1
|
Kana S, Basu D, Kar R, Nachiappa Ganesh R, Dubashi B, Kt H. Morphology, Morphometry, and Immunohistochemical Profile of Megakaryocytes and Bone Marrow Microenvironment in Disease Progression and Therapy Resistance in Chronic Myeloid Leukemia. Cureus 2024; 16:e67772. [PMID: 39328663 PMCID: PMC11424236 DOI: 10.7759/cureus.67772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/25/2024] [Indexed: 09/28/2024] Open
Abstract
Background Tyrosine kinase inhibitors have revolutionized the treatment of chronic myeloid leukemia (CML) since the beginning of the century. However, resistance to therapy and the progression of disease tend to occur in certain patients. The bone marrow microenvironment may play a role in the disease outcome. Megakaryocytes have multiple roles in the regulation and maintenance of the hematopoietic stem cell microenvironment. In the current study, we evaluated the association of megakaryocyte morphology, morphometry, and microenvironment with disease progression and therapy resistance in CML. Methodology Megakaryocyte morphology and morphometry were analyzed and compared between the different phases (chronic and advanced) at diagnosis in 150 cases of BCR-ABL-positive CML. All CML-CP patients (n = 119) were followed up on tyrosine kinase inhibitor therapy for a minimum of 15 months and classified based on their treatment outcome as a response, resistance to therapy, or progression of disease based on standard criteria. Immunohistochemistry on a bone marrow trephine biopsy was done for vascular endothelial growth factor (VEGF), FOXP3, CD150, CD48, CD44, osteopontin, CXCL12, N-cadherin, PDL-1, and IL-7, and their expression on megakaryocytes and their association with treatment outcome was evaluated. Results The morphology and morphometry of megakaryocytes showed a heterogeneous population in CML. Morphology and morphometric parameters, when compared between the chronic and advanced phases of disease at diagnosis, did not show any statistical difference. Megakaryocytes were variably positive for VEGF, FOXP3, CD150, CD48, osteopontin, N-cadherin, CXCL12, CD44, PDL-1, and IL-7. However, only CD44-positive megakaryocytes were statistically associated with the treatment outcome. The patients with a higher expression of CD44 megakaryocytes progressed to the advanced phase of the disease during therapy compared to those who responded. Conclusion Megakaryocyte morphology and morphometry were heterogeneous in CML; however, they did not show any significant difference with either the phase of the disease or with treatment outcomes. Among the various immunohistochemical markers of the microenvironment, only CD44-positivity on megakaryocytes was associated with poor treatment outcomes.
Collapse
Affiliation(s)
- Sreerag Kana
- Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Debdatta Basu
- Pathology, Mahatma Gandhi Medical College and Research Institute, Puducherry, IND
| | - Rakhee Kar
- Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Rajesh Nachiappa Ganesh
- Pathology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Biswajit Dubashi
- Medical Oncology, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| | - Harichandrakumar Kt
- Biostatistics, Jawaharlal Institute of Postgraduate Medical Education and Research, Puducherry, IND
| |
Collapse
|
2
|
Farkas DL. Biomedical Applications of Translational Optical Imaging: From Molecules to Humans. Molecules 2021; 26:molecules26216651. [PMID: 34771060 PMCID: PMC8587670 DOI: 10.3390/molecules26216651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 11/16/2022] Open
Abstract
Light is a powerful investigational tool in biomedicine, at all levels of structural organization. Its multitude of features (intensity, wavelength, polarization, interference, coherence, timing, non-linear absorption, and even interactions with itself) able to create contrast, and thus images that detail the makeup and functioning of the living state can and should be combined for maximum effect, especially if one seeks simultaneously high spatiotemporal resolution and discrimination ability within a living organism. The resulting high relevance should be directed towards a better understanding, detection of abnormalities, and ultimately cogent, precise, and effective intervention. The new optical methods and their combinations needed to address modern surgery in the operating room of the future, and major diseases such as cancer and neurodegeneration are reviewed here, with emphasis on our own work and highlighting selected applications focusing on quantitation, early detection, treatment assessment, and clinical relevance, and more generally matching the quality of the optical detection approach to the complexity of the disease. This should provide guidance for future advanced theranostics, emphasizing a tighter coupling-spatially and temporally-between detection, diagnosis, and treatment, in the hope that technologic sophistication such as that of a Mars rover can be translationally deployed in the clinic, for saving and improving lives.
Collapse
Affiliation(s)
- Daniel L. Farkas
- PhotoNanoscopy and Acceleritas Corporations, 13412 Ventura Boulevard, Sherman Oaks, CA 91423, USA; ; Tel.: +1-310-600-7102
- Clinical Photonics Corporation, 8591 Skyline Drive, Los Angeles, CA 90046, USA
- Department of Biomedical Engineering, University of Southern California, Los Angeles, CA 90089, USA
| |
Collapse
|
3
|
Bonaud A, Lemos JP, Espéli M, Balabanian K. Hematopoietic Multipotent Progenitors and Plasma Cells: Neighbors or Roommates in the Mouse Bone Marrow Ecosystem? Front Immunol 2021; 12:658535. [PMID: 33936091 PMCID: PMC8083056 DOI: 10.3389/fimmu.2021.658535] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022] Open
Abstract
The bone marrow is a complex ecosystem in which hematopoietic and non-hematopoietic cells reside. In this review, we discuss the bone marrow niches in mice that facilitate the survival, maintenance, and differentiation of cells of hematopoietic origin based on the recent literature. Our review places a special focus on the hematopoietic multipotent progenitors and on plasma cells, corresponding to the last stage of the B-cell lineage, that play a key role in the humoral memory response. We highlight the similarities between the microenvironments necessary for the establishment and the maintenance of these two immune cell subsets, and how the chemokine CXCL12/CXCR4 signaling axis contributes to these processes. Finally, we bring elements to address the following question: are multipotent progenitors and plasma cells neighbors or roommates within the bone marrow?
Collapse
Affiliation(s)
- Amélie Bonaud
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Julia P Lemos
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Marion Espéli
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Karl Balabanian
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
4
|
Multi-Modal Multi-Spectral Intravital Microscopic Imaging of Signaling Dynamics in Real-Time during Tumor-ImmuneInteractions. Cells 2021; 10:cells10030499. [PMID: 33652682 PMCID: PMC7996937 DOI: 10.3390/cells10030499] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 02/11/2021] [Accepted: 02/22/2021] [Indexed: 11/21/2022] Open
Abstract
Intravital microscopic imaging (IVM) allows for the study of interactions between immune cells and tumor cells in a dynamic, physiologically relevant system in vivo. Current IVM strategies primarily use fluorescence imaging; however, with the advances in bioluminescence imaging and the development of new bioluminescent reporters with expanded emission spectra, the applications for bioluminescence are extending to single cell imaging. Herein, we describe a molecular imaging window chamber platform that uniquely combines both bioluminescent and fluorescent genetically encoded reporters, as well as exogenous reporters, providing a powerful multi-plex strategy to study molecular and cellular processes in real-time in intact living systems at single cell resolution all in one system. We demonstrate that our molecular imaging window chamber platform is capable of imaging signaling dynamics in real-time at cellular resolution during tumor progression. Importantly, we expand the utility of IVM by modifying an off-the-shelf commercial system with the addition of bioluminescence imaging achieved by the addition of a CCD camera and demonstrate high quality imaging within the reaches of any biology laboratory.
Collapse
|
5
|
Zanetti C, Krause DS. "Caught in the net": the extracellular matrix of the bone marrow in normal hematopoiesis and leukemia. Exp Hematol 2020; 89:13-25. [PMID: 32755619 DOI: 10.1016/j.exphem.2020.07.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 07/28/2020] [Accepted: 07/30/2020] [Indexed: 12/14/2022]
Abstract
The influence of the bone marrow microenvironment on normal hematopoiesis, but also leukemia, has largely been accepted. However, the focus has been predominantly on the role of various cell types or cytokines maintaining hematopoietic stem cells or protecting leukemia stem cells from different therapies. A frequently overlooked component of the bone marrow microenvironment is the extracellular matrix, which not only provides a mechanical scaffold, but also serves as a source of growth factors. We discuss here how extracellular matrix proteins directly or indirectly modulate hematopoietic stem cell physiology and influence leukemia progression. It is hoped that existing and future studies on this topic may propel forward the possibility of augmenting normal hematopoiesis and improving therapies for leukemia, for instance, by targeting of the extracellular matrix in the bone marrow.
Collapse
Affiliation(s)
- Costanza Zanetti
- Georg-Speyer-Haus, Institute for Tumor Biology and Experimental Therapy, Frankfurt am Main, Germany
| | - Daniela S Krause
- German Cancer Research Center (DKFZ), Heidelberg, Germany; German Cancer Consortium (DKTK), Germany; Frankfurt Cancer Institute, Frankfurt, Germany; Faculty of Medicine, Johann Wolfgang Goethe University, Frankfurt, Germany.
| |
Collapse
|
6
|
Ly CH, Lynch GS, Ryall JG. A Metabolic Roadmap for Somatic Stem Cell Fate. Cell Metab 2020; 31:1052-1067. [PMID: 32433923 DOI: 10.1016/j.cmet.2020.04.022] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 02/13/2020] [Accepted: 04/29/2020] [Indexed: 01/14/2023]
Abstract
While metabolism was initially thought to play a passive role in cell biology by generating ATP to meet bioenergetic demands, recent studies have identified critical roles for metabolism in the generation of new biomass and provision of obligate substrates for the epigenetic modification of histones and DNA. This review details how metabolites generated through glycolysis and the tricarboxylic acid cycle are utilized by somatic stem cells to support cell proliferation and lineage commitment. Importantly, we also discuss the evolving hypothesis that histones can act as an energy reservoir during times of energy stress. Finally, we discuss how cells integrate both extrinsic metabolic cues and intrinsic metabolic machinery to regulate cell fate.
