51
|
Zavala F, Nombela-Arrieta C, Ben Nasr M, Fiorina P. Editorial: The Role of Hematopoietic Progenitors in Immune Regulation and Memory. Front Immunol 2021; 12:789139. [PMID: 34868075 PMCID: PMC8633563 DOI: 10.3389/fimmu.2021.789139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 10/28/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Flora Zavala
- Université de Paris, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades, Paris, France
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University of Zurich and University Hospital Zurich, Zurich, Switzerland
| | - Moufida Ben Nasr
- International Center for Type 1 Diabetes, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science, Università di Milano, Milan, Italy.,Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Paolo Fiorina
- International Center for Type 1 Diabetes, Pediatric Clinical Research Center Romeo ed Enrica Invernizzi, Department of Biomedical and Clinical Science, Università di Milano, Milan, Italy.,Nephrology Division, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States.,Division of Endocrinology, Territorial Healthcare Company Fatebenefratelli-Sacco, Milan, Italy
| |
Collapse
|
52
|
Isringhausen S, Mun Y, Kovtonyuk L, Kräutler NJ, Suessbier U, Gomariz A, Spaltro G, Helbling PM, Wong HC, Nagasawa T, Manz MG, Oxenius A, Nombela-Arrieta C. Chronic viral infections persistently alter marrow stroma and impair hematopoietic stem cell fitness. J Exp Med 2021; 218:e20192070. [PMID: 34709350 PMCID: PMC8558839 DOI: 10.1084/jem.20192070] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Revised: 08/11/2021] [Accepted: 10/05/2021] [Indexed: 11/04/2022] Open
Abstract
Chronic viral infections are associated with hematopoietic suppression, bone marrow (BM) failure, and hematopoietic stem cell (HSC) exhaustion. However, how persistent viral challenge and inflammatory responses target BM tissues and perturb hematopoietic competence remains poorly understood. Here, we combine functional analyses with advanced 3D microscopy to demonstrate that chronic infection with lymphocytic choriomeningitis virus leads to (1) long-lasting decimation of the BM stromal network of mesenchymal CXCL12-abundant reticular cells, (2) proinflammatory transcriptional remodeling of remaining components of this key niche subset, and (3) durable functional defects and decreased competitive fitness in HSCs. Mechanistically, BM immunopathology is elicited by virus-specific, activated CD8 T cells, which accumulate in the BM via interferon-dependent mechanisms. Combined antibody-mediated inhibition of type I and II IFN pathways completely preempts degeneration of CARc and protects HSCs from chronic dysfunction. Hence, viral infections and ensuing immune reactions durably impact BM homeostasis by persistently decreasing the competitive fitness of HSCs and disrupting essential stromal-derived, hematopoietic-supporting cues.
Collapse
Affiliation(s)
- Stephan Isringhausen
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - YeVin Mun
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Larisa Kovtonyuk
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - Ute Suessbier
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Alvaro Gomariz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Gianluca Spaltro
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Patrick M. Helbling
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Hui Chyn Wong
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | - Takashi Nagasawa
- Department of Microbiology and Immunology, Osaka University, Osaka, Japan
| | - Markus G. Manz
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| | | | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
53
|
Wu Q, Zhang J, Lucas D. Anatomy of Hematopoiesis and Local Microenvironments in the Bone Marrow. Where to? Front Immunol 2021; 12:768439. [PMID: 34858426 PMCID: PMC8632041 DOI: 10.3389/fimmu.2021.768439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 10/13/2021] [Indexed: 11/28/2022] Open
Abstract
The shape and spatial organization -the anatomy- of a tissue profoundly influences its function. Knowledge of the anatomical relationships between parent and daughter cells is necessary to understand differentiation and how the crosstalk between the different cells in the tissue leads to physiological maintenance and pathological perturbations. Blood cell production takes place in the bone marrow through the progressive differentiation of stem cells and progenitors. These are maintained and regulated by a heterogeneous microenvironment composed of stromal and hematopoietic cells. While hematopoiesis has been studied in extraordinary detail through functional and multiomics approaches, much less is known about the spatial organization of blood production and how local cues from the microenvironment influence this anatomy. Here, we discuss some of the studies that revealed a complex anatomy of hematopoiesis where discrete local microenvironments spatially organize and regulate specific subsets of hematopoietic stem cells and/or progenitors. We focus on the open questions in the field and discuss how new tools and technological advances are poised to transform our understanding of the anatomy of hematopoiesis.