Collapse
Affiliation(s)
- C Hai Ly
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Gordon S Lynch
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - James G Ryall
- Centre for Muscle Research, Department of Physiology, The University of Melbourne, Melbourne, VIC 3010, Australia.
| |
Collapse
|
7
|
Stefanowski J, Fiedler AF, Köhler M, Günther R, Liublin W, Tschaikner M, Rauch A, Reismann D, Matthys R, Nützi R, Bixel MG, Adams RH, Niesner RA, Duda GN, Hauser AE. Limbostomy: Longitudinal Intravital Microendoscopy in Murine Osteotomies. Cytometry A 2020; 97:483-495. [PMID: 32196971 DOI: 10.1002/cyto.a.23997] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/20/2020] [Accepted: 02/24/2020] [Indexed: 12/28/2022]
Abstract
Bone healing involves the interplay of immune cells, mesenchymal cells, and vasculature over the time course of regeneration. Approaches to quantify the spatiotemporal aspects of bone healing at cellular resolution during long bone healing do not yet exist. Here, a novel technique termed Limbostomy is presented, which combines intravital microendoscopy with an osteotomy. This design allows a modular combination of an internal fixator plate with a gradient refractive index (GRIN) lens at various depths in the bone marrow and can be combined with a surgical osteotomy procedure. The field of view (FOV) covers a significant area of the fracture gap and allows monitoring cellular processes in vivo. The GRIN lens causes intrinsic optical aberrations which have to be corrected. The optical system was characterized and a postprocessing algorithm was developed. It corrects for wave front aberration-induced image plane deformation and for background and noise signals, enabling us to observe subcellular processes. Exemplarily, we quantitatively and qualitatively analyze angiogenesis in bone regeneration. We make use of a transgenic reporter mouse strain with nucleargreen fluorescent protein and membrane-bound tdTomato under the Cadherin-5 promoter. We observe two phases of vascularization. First, rapid vessel sprouting pervades the FOV within 3-4 days after osteotomy. Second, the vessel network continues to be dynamically remodeled until the end of our observation time, 14 days after surgery. Limbostomy opens a unique set of opportunities and allows further insight on spatiotemporal aspects of bone marrow biology, for example, hematopoiesis, analysis of cellular niches, immunological memory, and vascularization in the bone marrow during health and disease. © 2020 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Jonathan Stefanowski
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Alexander F Fiedler
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Freie Universitat Berlin, Veterinary Medicine, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Markus Köhler
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Freie Universitat Berlin, Veterinary Medicine, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Robert Günther
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Wjatscheslaw Liublin
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Martin Tschaikner
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Ariana Rauch
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - David Reismann
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | | | | | | | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Raluca A Niesner
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Freie Universitat Berlin, Veterinary Medicine, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Georg N Duda
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Julius Wolff Institute, Berlin, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| |
Collapse
|
8
|
Live-animal imaging of native haematopoietic stem and progenitor cells. Nature 2020; 578:278-283. [PMID: 32025033 PMCID: PMC7021587 DOI: 10.1038/s41586-020-1971-z] [Citation(s) in RCA: 148] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/06/2019] [Indexed: 12/26/2022]
Abstract
The biology of hematopoietic stem cells (HSCs) has predominantly been studied under transplantation conditions1,2. Particularly challenging has been the study of dynamic HSC behaviors given that live animal HSC visualization in the native niche still represents an elusive goal in the field. Here, we describe a dual genetic strategy in mice that restricts reporter labeling to a subset of the most quiescent long-term HSCs (LT-HSCs) and that is compatible with current intravital imaging approaches in the calvarial bone marrow (BM)3–5. We find that this subset of LT-HSCs resides in close proximity to both sinusoidal blood vessels and the endosteal surface. In contrast, multipotent progenitor cells (MPPs) display a broader distance distribution from the endosteum and are more likely to be associated with transition zone vessels. LT-HSCs are not found in BM niches with the deepest hypoxia and instead are found in similar hypoxic environments as MPPs. In vivo time-lapse imaging reveals that LT-HSCs display limited motility at steady-state. Following activation, LT-HSCs display heterogenous responses, with some cells becoming highly motile and a fraction of HSCs expanding clonally within spatially restricted domains. These domains have defined characteristics, as HSC expansion is found almost exclusively in a subset of BM cavities exhibiting bone-remodeling activities. In contrast, cavities with low bone-resorbing activities do not harbor expanding HSCs. These findings point to a new degree of heterogeneity within the BM microenvironment, imposed by the stages of bone turnover. Overall, our approach enables direct visualization of HSC behaviors and dissection of heterogeneity in HSC niches.
Collapse
|
9
|
Wang F, Wei D, Suo Y, Zhu X, Yuan Y, Gao W, Jiang H, Wei X, Chen T. In vivo flow cytometry combined with intravital microscopy to monitor kinetics of transplanted bone marrow mononuclear cells in peripheral blood and bone marrow. Mol Biol Rep 2019; 47:1-10. [DOI: 10.1007/s11033-019-04608-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/16/2019] [Indexed: 12/26/2022]
|
10
|
Kim J, Bixel MG. Intravital Multiphoton Imaging of the Bone and Bone Marrow Environment. Cytometry A 2019; 97:496-503. [DOI: 10.1002/cyto.a.23937] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Affiliation(s)
- JungMo Kim
- Department of Tissue MorphogenesisMax Planck Institute for Molecular Biomedicine D‐48149 Münster Germany
| | - Maria Gabriele Bixel
- Department of Tissue MorphogenesisMax Planck Institute for Molecular Biomedicine D‐48149 Münster Germany
| |
Collapse
|
11
|
De Niz M, Meehan GR, Tavares J. Intravital microscopy: Imaging host-parasite interactions in lymphoid organs. Cell Microbiol 2019; 21:e13117. [PMID: 31512335 DOI: 10.1111/cmi.13117] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/25/2019] [Accepted: 09/01/2019] [Indexed: 12/11/2022]
Abstract
Intravital microscopy allows imaging of biological phenomena within living animals, including host-parasite interactions. This has advanced our understanding of both, the function of lymphoid organs during parasitic infections, and the effect of parasites on such organs to allow their survival. In parasitic research, recent developments in this technique have been crucial for the direct study of host-parasite interactions within organs at depths, speeds and resolution previously difficult to achieve. Lymphoid organs have gained more attention as we start to understand their function during parasitic infections and the effect of parasites on them. In this review, we summarise technical and biological findings achieved by intravital microscopy with respect to the interaction of various parasites with host lymphoid organs, namely the bone marrow, thymus, lymph nodes, spleen and the mucosa-associated lymphoid tissue, and present a view into possible future applications.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, Heussler Lab, University of Bern, Bern, Switzerland
| | - Gavin R Meehan
- Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Joana Tavares
- i3S-Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.,IBMC-Instituto de Biologia Molecular e Celular, University of Porto, Porto, Portugal
| |
Collapse
|
12
|
Shiflett LA, Tiede-Lewis LM, Xie Y, Lu Y, Ray EC, Dallas SL. Collagen Dynamics During the Process of Osteocyte Embedding and Mineralization. Front Cell Dev Biol 2019; 7:178. [PMID: 31620436 PMCID: PMC6759523 DOI: 10.3389/fcell.2019.00178] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 08/15/2019] [Indexed: 12/20/2022] Open
Affiliation(s)
- Lora A. Shiflett
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, United States
| | - LeAnn M. Tiede-Lewis
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Yixia Xie
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Yongbo Lu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, United States
| | - Eleanor C. Ray
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, United States
| | - Sarah L. Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, United States
- *Correspondence: Sarah L. Dallas,
| |
Collapse
|
13
|
Gomariz A, Isringhausen S, Helbling PM, Nombela-Arrieta C. Imaging and spatial analysis of hematopoietic stem cell niches. Ann N Y Acad Sci 2019; 1466:5-16. [PMID: 31368140 DOI: 10.1111/nyas.14184] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Revised: 05/30/2019] [Accepted: 06/06/2019] [Indexed: 01/21/2023]
Abstract
Hematopoietic stem cells (HSCs) have been long proposed to reside in defined anatomical locations within bone marrow (BM) tissues in direct contact or close proximity to nurturing cell types. Imaging techniques that allow the simultaneous mapping of HSCs and interacting cell types have been central to the discovery of basic principles of these so-called HSC niches. Despite major progress in the field, a quantitative and comprehensive model of the cellular and molecular components that define these specialized microenvironments is lacking to date, and uncertainties remain on the preferential localization of HSCs in the context of complex BM tissue landscapes. Recent technological breakthroughs currently allow for the quantitative spatial analysis of BM cellular components with extraordinary precision. Here, we critically discuss essential technical aspects related to imaging approaches, image processing tools, and spatial statistics, which constitute the three basic elements of rigorous quantitative spatial analyses of HSC niches in the BM microenvironment.
Collapse
Affiliation(s)
- Alvaro Gomariz
- Department of Medical Oncology and Hematology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Stephan Isringhausen
- Department of Medical Oncology and Hematology, University Hospital, University of Zurich, Zurich, Switzerland
| | - Patrick M Helbling
- Department of Medical Oncology and Hematology, University Hospital, University of Zurich, Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital, University of Zurich, Zurich, Switzerland
| |
Collapse
|
14
|
Abstract
Over the past two decades there have been unprecedented advances in the capabilities for live cell imaging using light and confocal microscopy. Together with the discovery of green fluorescent protein and its derivatives and the development of a vast array of fluorescent imaging probes and conjugates, it is now possible to image virtually any intracellular or extracellular protein or structure. Traditional static imaging of fixed bone cells and tissues takes a snapshot view of events at a specific time point, but can often miss the dynamic aspects of the events being investigated. This chapter provides an overview of the application of live cell imaging approaches for the study of bone cells and bone organ cultures. Rather than emphasizing technical aspects of the imaging equipment, which may vary in different laboratories, we focus on what we consider to be the important principles that are of most practical use for an investigator setting up these techniques in their own laboratory. We also provide detailed protocols that our laboratory has used for live imaging of bone cell and organ cultures.
Collapse
Affiliation(s)
- Sarah L Dallas
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, Kansas City, MO, USA.
| | - Patricia A Veno
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, Kansas City, MO, USA
| | - LeAnn M Tiede-Lewis
- Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri, Kansas City, Kansas City, MO, USA
| |
Collapse
|
15
|
Metabolic Regulations in Hematopoietic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1143:59-74. [PMID: 31338815 DOI: 10.1007/978-981-13-7342-8_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
One of the bottlenecks of the treatments for malignant hematopoietic disorders is the unavailability of sufficient amount of hematopoietic stem cells (HSCs). HSCs are considered to be originated from the aorta-gonad-mesonephros and gradually migrates into fetal liver and resides in a unique microenvironment/niche of bone marrow. Although many intrinsic and extrinsic factors (niche components) are reported to be involved in the origination, maturation, migration, and localization of HSCs at different developmental stages, the detailed molecular mechanisms still remain largely unknown. Previous studies have shown that intrinsic metabolic networks may be critical for the cell fate determinations of HSCs. For example, HSCs mainly utilize glycolysis as the main energy sources; oxidative phosphorylation is required for the homeostasis of HSCs; lipid or amino acid metabolisms may also sustain HSC stemness. Mechanistically, lots of regulatory pathways, such as MEIS1/HIF1A and PI3K/AKT/mTOR signaling, are found to fine-tune the different nutrient metabolisms and cell fate commitments of HSCs. However, more efforts are required for the optimization and establishment of precise metabolic techniques specific for the HSCs with relatively rare cell frequency and understanding of the basic metabolic properties and their underlying regulatory mechanisms of different nutrients (such as glucose) during the different developmental stages of HSCs.
Collapse
|
16
|
Behrmann L, Wellbrock J, Fiedler W. Acute Myeloid Leukemia and the Bone Marrow Niche-Take a Closer Look. Front Oncol 2018; 8:444. [PMID: 30370251 PMCID: PMC6195156 DOI: 10.3389/fonc.2018.00444] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/24/2018] [Indexed: 12/20/2022] Open
Abstract
The bone marrow is the home of hematopoiesis and is therefore a hotspot for the development of hematopoietic diseases. Complex interactions between the bone marrow microenvironment and hematopoietic stem cells must find a balance between proliferation, differentiation and homeostasis of the stem cell compartment. Changes in this tightly regulated network can provoke malignant transformation, leading to hematopoietic diseases. Here we focus on acute myeloid leukemia (AML), since this is the most frequent acute leukemia in adulthood with very poor overall survival rates and where relapse after chemotherapy continues to be a major challenge, driving demand for new therapeutic strategies. Current research is focusing on the identification of specific interactions between leukemic blasts and their niche components, which may be exploited as novel treatment targets along with induction chemotherapy. Significant progress has been gained over the last few years in the field of high-resolution imaging. Confocal ex vivo and intravital microscopy have revealed a detailed map of bone marrow structures and components; as well as identifying numerous alterations in the stem cell niche that correspond to disease progression. However, the underlying mechanisms are still not completely understood and due to the complexity, their elucidation remains a challenging. This review discusses the constitution of the AML niche in the bone marrow, the improvement in visualization of the complex three-dimensional niche structures and points out new therapeutic strategies to increase the overall survival of AML patients.
Collapse
Affiliation(s)
- Lena Behrmann
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Jasmin Wellbrock
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section Pneumology, Hubertus Wald University Cancer Center, University Medical Center Hamburg Eppendorf, Hamburg, Germany
| |
Collapse
|
17
|
Holzwarth K, Köhler R, Philipsen L, Tokoyoda K, Ladyhina V, Wählby C, Niesner RA, Hauser AE. Multiplexed fluorescence microscopy reveals heterogeneity among stromal cells in mouse bone marrow sections. Cytometry A 2018; 93:876-888. [DOI: 10.1002/cyto.a.23526] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Revised: 05/22/2018] [Accepted: 06/20/2018] [Indexed: 01/07/2023]
Affiliation(s)
| | - Ralf Köhler
- Deutsches Rheumaforschungszentrum, a Leibniz Institute; Berlin Germany
| | - Lars Philipsen
- Institute of Molecular and Clinical Immunology, Medical Faculty; Otto von Guericke University; Magdeburg Germany
| | - Koji Tokoyoda
- Deutsches Rheumaforschungszentrum, a Leibniz Institute; Berlin Germany
| | - Valeriia Ladyhina
- Centre for Image Analysis, Department of Information Technology, Division of Visual Information and Interaction; Uppsala University; Uppsala Sweden
| | - Carolina Wählby
- Centre for Image Analysis, Department of Information Technology, Division of Visual Information and Interaction; Uppsala University; Uppsala Sweden
| | - Raluca A. Niesner
- Deutsches Rheumaforschungszentrum, a Leibniz Institute; Berlin Germany
| | - Anja E. Hauser
- Immune Dynamics; Charité - Universitätsmedizin; Berlin Germany
- Deutsches Rheumaforschungszentrum, a Leibniz Institute; Berlin Germany
| |
Collapse
|
18
|
Systems for localized release to mimic paracrine cell communication in vitro. J Control Release 2018; 278:24-36. [PMID: 29601931 DOI: 10.1016/j.jconrel.2018.03.028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/24/2018] [Accepted: 03/26/2018] [Indexed: 12/27/2022]
Abstract
Paracrine cell communication plays a pivotal role for signal exchange between proximal cells in vivo. However, this localized, gradient type release of mediators at very low concentrations (pg/ml), relevant during physiological and pathological processes, is rarely reflected within in vitro approaches. This review gives an overview on state-of-the-art approaches, which transfer the paracrine cell-to-cell communication into in vitro cell culture model setups. The traditional methods like trans-well assays and more advanced microfluidic approaches are included. The review focusses on systems for localized release, mostly based on microparticles, which tightly mimic the paracrine interaction between single cells in 3D microenvironments. Approaches based on single microparticles, with the main focus on affinity-controlled storage and release of cytokines, are reviewed and their importance for understanding paracrine communication is highlighted. Various methods to study the cytokine release and their advantages and disadvantages are discussed. Basic principles of the release characteristics, like diffusion mechanisms, are quantitatively described, including the formation of resulting gradients around the local sources. In vitro cell experiments using such localized microparticle release systems in approaches to increase understanding of stem cell behavior within their niches and regulation of wound healing are highlighted as examples of successful localized release systems for mimicking paracrine cell communication.
Collapse
|
19
|
Bixel MG, Kusumbe AP, Ramasamy SK, Sivaraj KK, Butz S, Vestweber D, Adams RH. Flow Dynamics and HSPC Homing in Bone Marrow Microvessels. Cell Rep 2017; 18:1804-1816. [PMID: 28199850 PMCID: PMC5318670 DOI: 10.1016/j.celrep.2017.01.042] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/28/2016] [Accepted: 01/18/2017] [Indexed: 12/17/2022] Open
Abstract
Measurements of flow velocities at the level of individual arterial vessels and sinusoidal capillaries are crucial for understanding the dynamics of hematopoietic stem and progenitor cell homing in the bone marrow vasculature. We have developed two complementary intravital two-photon imaging approaches to determine blood flow dynamics and velocities in multiple vessel segments by capturing the motion of red blood cells. High-resolution spatiotemporal measurements through a cranial window to determine short-time dynamics of flowing blood cells and repetitive centerline scans were used to obtain a detailed flow-profile map with hemodynamic parameters. In addition, we observed the homing of individual hematopoietic stem and progenitor cells and obtained detailed information on their homing behavior. With our imaging setup, we determined flow patterns at cellular resolution, blood flow velocities and wall shear stress in small arterial vessels and highly branched sinusoidal capillaries, and the cellular dynamics of hematopoietic stem and progenitor cell homing.
Collapse
Affiliation(s)
- M Gabriele Bixel
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; Faculty of Medicine, University of Münster, 48149 Münster, Germany.
| | - Anjali P Kusumbe
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Saravana K Ramasamy
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Kishor K Sivaraj
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Stefan Butz
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; Faculty of Medicine, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
20
|
Intravital longitudinal wide-area imaging of dynamic bone marrow engraftment and multilineage differentiation through nuclear-cytoplasmic labeling. PLoS One 2017; 12:e0187660. [PMID: 29099870 PMCID: PMC5669471 DOI: 10.1371/journal.pone.0187660] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 10/24/2017] [Indexed: 12/14/2022] Open
Abstract
Bone marrow is a vital tissue that produces the majority of erythrocytes, thrombocytes, and immune cells. Bone marrow transplantation (BMT) has been widely performed in patients with blood disorders and cancers. However, the cellular-level behaviors of the transplanted bone marrow cells over wide-areas of the host bone marrow after the BMT are not fully understood yet. In this work, we performed a longitudinal wide-area cellular-level observation of the calvarial bone marrow after the BMT in vivo. Using a H2B-GFP/β-actin-DsRed double-transgenic mouse model as a donor, a subcellular-level nuclear-cytoplasmic visualization of the transplanted bone marrow cells was achieved, which enabled a direct in vivo dynamic monitoring of the distribution and proliferation of the transplanted bone marrow cells. The same spots in the wide-area of the calvarial bone marrow were repeatedly identified using fluorescently labeled vasculature as a distinct landmark. It revealed various dynamic cellular-level behaviors of the transplanted BM cells in early stage such as cluster formation, migration, and active proliferation in vivo.