Collapse
Affiliation(s)
- Qingqing Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, United States
| | - Jizhou Zhang
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, United States
| | - Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
54
|
Mun Y, Fazio S, Arrieta CN. Remodeling of the Bone Marrow Stromal Microenvironment During Pathogenic Infections. Curr Top Microbiol Immunol 2021; 434:55-81. [PMID: 34850282 DOI: 10.1007/978-3-030-86016-5_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
The bone marrow (BM) is the primary hematopoietic organ and a hub in which organismal demands for blood cellular output are systematically monitored. BM tissues are additionally home to a plethora of mature immune cell types, providing functional environments for the activation of immune responses and acting as preferred anatomical reservoirs for cells involved in immunological memory. Stromal cells of the BM microenvironment crucially govern different aspects of organ function, by structuring tissue microanatomy and by directly providing essential regulatory cues to hematopoietic and immune components in distinct niches. Emerging evidence demonstrates that stromal networks are endowed with remarkable functional and structural plasticity. Stress-induced adaptations of stromal cells translate into demand-driven hematopoiesis. Furthermore, aberrations of stromal integrity arising from pathological conditions critically contribute to the dysregulation of BM function. Here, we summarize our current understanding of the alterations that pathogenic infections and ensuing inflammatory conditions elicit on the global topography of the BM microenvironment, the integrity of anatomical niches and cellular interactions, and ultimately, on the regulatory function of diverse stromal subsets.
Collapse
Affiliation(s)
- YeVin Mun
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Häldeliweg 4, 8032, Zurich, Switzerland
| | - Serena Fazio
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Häldeliweg 4, 8032, Zurich, Switzerland
| | - César Nombela Arrieta
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Häldeliweg 4, 8032, Zurich, Switzerland.
| |
Collapse
|
55
|
Omatsu Y, Higaki K, Nagasawa T. Cellular Niches for Hematopoietic Stem Cells and Lympho-Hematopoiesis in Bone Marrow During Homeostasis and Blood Cancers. Curr Top Microbiol Immunol 2021; 434:33-54. [PMID: 34850281 DOI: 10.1007/978-3-030-86016-5_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Most types of blood cells, including immune cells are generated from hematopoietic stem cells (HSCs) within bone marrow in the adult. Most HSCs are in contact with and require the special microenvironment known as a niche for their maintenance. It has been thought that HSC niches comprise various types of support cells that provide critical signals, including cytokines and extracellular matrix for HSC regulation. However, among these cells, several lines of evidence have demonstrated that the population of bone marrow-specific mesenchymal stem cells, termed CXC chemokine ligand 12 (CXCL12)-abundant reticular (CAR) cells, which overlap strongly with leptin receptor-expressing (LepR+) cells, is the major cellular component of HSC niches. CAR/LepR+ cells give rise to most adipocytes and osteoblasts in adult bone marrow and express much higher levels of HSC niche factors, including cytokines CXCL12 and stem cell factor (SCF), which are essential for HSC maintenance, and transcription factors Foxc1 and Ebf3, which are essential for the formation and maintenance of HSC niches than other types of cells. CAR/LepR+ cells are present in human bone marrow, undergo fibrotic expansion, and have reduced expression of HSC niche factors in hematopoietic malignancies.
Collapse
Affiliation(s)
- Yoshiki Omatsu
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Kei Higaki
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, 565-0871, Osaka, Japan
| | - Takashi Nagasawa
- Laboratory of Stem Cell Biology and Developmental Immunology, Graduate School of Frontier Biosciences and Graduate School of Medicine, WPI Immunology Frontier Research Center, Osaka University, Suita, 565-0871, Osaka, Japan.
| |
Collapse
|
56
|
Dalisson B, Charbonnier B, Aoude A, Gilardino M, Harvey E, Makhoul N, Barralet J. Skeletal regeneration for segmental bone loss: Vascularised grafts, analogues and surrogates. Acta Biomater 2021; 136:37-55. [PMID: 34626818 DOI: 10.1016/j.actbio.2021.09.053] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 09/25/2021] [Accepted: 09/28/2021] [Indexed: 02/08/2023]
Abstract
Massive segmental bone defects (SBD) are mostly treated by removing the fibula and transplanting it complete with blood supply. While revolutionary 50 years ago, this remains the standard treatment. This review considers different strategies to repair SBD and emerging potential replacements for this highly invasive procedure. Prior to the technical breakthrough of microsurgery, researchers in the 1960s and 1970s had begun to make considerable progress in developing non autologous routes to repairing SBD. While the breaktthrough of vascularised bone transplantation solved the immediate problem of a lack of reliable repair strategies, much of their prior work is still relevant today. We challenge the assumption that mimicry is necessary or likely to be successful and instead point to the utility of quite crude (from a materials technology perspective), approaches. Together there are quite compelling indications that the body can regenerate entire bone segments with few or no exogenous factors. This is important, as there is a limit to how expensive a bone repair can be and still be widely available to all patients since cost restraints within healthcare systems are not likely to diminish in the near future. STATEMENT OF SIGNIFICANCE: This review is significant because it is a multidisciplinary view of several surgeons and scientists as to what is driving improvement in segmental bone defect repair, why many approaches to date have not succeeded and why some quite basic approaches can be as effective as they are. While there are many reviews of the literature of grafting and bone repair the relative lack of substantial improvement and slow rate of progress in clinical translation is often overlooked and we seek to challenge the reader to consider the issue more broadly.