Collapse
|
21
|
Tamrin SH, Majedi FS, Tondar M, Sanati-Nezhad A, Hasani-Sadrabadi MM. Electromagnetic Fields and Stem Cell Fate: When Physics Meets Biology. Rev Physiol Biochem Pharmacol 2017; 171:63-97. [PMID: 27515674 DOI: 10.1007/112_2016_4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Controlling stem cell (SC) fate is an extremely important topic in the realm of SC research. A variety of different external cues mainly mechanical, chemical, or electrical stimulations individually or in combination have been incorporated to control SC fate. Here, we will deconstruct the probable relationship between the functioning of electromagnetic (EMF) and SC fate of a variety of different SCs. The electromagnetic (EM) nature of the cells is discussed with the emphasis on the effects of EMF on the determinant factors that directly and/or indirectly influence cell fate. Based on the EM effects on a variety of cellular processes, it is believed that EMFs can be engineered to provide a controlled signal with the highest impact on the SC fate decision. Considering the novelty and broad applications of applying EMFs to change SC fate, it is necessary to shed light on many unclear mechanisms underlying this phenomenon.
Collapse
Affiliation(s)
- Sara Hassanpour Tamrin
- Center of Excellence in Biomaterials, Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran
| | | | - Mahdi Tondar
- Department of Biochemistry and Molecular & Cellular Biology, School of Medicine, Georgetown University, Washington, DC, USA
| | - Amir Sanati-Nezhad
- BioMEMS and BioInspired Microfluidic Laboratory, Department of Mechanical and Manufacturing Engineering, Center for Bioengineering Research and Education, University of Calgary, Calgary, AB, Canada, T2N1N4.
| | - Mohammad Mahdi Hasani-Sadrabadi
- Department of Chemistry & Biochemistry, and California NanoSystems Institute, University of California at Los Angeles, Los Angeles, CA, 90095, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience and G.W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
22
|
In vivo longitudinal visualization of bone marrow engraftment process in mouse calvaria using two-photon microscopy. Sci Rep 2017; 7:44097. [PMID: 28276477 PMCID: PMC5343427 DOI: 10.1038/srep44097] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Accepted: 01/31/2017] [Indexed: 12/13/2022] Open
Abstract
Intravital microscopy of mouse calvarial bone marrow (BM) is a powerful method for studying hematopoietic stem cells (HSCs) and the BM microenvironment at the cellular level. However, the current method used to access the mouse calvaria allows for only a few imaging times in the same mouse because of scar formation and inflammation induced by multiple surgeries. Longitudinal imaging of the BM may help better understand its microenvironment. In this study, a mouse calvarial window model was developed for longitudinal imaging that involves attaching a cover glass window onto the mouse calvaria and sealing the surrounding exposed area with cyanoacrylate glue and dental cement. The model was used for the longitudinal two-photon microscopy (TPM) imaging of the BM engraftment process. The same BM cavity sites were imaged multiple times over 4 weeks after BM transplantation (BMT). Temporal changes in the BM microenvironment, such as the reconstitution of transplanted BM cells and the recovery of vasculature, were observed and analysed qualitatively and quantitatively. Longitudinal intravital microscopy using the mouse calvarial window model was successfully demonstrated and may be useful for further BM studies.
Collapse
|
23
|
Nombela-Arrieta C, Isringhausen S. The Role of the Bone Marrow Stromal Compartment in the Hematopoietic Response to Microbial Infections. Front Immunol 2017; 7:689. [PMID: 28163704 PMCID: PMC5247475 DOI: 10.3389/fimmu.2016.00689] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/23/2016] [Indexed: 12/18/2022] Open
Abstract
Continuous production of blood cells unfolds within a complex three-dimensional tissue scaffold established by highly organized stromal cell networks of mesenchymal, neural, and vascular origin inside bone marrow (BM) cavities. Collectively, stromal cells have been shown to serve two principal roles; first as primary participants of bone remodeling and metabolism and second as master regulators of different stages of blood cell development and production. Indeed, ample evidence demonstrates that stromal cells can sense and integrate systemic signals to shape hematopoietic responses and that these regulatory mechanisms are subverted in multiple pathologic conditions. Microbial infections are stressors that elicit potent inflammatory reactions and induce substantial alterations of hematopoietic output. Whether the cellular components of the BM stromal microenvironment are targeted by infections and participate in infection-induced hematopoiesis has not been investigated in sufficient detail to date. In this manuscript, we provide a succinct updated overview of the different cell populations that are currently known to form BM stroma. We discuss experimental evidence demonstrating that different stromal components are actively damaged or functionally altered by pathogens and/or ensuing inflammatory signals and review how these effects are known to contribute to the hematologic manifestations observed during infections.
Collapse
|
24
|
Extended time-lapse in vivo imaging of tibia bone marrow to visualize dynamic hematopoietic stem cell engraftment. Leukemia 2016; 31:1582-1592. [PMID: 27890929 PMCID: PMC5498248 DOI: 10.1038/leu.2016.354] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/03/2016] [Accepted: 11/07/2016] [Indexed: 12/12/2022]
Abstract
Homing, engraftment and proliferation of hematopoietic stem/progenitor cell (HSC/HPCs) are crucial steps required for success of a bone marrow transplant. Observation of these critical events is limited by the opaque nature of bone. Here we demonstrate how individual HSCs engraft in long bones by thinning one side of the tibia for direct and unbiased observation. Intravital imaging enabled detailed visualization of single Sca-1+, c-Kit+, Lineage− (SKL) cell migration to bone marrow niches and subsequent proliferation to reconstitute hematopoiesis. This longitudinal study allowed direct observation of dynamic HSC/HPC activities during engraftment in full color for up to six days in live recipients. Individual SKL cells, but not mature or committed progenitor cells, preferentially homed to a limited number of niches near highly vascularized endosteal regions, and clonally expanded. Engraftment of SKL cells in P-selectin and osteopontin knockout mice showed abnormal homing and expansion of SKL cells. CD150+, CD48− SKL populations initially engrafted in the central marrow region, utilizing only a subset of niches occupied by the parent SKL cells. Our study demonstrates that time-lapse imaging of tibia can be a valuable tool to understand the dynamic characteristics of functional HSC and niche components in various mouse models.
Collapse
|
25
|
Digesting the role of bone marrow macrophages on hematopoiesis. Immunobiology 2016; 222:814-822. [PMID: 27890297 DOI: 10.1016/j.imbio.2016.11.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 10/27/2016] [Accepted: 11/12/2016] [Indexed: 01/07/2023]
Abstract
Tissue resident macrophages are found in various tissues like Langerhans cells in the skin or alveolar macrophages in the lung, and their main function is to regulate organ homeostasis. They have also been observed in the bone marrow and these cells in particular have been gaining importance in recent years as they are key players in hematopoiesis. However, as the characterization and classification of these putatively different bone marrow resident macrophages is far from established there is a need to generate an overview of tissue resident macrophages of the bone marrow. Here, we will review the current knowledge of bone marrow resident macrophages both in mouse and human. We will discuss the state of the art on the origin of bone marrow macrophages, specialized microenvironments where they reside and their unique characteristics. We will emphasize the two best studied examples of macrophage homeostatic function in the bone marrow, specifically within erythroblastic islands and the hematopoietic stem cell niche. Although increasing evidence shows that bone marrow resident macrophages are indispensable for hematopoietic stem cell function and bone marrow erythroid output, the field of bone marrow macrophages is in its infancy. This field is in dire need for a unified nomenclature to support functional experiments, model systems, and the identification of niches.
Collapse
|
26
|
Kong L, Tang J, Cui M. Multicolor multiphoton in vivo imaging flow cytometry. OPTICS EXPRESS 2016; 24:6126-35. [PMID: 27136806 PMCID: PMC5025233 DOI: 10.1364/oe.24.006126] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 03/06/2016] [Accepted: 03/07/2016] [Indexed: 05/20/2023]
Abstract
In vivo flow cytometry provides a non-invasive way of probing the biology of circulating cells during disease progression and studying cellular response to therapy. However, current methods provide little morphological information which potentially could be new biological marker for early disease diagnosis, and fail to reveal intercellular interactions. Here we report a multi-color, multiphoton in vivo imaging flow cytometry, to image circulating cells within the vasculature of scattering tissues at high spatiotemporal resolution. We apply it in imaging of cellular dynamics in bone marrow through the intact mouse skull, in situ deformability cytometry, distinguishing cellular clusters, and simultaneously monitoring multiple types of trafficking cells based on their morphologies and fluorescence emission colors.
Collapse
Affiliation(s)
- Lingjie Kong
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907,
USA
| | - Jianyong Tang
- Laboratory of Systems Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892,
USA
| | - Meng Cui
- School of Electrical and Computer Engineering, Purdue University, West Lafayette, IN 47907,
USA
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907,
USA
- Integrated Imaging Cluster, Purdue University, West Lafayette, IN 47907,
USA
- Bindley Bioscience Center, Purdue University, West Lafayette, IN 47907,
USA
| |
Collapse
|
27
|
Peris P, Roforth MM, Nicks KM, Fraser D, Fujita K, Jilka RL, Khosla S, McGregor U. Ability of circulating human hematopoietic lineage negative cells to support hematopoiesis. J Cell Biochem 2016; 116:58-66. [PMID: 25145595 DOI: 10.1002/jcb.24942] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 08/15/2014] [Indexed: 12/24/2022]
Abstract
Hematopoietic stem cell (HSC) self-renewal is regulated by osteoblast and/or endothelial cells within the hematopoietic niche. However, the true identity of the supporting cells and the nature of the secreted factors remain uncertain. We developed a novel mouse model and analyzed whether circulating human peripheral hematopoietic lineage negative/AP+ (lin-/AP+) cells support hematopoiesis in vivo. Thus, immunocompromised (Rag) mice expressing thymidine kinase (Tk) under the control of the 3.6Col1α1 promoter (Tk-Rag) were treated with ganciclovir, resulting in osteoblast progenitor cell ablation and subsequent loss of hematopoiesis (evaluated by measuring mouse Ter119+ erythroid cells). Following hematopoietic cell depletion, human bone marrow-derived marrow stromal cells (MSCs) or lin-/AP+ cells were infused into Tk-Rag mice and compared with saline infusions. Ganciclovir significantly reduced (7.4-fold) Ter119+ cells in the bone marrow of Tk-Rag mice compared to saline injections. Infusion of either MSCs or lin-/AP+ cells into ganciclovir-treated mice resulted in a 3.3-fold and 2.7-fold increase (P < 0.01), respectively, in Ter119+ cells compared to mice receiving saline. Relative to lin-/AP- cells, lin-/AP+ cells expressed high levels of mesenchymal, endothelial, and hematopoiesis supporting genes. Thus, human peripheral blood lin-/AP+ cells represent a novel cell type capable of supporting hematopoiesis in a manner comparable to MSCs.
Collapse
Affiliation(s)
- Pilar Peris
- Endocrine Research Unit, College of Medicine, Mayo Clinic, Rochester, Minnesota; Rheumatology Department, Hospital Clinic, IDIBAPS, CIBERehd, Barcelona, Spain
| | | | | | | | | | | | | | | |
Collapse
|
28
|
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: 3.1] [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
|
29
|
Heideveld E, Masiello F, Marra M, Esteghamat F, Yağcı N, von Lindern M, Migliaccio ARF, van den Akker E. CD14+ cells from peripheral blood positively regulate hematopoietic stem and progenitor cell survival resulting in increased erythroid yield. Haematologica 2015; 100:1396-406. [PMID: 26294724 DOI: 10.3324/haematol.2015.125492] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 08/12/2015] [Indexed: 12/28/2022] Open
Abstract
Expansion of erythroblasts from human peripheral blood mononuclear cells is 4- to 15-fold more efficient than that of CD34(+) cells purified from peripheral blood mononuclear cells. In addition, purified CD34(+) and CD34(-) populations from blood do not reconstitute this erythroid yield, suggesting a role for feeder cells present in blood mononuclear cells that increase hematopoietic output. Immunodepleting peripheral blood mononuclear cells for CD14(+) cells reduced hematopoietic stem and progenitor cell expansion. Conversely, the yield was increased upon co-culture of CD34(+) cells with CD14(+) cells (full contact or transwell assays) or CD34(+) cells re-constituted in conditioned medium from CD14(+) cells. In particular, CD14(++)CD16(+) intermediate monocytes/macrophages enhanced erythroblast outgrowth from CD34(+) cells. No effect of CD14(+) cells on erythroblasts themselves was observed. However, 2 days of co-culturing CD34(+) and CD14(+) cells increased CD34(+) cell numbers and colony-forming units 5-fold. Proliferation assays suggested that CD14(+) cells sustain CD34(+) cell survival but not proliferation. These data identify previously unrecognized erythroid and non-erythroid CD34(-) and CD34(+) populations in blood that contribute to the erythroid yield. A flow cytometry panel containing CD34/CD36 can be used to follow specific stages during CD34(+) differentiation to erythroblasts. We have shown modulation of hematopoietic stem and progenitor cell survival by CD14(+) cells present in peripheral blood mononuclear cells which can also be found near specific hematopoietic niches in the bone marrow.
Collapse
Affiliation(s)
- Esther Heideveld
- Sanquin Research, Dept. of Hematopoiesis, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Francesca Masiello
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanita, Rome, Italy
| | - Manuela Marra
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanita, Rome, Italy
| | - Fatemehsadat Esteghamat
- Sanquin Research, Dept. of Hematopoiesis, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Nurcan Yağcı
- Sanquin Research, Dept. of Hematopoiesis, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Marieke von Lindern
- Sanquin Research, Dept. of Hematopoiesis, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Anna Rita F Migliaccio
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanita, Rome, Italy Division of Hematology and Medical Oncology, Mount Sinai School of Medicine and the Myeloproliferative Disorders Research Consortium, New York, NY, USA
| | - Emile van den Akker
- Sanquin Research, Dept. of Hematopoiesis, and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, The Netherlands
| |
Collapse
|
30
|
Lin HD, Fong CY, Biswas A, Choolani M, Bongso A. Human Wharton's jelly stem cells, its conditioned medium and cell-free lysate inhibit the growth of human lymphoma cells. Stem Cell Rev Rep 2015; 10:573-86. [PMID: 24789672 DOI: 10.1007/s12015-014-9514-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Several groups have reported that primitive mesenchymal stem cells from the gelatinous matrix of the Wharton's jelly of the human umbilical cord (hWJSCs) possess tumoricidal properties and inhibit the growth of solid tumours such as human mammary carcinoma, ovarian carcinoma and osteosarcoma. This unique characteristic led to the hypothesis that hWJSCs serve as a natural defence against migrating cancer cells from mother to fetus thus explaining why tumorigenesis in the fetus is rare. However, it is not known whether non-solid malignant hematopoietic cells are also inhibited by hWJSCs and what the exact tumoricidal mechanisms are. We therefore evaluated the influence of hWJSCs and its extracts on Burkitt's lymphoma cells. Cell proliferation (BrdU and Ki67+), viability (MTT) and cell death (Annexin V-Propidium iodide and live/dead) assays showed significant inhibition of lymphoma cell growth after 48 h exposure to hWJSCs or its extracts compared to controls. Increased cell death was observed at sub-G1 and S and decreased proliferation at G2/M phases of the mitotic cycle. Superoxide dismutase and hydrogen peroxide activity were significantly increased and glutathione peroxidase significantly decreased in treated lymphoma cells. Time lapse imaging and confocal z-stack images showed yellow fluorescent in situ hybridization (FISH) signals of lymphoma cell Y chromosomes within the cytoplasm of female red labelled hWJSCs. We hypothesize that the growth of lymphoma cells is inhibited by the molecules secreted by hWJSCs that use oxidative stress pathways to induce cell death followed by engulfment of the apoptotic remains of the lymphoma cells by the hWJSCs.
Collapse
Affiliation(s)
- Hao Daniel Lin
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, Kent Ridge, Singapore, Singapore, 119228
| | | | | | | | | |
Collapse
|
31
|
Turcotte R, Alt C, Mortensen LJ, Lin CP. Characterization of multiphoton microscopy in the bone marrow following intravital laser osteotomy. BIOMEDICAL OPTICS EXPRESS 2014; 5:3578-88. [PMID: 25360374 PMCID: PMC4206326 DOI: 10.1364/boe.5.003578] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 08/15/2014] [Accepted: 08/17/2014] [Indexed: 05/20/2023]
Abstract
The bone marrow is an important site where all blood cells are formed from hematopoietic stem cells and where hematologic malignancies such as leukemia emerge. It is also a frequent site for metastasis of solid tumors such as breast cancer and prostate cancer. Intravital microscopy is a powerful tool for studying the bone marrow with single cell and sub-cellular resolution. To improve optical access to this rich biological environment, plasma-mediated laser ablation with sub-microjoule femtosecond pulses was used to thin cortical bone. By locally removing a superficial layer of bone (local laser osteotomy), significant improvements in multiphoton imaging were observed in individual bone marrow compartments in vivo. This work demonstrates the utility of scanning laser ablation of hard tissue with sub-microjoule pulses as a preparatory step to imaging.