Collapse
|
57
|
Yip RKH, Rimes JS, Capaldo BD, Vaillant F, Mouchemore KA, Pal B, Chen Y, Surgenor E, Murphy AJ, Anderson RL, Smyth GK, Lindeman GJ, Hawkins ED, Visvader JE. Mammary tumour cells remodel the bone marrow vascular microenvironment to support metastasis. Nat Commun 2021; 12:6920. [PMID: 34836954 PMCID: PMC8626461 DOI: 10.1038/s41467-021-26556-6] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Accepted: 09/27/2021] [Indexed: 12/11/2022] Open
Abstract
Bone marrow is a preferred metastatic site for multiple solid tumours and is associated with poor prognosis and significant morbidity. Accumulating evidence indicates that cancer cells colonise specialised niches within the bone marrow to support their long-term propagation, but the precise location and mechanisms that mediate niche interactions are unknown. Using breast cancer as a model of solid tumour metastasis to the bone marrow, we applied large-scale quantitative three-dimensional imaging to characterise temporal changes in the bone marrow microenvironment during disease progression. We show that mouse mammary tumour cells preferentially home to a pre-existing metaphyseal domain enriched for type H vessels. Metastatic lesion outgrowth rapidly remodelled the local vasculature through extensive sprouting to establish a tumour-supportive microenvironment. The evolution of this tumour microenvironment reflects direct remodelling of the vascular endothelium through tumour-derived granulocyte-colony stimulating factor (G-CSF) in a hematopoietic cell-independent manner. Therapeutic targeting of the metastatic niche by blocking G-CSF receptor inhibited pathological blood vessel remodelling and reduced bone metastasis burden. These findings elucidate a mechanism of 'host' microenvironment hijacking by mammary tumour cells to subvert the local microvasculature to form a specialised, pro-tumorigenic niche.
Collapse
Affiliation(s)
- Raymond K. H. Yip
- grid.1042.7ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC 3010 Australia
| | - Joel S. Rimes
- grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1042.7Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia
| | - Bianca D. Capaldo
- grid.1042.7ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC 3010 Australia
| | - François Vaillant
- grid.1042.7ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC 3010 Australia
| | - Kellie A. Mouchemore
- grid.1018.80000 0001 2342 0938School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086 Australia ,grid.482637.cOlivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084 Australia
| | - Bhupinder Pal
- grid.1042.7ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia ,grid.1018.80000 0001 2342 0938School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086 Australia ,grid.482637.cOlivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084 Australia
| | - Yunshun Chen
- grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1042.7Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia
| | - Elliot Surgenor
- grid.1042.7ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia
| | - Andrew J. Murphy
- grid.1002.30000 0004 1936 7857Department of Immunology, Monash University, Melbourne, VIC Australia ,grid.1051.50000 0000 9760 5620Division of Immunometabolism, Baker Heart & Diabetes Institute, Melbourne, VIC Australia
| | - Robin L. Anderson
- grid.1018.80000 0001 2342 0938School of Cancer Medicine, La Trobe University, Bundoora, VIC 3086 Australia ,grid.482637.cOlivia Newton-John Cancer Research Institute, Heidelberg, VIC 3084 Australia
| | - Gordon K. Smyth
- grid.1042.7Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XSchool of Mathematics and Statistics, The University of Melbourne, Parkville, VIC 3010 Australia
| | - Geoffrey J. Lindeman
- grid.1042.7ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medicine, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1055.10000000403978434Department of Medical Oncology and Parkville Familial Cancer Centre, The Peter MacCallum Cancer Centre and Royal Melbourne Hospital, Parkville, VIC 3050 Australia
| | - Edwin D. Hawkins
- grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC 3010 Australia ,grid.1042.7Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia
| | - Jane E. Visvader
- grid.1042.7ACRF Cancer Biology and Stem Cells Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia ,grid.1008.90000 0001 2179 088XDepartment of Medical Biology, The University of Melbourne, Parkville, VIC 3010 Australia
| |
Collapse
|
58
|
Matteini F, Mulaw MA, Florian MC. Aging of the Hematopoietic Stem Cell Niche: New Tools to Answer an Old Question. Front Immunol 2021; 12:738204. [PMID: 34858399 PMCID: PMC8631970 DOI: 10.3389/fimmu.2021.738204] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 10/11/2021] [Indexed: 12/31/2022] Open