Collapse
Affiliation(s)
- Raphaël Turcotte
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, CPZN 8238, 185 Cambridge Street, Boston, MA 02114,
USA
- Department of Biomedical Engineering, Boston University, Boston, MA 02215,
USA
| | - Clemens Alt
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, CPZN 8238, 185 Cambridge Street, Boston, MA 02114,
USA
| | - Luke J. Mortensen
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, CPZN 8238, 185 Cambridge Street, Boston, MA 02114,
USA
| | - Charles P. Lin
- Advanced Microscopy Program, Center for Systems Biology and Wellman Center for Photomedicine, Massachusetts General Hospital, Harvard Medical School, CPZN 8238, 185 Cambridge Street, Boston, MA 02114,
USA
| |
Collapse
|
32
|
Glutathione S-transferase P influences redox and migration pathways in bone marrow. PLoS One 2014; 9:e107478. [PMID: 25216273 PMCID: PMC4162606 DOI: 10.1371/journal.pone.0107478] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2014] [Accepted: 08/11/2014] [Indexed: 01/22/2023] Open
Abstract
To interrogate why redox homeostasis and glutathione S-transferase P (GSTP) are important in regulating bone marrow cell proliferation and migration, we isolated crude bone marrow, lineage negative and bone marrow derived-dendritic cells (BMDDCs) from both wild type (WT) and knockout (Gstp1/p2(-/-)) mice. Comparison of the two strains showed distinct thiol expression patterns. WT had higher baseline and reactive oxygen species-induced levels of S-glutathionylated proteins, some of which (sarco-endoplasmic reticulum Ca2(+)-ATPase) regulate Ca(2+) fluxes and subsequently influence proliferation and migration. Redox status is also a crucial determinant in the regulation of the chemokine system. CXCL12 chemotactic response was stronger in WT cells, with commensurate alterations in plasma membrane polarization/permeability and intracellular calcium fluxes; activities of the downstream kinases, ERK and Akt were also higher in WT. In addition, expression levels of the chemokine receptor CXCR4 and its associated phosphatase, SHP-2, were higher in WT. Inhibition of CXCR4 or SHP2 decreased the extent of CXCL12-induced migration in WT BMDDCs. The differential surface densities of CXCR4, SHP-2 and inositol trisphosphate receptor in WT and Gstp1/p2(-/-) cells correlated with the differential CXCR4 functional activities, as measured by the extent of chemokine-induced directional migration and differences in intracellular signaling. These observed differences contribute to our understanding of how genetic ablation of GSTP causes different levels of myeloproliferation and migration [corrected]
Collapse
|
33
|
Abstract
SIGNIFICANCE The effect of redox signaling on hematopoietic stem cell (HSC) function is not clearly understood. RECENT ADVANCES A growing body of evidence suggests that adult HSCs reside in the hypoxic bone marrow microenvironment or niche during homeostasis. It was recently shown that primitive HSCs in the bone marrow prefer to utilize anaerobic glycolysis to meet their energy demands and have lower rates of oxygen consumption and lower ATP levels. Hypoxia-inducible factor-α (Hif-1α) is a master regulator of cellular metabolism. With hundreds of downstream target genes and crosstalk with other signaling pathways, it regulates various aspects of metabolism from the oxidative stress response to glycolysis and mitochondrial respiration. Hif-1α is highly expressed in HSCs, where it regulates their function and metabolic phenotype. However, the regulation of Hif-1α in HSCs is not entirely understood. The homeobox transcription factor myeloid ecotropic viral integration site 1 (Meis1) is expressed in the most primitive HSCs populations, and it is required for primitive hematopoiesis. Recent reports suggest that Meis1 is required for normal adult HSC function by regulating the metabolism and redox state of HSCs transcriptionally through Hif-1α and Hif-2α. CRITICAL ISSUES Given the profound effect of redox status on HSC function, it is critical to fully characterize the intrinsic, and microenvironment-related mechanisms of metabolic and redox regulation in HSCs. FUTURE DIRECTIONS Future studies will be needed to elucidate the link between HSC metabolism and HSC fates, including quiescence, self-renewal, differentiation, apoptosis, and migration.
Collapse
Affiliation(s)
- Cheng Cheng Zhang
- Division of Cardiology, Departments of Physiology and Developmental Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hesham A. Sadek
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
34
|
Forgacova K, Savvulidi F, Sefc L, Linhartova J, Necas E. All hematopoietic stem cells engraft in submyeloablatively irradiated mice. Biol Blood Marrow Transplant 2013; 19:713-9. [PMID: 23422843 DOI: 10.1016/j.bbmt.2013.02.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/11/2013] [Indexed: 12/24/2022]
Abstract
Significant controversy exists regarding the impact of hematopoietic stroma damage by irradiation on the efficiency of engraftment of intravenously transplanted stem cells. It was previously demonstrated that in normal syngenic mice, all intravenously transplanted donor stem cells, present in the bone marrow, compete equally with those of the host. In this study, we comprehensively compared the blood cell production derived from transplanted donor stem cells with that from the host stem cells surviving various doses of submyeloablative irradiation. We compared the partial chimerism resulting from transplantation with theoretical estimates that assumed transplantation efficiencies ranging from 100% to 20%. The highest level of consensus between the experimental and the theoretical results was 100% for homing and engraftment (ie, the utilization of all transplanted stem cells). These results point to a very potent mechanism through which intravenously administered hematopoietic stem cells are captured from circulation, engraft in the hematopoietic tissue, and contribute to blood cell production in irradiated recipients. The damage done to hematopoietic stroma and to the trabecular bone by submyeloablative doses of ionizing radiation does not negatively affect the homing and engraftment mechanisms of intravenously transplanted hematopoietic progenitor and stem cells.
Collapse
Affiliation(s)
- Katarina Forgacova
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, Prague, Czech Republic
| | | | | | | | | |
Collapse
|
35
|
MacLean AL, Lo Celso C, Stumpf MPH. Population dynamics of normal and leukaemia stem cells in the haematopoietic stem cell niche show distinct regimes where leukaemia will be controlled. J R Soc Interface 2013; 10:20120968. [PMID: 23349436 PMCID: PMC3627104 DOI: 10.1098/rsif.2012.0968] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Haematopoietic stem cells (HSCs) are responsible for maintaining immune cells, red blood cells and platelets throughout life. HSCs must be located in their ecological niche (the bone marrow) to function correctly, that is, to regenerate themselves and their progeny; the latter eventually exit the bone marrow and enter circulation. We propose that cells with oncogenic potential-cancer/leukaemia stem cells (LSC)-and their progeny will also occupy this niche. Mathematical models, which describe the dynamics of HSCs, LSCs and their progeny allow investigation into the conditions necessary for defeating a malignant invasion of the niche. Two such models are developed and analysed here. To characterize their behaviour, we use an inferential framework that allows us to study regions in parameter space that give rise to desired behaviour together with an assessment of the robustness of the dynamics. Using this approach, we map out conditions under which HSCs can outcompete LSCs. In therapeutic applications, we clearly want to drive haematopoiesis into such regimes and the current analysis provide some guidance as to how we can identify new therapeutic targets. Our results suggest that maintaining a viable population of HSCs and their progenies in the niche may often already be nearly sufficient to eradicate LSCs from the system.
Collapse
Affiliation(s)
- Adam L MacLean
- Theoretical Systems Biology, Division of Molecular Biosciences, Imperial College London, Sir Ernst Chain Building, London SW7 2AZ, UK
| | | | | |
Collapse
|
36
|
Amorin B, Alegretti AP, Valim VDS, Silva AMPD, Silva MALD, Sehn F, Silla L. Characteristics of Mesenchymal Stem Cells under Hypoxia. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/cellbio.2013.21002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
37
|
Lee J, Li M, Milwid J, Dunham J, Vinegoni C, Gorbatov R, Iwamoto Y, Wang F, Shen K, Hatfield K, Enger M, Shafiee S, McCormack E, Ebert BL, Weissleder R, Yarmush ML, Parekkadan B. Implantable microenvironments to attract hematopoietic stem/cancer cells. Proc Natl Acad Sci U S A 2012; 109:19638-43. [PMID: 23150542 PMCID: PMC3511730 DOI: 10.1073/pnas.1208384109] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The environments that harbor hematopoietic stem and progenitor cells are critical to explore for a better understanding of hematopoiesis during health and disease. These compartments often are inaccessible for controlled and rapid experimentation, thus limiting studies to the evaluation of conventional cell culture and transgenic animal models. Here we describe the manufacture and image-guided monitoring of an engineered microenvironment with user-defined properties that recruits hematopoietic progenitors into the implant. Using intravital imaging and fluorescence molecular tomography, we show in real time that the cell homing and retention process is efficient and durable for short- and long-term engraftment studies. Our results indicate that bone marrow stromal cells, precoated on the implant, accelerate the formation of new sinusoidal blood vessels with vascular integrity at the microcapillary level that enhances the recruitment hematopoietic progenitor cells to the site. This implantable construct can serve as a tool enabling the study of hematopoiesis.
Collapse
Affiliation(s)
- Jungwoo Lee
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children in Boston, MA 02114
| | - Matthew Li
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children in Boston, MA 02114
- Harvard-MIT Health Sciences and Technology, Cambridge, MA 02139
| | - Jack Milwid
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children in Boston, MA 02114
- Harvard-MIT Health Sciences and Technology, Cambridge, MA 02139
| | - Joshua Dunham
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Claudio Vinegoni
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Rostic Gorbatov
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Yoshiko Iwamoto
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Fangjing Wang
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children in Boston, MA 02114
| | - Keyue Shen
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children in Boston, MA 02114
| | - Kimberley Hatfield
- Section of Hematology, Department of Medicine, Haukeland University Hospital, 5021 Bergen, Norway
| | - Marianne Enger
- Gade Institute, University of Bergen, 5020 Bergen, Norway
| | - Sahba Shafiee
- Department of Hematology, Institute of Internal Medicine, Haukeland University Hospital, University of Bergen, 5020 Bergen, Norway
| | - Emmet McCormack
- Department of Hematology, Institute of Internal Medicine, Haukeland University Hospital, University of Bergen, 5020 Bergen, Norway
| | - Benjamin L. Ebert
- Department of Hematology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02114
- The Harvard Stem Cell Institute, Boston, MA 02115; and
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114
| | - Martin L. Yarmush
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children in Boston, MA 02114
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ 08854
| | - Biju Parekkadan
- Center for Engineering in Medicine and Surgical Services, Massachusetts General Hospital, Harvard Medical School and Shriners Hospital for Children in Boston, MA 02114
- The Harvard Stem Cell Institute, Boston, MA 02115; and
| |
Collapse
|
38
|
Liu Y, Chen XH, Si YJ, Li ZJ, Gao L, Gao L, Zhang C, Zhang X. Reconstruction of hematopoietic inductive microenvironment after transplantation of VCAM-1-modified human umbilical cord blood stromal cells. PLoS One 2012; 7:e31741. [PMID: 22384064 PMCID: PMC3285638 DOI: 10.1371/journal.pone.0031741] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 01/12/2012] [Indexed: 01/13/2023] Open
Abstract
The hematopoietic inductive microenvironment (HIM) is where hematopoietic stem/progenitor cells grow and develop. Hematopoietic stromal cells were the key components of the HIM. In our previous study, we had successfully cultured and isolated human cord blood–derived stromal cells (HUCBSCs) and demonstrated that they could secret hemopoietic growth factors such as GM-CSF, TPO, and SCF. However, it is still controversial whether HUCBSCs can be used for reconstruction of HIM. In this study, we first established a co-culture system of HUCBSCs and cord blood CD34+ cells and then determined that using HUCBSCs as the adherent layer had significantly more newly formed colonies of each hematopoietic lineage than the control group, indicating that HUCBSCs had the ability to promote the proliferation of hematopoietic stem cells/progenitor cells. Furthermore, the number of colonies was significantly higher in vascular cell adhesion molecule-1 (VCAM-1)-modified HUCBSCs, suggesting that the ability of HUCBSCs in promoting the proliferation of hematopoietic stem cells/progenitor cells was further enhanced after having been modified with VCAM-1. Next, HUCBSCs were infused into a radiation-damaged animal model, in which the recovery of hematopoiesis was observed. The results demonstrate that the transplanted HUCBSCs were “homed in” to bone marrow and played roles in promoting the recovery of irradiation-induced hematopoietic damage and repairing HIM. Compared with the control group, the HUCBSC group had significantly superior effectiveness in terms of the recovery time for hemogram and myelogram, CFU-F, CFU-GM, BFU-E, and CFU-Meg. Such differences were even more significant in VCAM-1-modified HUCBSCs group. We suggest that HUCBSCs are able to restore the functions of HIM and promote the recovery of radiation-induced hematopoietic damage. VCAM-1 plays an important role in supporting the repair of HIM damage.
Collapse
Affiliation(s)
- Yao Liu
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Xing-hua Chen
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Ying-jian Si
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
- Department of Pediatric Hematology/Oncology, BaYi Children's Hospital, The Military General Hospital of Beijing, Beijing, China
| | - Zhong-jun Li
- Department of Blood Transfusion, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Lei Gao
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Li Gao
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Cheng Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Xi Zhang
- Department of Hematology, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
- * E-mail:
| |
Collapse
|
39
|
Poncin G, Beaulieu A, Humblet C, Thiry A, Oda K, Boniver J, Defresne MP. Characterization of spontaneous bone marrow recovery after sublethal total body irradiation: importance of the osteoblastic/adipocytic balance. PLoS One 2012; 7:e30818. [PMID: 22363493 PMCID: PMC3281884 DOI: 10.1371/journal.pone.0030818] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 12/21/2011] [Indexed: 12/16/2022] Open
Abstract
Many studies have already examined the hematopoietic recovery after irradiation but paid with very little attention to the bone marrow microenvironment. Nonetheless previous studies in a murine model of reversible radio-induced bone marrow aplasia have shown a significant increase in alkaline phosphatase activity (ALP) prior to hematopoietic regeneration. This increase in ALP activity was not due to cell proliferation but could be attributed to modifications of the properties of mesenchymal stem cells (MSC). We thus undertook a study to assess the kinetics of the evolution of MSC correlated to their hematopoietic supportive capacities in mice treated with sub lethal total body irradiation. In our study, colony-forming units – fibroblasts (CFU-Fs) assay showed a significant MSC rate increase in irradiated bone marrows. CFU-Fs colonies still possessed differentiation capacities of MSC but colonies from mice sacrificed 3 days after irradiation displayed high rates of ALP activity and a transient increase in osteoblastic markers expression while pparγ and neuropilin-1 decreased. Hematopoietic supportive capacities of CFU-Fs were also modified: as compared to controls, irradiated CFU-Fs significantly increased the proliferation rate of hematopoietic precursors and accelerated the differentiation toward the granulocytic lineage. Our data provide the first evidence of the key role exerted by the balance between osteoblasts and adipocytes in spontaneous bone marrow regeneration. First, (pre)osteoblast differentiation from MSC stimulated hematopoietic precursor's proliferation and granulopoietic regeneration. Then, in a second time (pre)osteoblasts progressively disappeared in favour of adipocytic cells which down regulated the proliferation and granulocytic differentiation and then contributed to a return to pre-irradiation conditions.
Collapse
Affiliation(s)
- Géraldine Poncin
- Department of Cytology & Histology, University of Liège, CHU-B23, Liège, Belgium.
| | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Over the past two decades there have been unprecedented advances in the capabilities for live cell imaging using light and confocal microscopy. Together with the discovery of green fluorescent protein and its derivatives and the development of a vast array of fluorescent imaging probes and conjugates, it is now possible to image virtually any intracellular or extracellular protein or structure. Traditional static imaging of fixed bone cells and tissues takes a snapshot view of events at a specific time point, but can often miss the dynamic aspects of the events being investigated. This chapter provides an overview of the application of live cell imaging approaches for the study of bone cells and bone organ cultures. Rather than emphasizing technical aspects of the imaging equipment, we have focused on what we consider to be the important principles that are of most practical use for an investigator setting up these techniques in their own laboratory, together with detailed protocols that our laboratory has used for live imaging of bone cell and organ cultures.
Collapse
Affiliation(s)
- Sarah L Dallas
- School of Dentistry/Department of Oral Biology, University of Missouri, Kansas City, MO, USA.
| | | |
Collapse
|
41
|
Barrett O, Sottocornola R, Lo Celso C. In vivo imaging of hematopoietic stem cells in the bone marrow niche. Methods Mol Biol 2012; 916:231-242. [PMID: 22914945 DOI: 10.1007/978-1-61779-980-8_18] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Even though hematopoietic stem cells (HSC) are amongst the first somatic stem cells exploited for therapeutic purposes, their application is still limited by the inability to expand them ex vivo without impairing their function. Moreover, it has recently emerged that several types of leukemia develop and relapse through complex interactions with bone marrow (BM) components and may directly affect the HSC and their niche. Increasing attention has therefore been dedicated to the BM microenvironment the HSC reside in, with the view that a better understanding of the molecular regulators of HSC-niche interaction in vivo will allow improving HSC mobilization, collection and transplantation and provide clues for the development of innovative leukemia treatments. This chapter focuses on a recently established technique for the visualization of transplanted hematopoietic stem and progenitor cells (HSPC) within the calvarium bone marrow of live mice (Lo Celso et al. Nature 457:92-96, 2007). Intravital microscopy is a rapidly developing field, driven by constant improvement in both detection technologies (i.e., spatial resolution, depth of penetration, spectral definition) and probe availability (i.e., increasingly sophisticated genetic and chemical reporter systems). We therefore discuss the current limitations and challenges related to intravital microscopy of the HSC niche and introduce a number of potential imaging approaches, which could be promising candidates for future development of this technique.
Collapse
Affiliation(s)
- Oliver Barrett
- Division of Cell and Molecular Biology, Imperial College, London, UK
| | | | | |
Collapse
|
42
|
Zhao W, Schafer S, Choi J, Yamanaka YJ, Lombardi ML, Bose S, Carlson AL, Phillips JA, Teo W, Droujinine IA, Cui CH, Jain RK, Lammerding J, Love JC, Lin CP, Sarkar D, Karnik R, Karp JM. Cell-surface sensors for real-time probing of cellular environments. NATURE NANOTECHNOLOGY 2011; 6:524-31. [PMID: 21765401 PMCID: PMC3163485 DOI: 10.1038/nnano.2011.101] [Citation(s) in RCA: 170] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2010] [Accepted: 06/02/2011] [Indexed: 05/16/2023]
Abstract
The ability to explore cell signalling and cell-to-cell communication is essential for understanding cell biology and developing effective therapeutics. However, it is not yet possible to monitor the interaction of cells with their environments in real time. Here, we show that a fluorescent sensor attached to a cell membrane can detect signalling molecules in the cellular environment. The sensor is an aptamer (a short length of single-stranded DNA) that binds to platelet-derived growth factor (PDGF) and contains a pair of fluorescent dyes. When bound to PDGF, the aptamer changes conformation and the dyes come closer to each other, producing a signal. The sensor, which is covalently attached to the membranes of mesenchymal stem cells, can quantitatively detect with high spatial and temporal resolution PDGF that is added in cell culture medium or secreted by neighbouring cells. The engineered stem cells retain their ability to find their way to the bone marrow and can be monitored in vivo at the single-cell level using intravital microscopy.