Abstract
The hematopoietic stem cell (HSC) niche is a specialized microenvironment, where a complex and dynamic network of interactions across multiple cell types regulates HSC function. During the last years, it became progressively clearer that changes in the HSC niche are responsible for specific alterations of HSC behavior. The aging of the bone marrow (BM) microenvironment has been shown to critically contribute to the decline in HSC function over time. Interestingly, while upon aging some niche structures within the BM are degenerated and negatively affect HSC functionality, other niche cells and specific signals are preserved and essential to retaining HSC function and regenerative capacity. These new findings on the role of the aging BM niche critically depend on the implementation of new technical tools, developed thanks to transdisciplinary approaches, which bring together different scientific fields. For example, the development of specific mouse models in addition to coculture systems, new 3D-imaging tools, ossicles, and ex-vivo BM mimicking systems is highlighting the importance of new technologies to unravel the complexity of the BM niche on aging. Of note, an exponential impact in the understanding of this biological system has been recently brought by single-cell sequencing techniques, spatial transcriptomics, and implementation of artificial intelligence and deep learning approaches to data analysis and integration. This review focuses on how the aging of the BM niche affects HSCs and on the new tools to investigate the specific alterations occurring in the BM upon aging. All these new advances in the understanding of the BM niche and its regulatory function on HSCs have the potential to lead to novel therapeutical approaches to preserve HSC function upon aging and disease.
Collapse
Affiliation(s)
- Francesca Matteini
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], Barcelona, Spain
| | - Medhanie A. Mulaw
- Institute for Molecular Medicine and Internal Medicine I, Ulm University and University Hospital Ulm, Ulm, Germany
| | - M. Carolina Florian
- Stem Cell Aging Group, Regenerative Medicine Program, The Bellvitge Institute for Biomedical Research (IDIBELL), Barcelona, Spain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia, P-CMR[C], Barcelona, Spain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| |
Collapse
|
59
|
Engraftment characterization of risk-stratified AML patients in NSGS mice. Blood Adv 2021; 5:4842-4854. [PMID: 34470043 PMCID: PMC9153030 DOI: 10.1182/bloodadvances.2020003958] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 05/19/2021] [Indexed: 11/24/2022] Open
Abstract
PDXs from risk-stratified AML samples are crucial for studying AML biology and testing novel therapeutics. We characterize human AML engraftment in NSGS mice, offering a valuable platform for in vivo testing of targeted therapies.
Acute myeloid leukemia (AML) is the most common acute leukemia in adults. Disease heterogeneity is well documented, and patient stratification determines treatment decisions. Patient-derived xenografts (PDXs) from risk-stratified AML are crucial for studying AML biology and testing novel therapeutics. Despite recent advances in PDX modeling of AML, reproducible engraftment of human AML is primarily limited to high-risk (HR) cases, with inconsistent or very protracted engraftment observed for favorable-risk (FR) and intermediate-risk (IR) patients. We used NSGS mice to characterize the engraftment robustness/kinetics of 28 AML patient samples grouped according to molecular/cytogenetic classification and assessed whether the orthotopic coadministration of patient-matched bone marrow mesenchymal stromal cells (BM MSCs) improves AML engraftment. PDX event-free survival correlated well with the predictable prognosis of risk-stratified AML patients. The majority (85-94%) of the mice were engrafted in bone marrow (BM) independently of the risk group, although HR AML patients showed engraftment levels that were significantly superior to those of FR or IR AML patients. Importantly, the engraftment levels observed in NSGS mice by week 6 remained stable over time. Serial transplantation and long-term culture-initiating cell (LTC-IC) assays revealed long-term engraftment limited to HR AML patients, fitter leukemia-initiating cells (LICs) in HR AML samples, and the presence of AML LICs in the CD34− leukemic fraction, regardless of the risk group. Finally, orthotopic coadministration of patient-matched BM MSCs and AML cells was dispensable for BM engraftment levels but favored peripheralization of engrafted AML cells. This comprehensive characterization of human AML engraftment in NSGS mice offers a valuable platform for in vivo testing of targeted therapies in risk-stratified AML patient samples.