Collapse
Affiliation(s)
- Weian Zhao
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Sebastian Schafer
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Jonghoon Choi
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Yvonne J. Yamanaka
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
- Department of Biological Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Maria L. Lombardi
- Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Suman Bose
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Alicia L. Carlson
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Wellman Center for Photomedicine and Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, 40 Blossom Street, Boston, Massachusetts 02114, USA
| | - Joseph A. Phillips
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Weisuong Teo
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Ilia A. Droujinine
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Cheryl H. Cui
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Rakesh K. Jain
- Edwin L. Steele Laboratory, Department of Radiation Oncology, Massachusetts General Hospital/Harvard Medical School, 55 Fruit Street, Boston, Massachusetts 02114, USA
| | - Jan Lammerding
- Department of Medicine, Brigham and Women's Hospital/Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - J. Christopher Love
- Department of Chemical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Charles P. Lin
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Wellman Center for Photomedicine and Center for Systems Biology, Massachusetts General Hospital/Harvard Medical School, 40 Blossom Street, Boston, Massachusetts 02114, USA
| | - Debanjan Sarkar
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| | - Rohit Karnik
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, Massachusetts 02139, USA
| | - Jeffrey M. Karp
- Center for Regenerative Therapeutics & Department of Medicine, Brigham & Women's Hospital, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Medical School, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard Stem Cell Institute, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
- Harvard-MIT Division of Health Science and Technology, 65 Landsdowne Street, Cambridge, Massachusetts 02139, USA
| |
Collapse
|
43
|
Tew KD, Manevich Y, Grek C, Xiong Y, Uys J, Townsend DM. The role of glutathione S-transferase P in signaling pathways and S-glutathionylation in cancer. Free Radic Biol Med 2011; 51:299-313. [PMID: 21558000 PMCID: PMC3125017 DOI: 10.1016/j.freeradbiomed.2011.04.013] [Citation(s) in RCA: 143] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2011] [Revised: 04/07/2011] [Accepted: 04/07/2011] [Indexed: 12/12/2022]
Abstract
Glutathione S-transferase P is abundantly expressed in some mammalian tissues, particularly those associated with malignancies. While the enzyme can catalyze thioether bond formation between some electrophilic chemicals and GSH, novel nondetoxification functions are now ascribed to it. This review summarizes recent material that implicates GSTP in mediating S-glutathionylation of specific clusters of target proteins and in reactions that define a negative regulatory role in some kinase pathways through ligand or protein:protein interactions. It is becoming apparent that GSTP participates in the maintenance of cellular redox homeostasis through a number of convergent and divergent mechanisms. Moreover, drug platforms that have GSTP as a target have produced some interesting preclinical and clinical candidates.
Collapse
Affiliation(s)
- Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Megens RTA, Kemmerich K, Pyta J, Weber C, Soehnlein O. Intravital imaging of phagocyte recruitment. Thromb Haemost 2011; 105:802-10. [PMID: 21437362 DOI: 10.1160/th10-11-0735] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 03/02/2011] [Indexed: 12/28/2022]
Abstract
Extravasation of neutrophils and monocytes is a hallmark event in acute and chronic inflammation. Owing to recent improvements in optical imaging techniques, the classical leukocyte extravasation cascade has been refined with intermediate steps being added. Further studies have shown tissue specific leukocyte recruitment patterns, thus allowing for more selective targeting. Here we focus on recent advances in intravital imaging of leukocyte recruitment by means of optical imaging techniques and emphasise the translation thereof into tissue-specific recruitment to the lungs, the liver and large arteries.
Collapse
Affiliation(s)
- R T A Megens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-University Munich, Munich, Germany.
| | | | | | | | | |
Collapse
|
45
|
Affiliation(s)
- Yoon-Young Jang
- From the Sidney Kimmel Comprehensive Cancer Center; Stem Cell Program, Institute for Cell Engineering; and Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Zhaohui Ye
- From the Sidney Kimmel Comprehensive Cancer Center; Stem Cell Program, Institute for Cell Engineering; and Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Linzhao Cheng
- From the Sidney Kimmel Comprehensive Cancer Center; Stem Cell Program, Institute for Cell Engineering; and Department of Medicine, Division of Hematology, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
46
|
Tew KD, Townsend DM. Regulatory functions of glutathione S-transferase P1-1 unrelated to detoxification. Drug Metab Rev 2011; 43:179-93. [PMID: 21351850 DOI: 10.3109/03602532.2011.552912] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Glutathione S-transferase P1-1 (GSTP) is one member of the family of GSTs and is ubiquitously expressed in human tissues. The literature is replete with reports of high levels of GSTP linked either with cancer incidence or drug resistance, and yet no entirely cogent explanation for these correlations exists. The catalytic detoxification properties of the GST isozyme family have been a primary research focus for the last four decades. However, it has become apparent that they have undergone structural and functional convergence where evolutionary selective pressures have favored the emergence of noncatalytic properties of GSTP that has imbued this isozyme with expanded biological importance. For example, GSTP has now been linked with two cell-signaling functions that are critical to survival. Through protein:protein interactions, GSTP can sequester c-jun N-terminal kinase (JNK) and act as a negative regulator of this stress kinase. Pharmacologically, this activity has been linked with the activity of GSTP inhibitors in stimulating myeloproliferation. In addition, GSTP is linked with the forward S-glutathionylation reaction, a post-translational modification that impacts the function/activity of a number of proteins. Catalytic reversal of S-glutathionylation is well characterized, but the role of GSTP in catalyzing the forward reaction contributes to the "glutathionylation cycle." Moreover, GSTP is itself susceptible to S-glutathionylation, providing an autoregulatory loop for the cycle. Because oxidative stress regulates both S-glutathionylation and JNK-signaling pathways, such links may help to explain the aberrant patterns of GSTP expression in the cancer phenotype. As such, there is an ongoing preclinical and clinical platform of drug discovery and development around GSTP.
Collapse
Affiliation(s)
- Kenneth D Tew
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425-5050, USA.
| | | |
Collapse
|
47
|
Lo Celso C, Lin CP, Scadden DT. In vivo imaging of transplanted hematopoietic stem and progenitor cells in mouse calvarium bone marrow. Nat Protoc 2011; 6:1-14. [PMID: 21212779 PMCID: PMC3382040 DOI: 10.1038/nprot.2010.168] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
In vivo imaging of transplanted hematopoietic stem and progenitor cells (HSPCs) was developed to investigate the relationship between HSPCs and components of their microenvironment in the bone marrow. In particular, it allows a direct observation of the behavior of hematopoietic cells during the first few days after transplantation, when the critical events in homing and early engraftment are occurring. By directly imaging these events in living animals, this method permits a detailed assessment of functions previously evaluated by crude assessments of cell counts (homing) or after prolonged periods (engraftment). This protocol offers a new means of investigating the role of cell-intrinsic and cell-extrinsic molecular regulators of hematopoiesis during the early stages of transplantation, and it is the first to allow the study of cell-cell interactions within the bone marrow in three dimensions and in real time. In this paper, we describe how to isolate, label and inject HSPCs, as well as how to perform calvarium intravital microscopy and analyze the resulting images. A typical experiment can be performed and analyzed in ∼1 week.
Collapse
|
48
|
Simsek T, Kocabas F, Zheng J, Deberardinis RJ, Mahmoud AI, Olson EN, Schneider JW, Zhang CC, Sadek HA. The distinct metabolic profile of hematopoietic stem cells reflects their location in a hypoxic niche. Cell Stem Cell 2010; 7:380-90. [PMID: 20804973 DOI: 10.1016/j.stem.2010.07.011] [Citation(s) in RCA: 816] [Impact Index Per Article: 58.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2010] [Revised: 05/24/2010] [Accepted: 07/14/2010] [Indexed: 12/15/2022]
Abstract
Bone marrow transplantation is the primary therapy for numerous hematopoietic disorders. The efficiency of bone marrow transplantation depends on the function of long-term hematopoietic stem cells (LT-HSCs), which is markedly influenced by their hypoxic niche. Survival in this low-oxygen microenvironment requires significant metabolic adaptation. Here, we show that LT-HSCs utilize glycolysis instead of mitochondrial oxidative phosphorylation to meet their energy demands. We used flow cytometry to identify a unique low mitochondrial activity/glycolysis-dependent subpopulation that houses the majority of hematopoietic progenitors and LT-HSCs. Finally, we demonstrate that Meis1 and Hif-1alpha are markedly enriched in LT-HSCs and that Meis1 regulates HSC metabolism through transcriptional activation of Hif-1alpha. These findings reveal an important transcriptional network that regulates HSC metabolism.
Collapse
Affiliation(s)
- Tugba Simsek
- Department of Internal Medicine, Division of Cardiology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Grek CL, Townsend DM, Tew KD. The impact of redox and thiol status on the bone marrow: Pharmacological intervention strategies. Pharmacol Ther 2010; 129:172-84. [PMID: 20951732 DOI: 10.1016/j.pharmthera.2010.09.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 09/14/2010] [Indexed: 10/18/2022]
Abstract
Imbalances in cancer cell redox homeostasis provide a platform for new opportunities in the development of anticancer drugs. The control of severe dose-limiting toxicities associated with redox regulation, including myelosuppression and immunosuppression, remains a challenge. Recent evidence implicates a critical role for redox regulation and thiol balance in pathways that control myeloproliferation, hematopoietic progenitor cell mobilization, and immune response. Hematopoietic stem cell (HSC) self-renewal and differentiation are dependent upon levels of intracellular reactive oxygen species (ROS) and niche microenvironments. Redox status and the equilibrium of free thiol:disulfide couples are important in modulating immune response and lymphocyte activation, proliferation and differentiation. This subject matter is the focus of the present review. The potential of redox modulating chemotherapeutics as myeloproliferative and immunomodulatory agents is also covered.
Collapse
Affiliation(s)
- Christina L Grek
- Department of Cell and Molecular Pharmacology and Experimental Therapeutics, Medical University of South Carolina, Charleston, SC 29425, USA
| | | | | |
Collapse
|
50
|
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 DOI: 10.1016/j.bcp.2010.04.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [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
| | | | | |
Collapse
|