Collapse
|
60
|
Multicolor 3D Confocal Imaging of Thick Tissue Sections. Methods Mol Biol 2021; 2350:95-104. [PMID: 34331281 DOI: 10.1007/978-1-0716-1593-5_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/17/2023]
Abstract
In multicellular organisms, most physiological and pathological processes involve an interplay between various cells and molecules that act both locally and systemically. To understand how these complex and dynamic processes occur in time and space, imaging techniques are key. Advances in tissue processing techniques and microscopy now allow us to probe these processes at a large scale and at the same time at a level of detail previously unachievable. Indeed, it is now possible to reliably quantify multiple protein expression levels at single-cell resolution in whole organs using three-dimensional fluorescence imaging techniques. Here we describe a method to prepare adult mouse bone tissue for multiplexed confocal imaging of thick tissue sections. Up to eight different fluorophores can be multiplexed using this technique and spectrally resolved using standard confocal microscopy. The optical clearing method described allows detection of these fluorophores up to a depth of >700 μm in the far-red. Although the method was initially developed for bone tissue imaging, we have successfully applied it to several other tissue types.
Collapse
|
61
|
Gomariz A, Portenier T, Helbling PM, Isringhausen S, Suessbier U, Nombela-Arrieta C, Goksel O. Modality Attention and Sampling Enables Deep Learning with Heterogeneous Marker Combinations in Fluorescence Microscopy. NAT MACH INTELL 2021; 3:799-811. [PMID: 34541455 PMCID: PMC7611676 DOI: 10.1038/s42256-021-00379-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 07/02/2021] [Indexed: 01/01/2023]
Abstract
Fluorescence microscopy allows for a detailed inspection of cells, cellular networks, and anatomical landmarks by staining with a variety of carefully-selected markers visualized as color channels. Quantitative characterization of structures in acquired images often relies on automatic image analysis methods. Despite the success of deep learning methods in other vision applications, their potential for fluorescence image analysis remains underexploited. One reason lies in the considerable workload required to train accurate models, which are normally specific for a given combination of markers, and therefore applicable to a very restricted number of experimental settings. We herein propose Marker Sampling and Excite - a neural network approach with a modality sampling strategy and a novel attention module that together enable (i) flexible training with heterogeneous datasets with combinations of markers and (ii) successful utility of learned models on arbitrary subsets of markers prospectively. We show that our single neural network solution performs comparably to an upper bound scenario where an ensemble of many networks is naïvely trained for each possible marker combination separately. In addition, we demonstrate the feasibility of this framework in high-throughput biological analysis by revising a recent quantitative characterization of bone marrow vasculature in 3D confocal microscopy datasets and further confirm the validity of our approach on an additional, significantly different dataset of microvessels in fetal liver tissues. Not only can our work substantially ameliorate the use of deep learning in fluorescence microscopy analysis, but it can also be utilized in other fields with incomplete data acquisitions and missing modalities.
Collapse
Affiliation(s)
- Alvaro Gomariz
- Computer-assisted Applications in Medicine, Computer Vision Lab, ETH Zurich, Switzerland
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Switzerland
| | - Tiziano Portenier
- Computer-assisted Applications in Medicine, Computer Vision Lab, ETH Zurich, Switzerland
| | - Patrick M. Helbling
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Switzerland
| | - Stephan Isringhausen
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Switzerland
| | - Ute Suessbier
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, Switzerland
| | - Orcun Goksel
- Computer-assisted Applications in Medicine, Computer Vision Lab, ETH Zurich, Switzerland
- Department of Information Technology, Uppsala University, Sweden
| |
Collapse
|
62
|
Gutjahr JC, Crawford KS, Jensen DR, Naik P, Peterson FC, Samson GPB, Legler DF, Duchene J, Veldkamp CT, Rot A, Volkman BF. The dimeric form of CXCL12 binds to atypical chemokine receptor 1. Sci Signal 2021; 14:14/696/eabc9012. [PMID: 34404752 DOI: 10.1126/scisignal.abc9012] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The pleiotropic chemokine CXCL12 is involved in diverse physiological and pathophysiological processes, including embryogenesis, hematopoiesis, leukocyte migration, and tumor metastasis. It is known to engage the classical receptor CXCR4 and the atypical receptor ACKR3. Differential receptor engagement can transduce distinct cellular signals and effects as well as alter the amount of free, extracellular chemokine. CXCR4 binds both monomeric and the more commonly found dimeric forms of CXCL12, whereas ACKR3 binds monomeric forms. Here, we found that CXCL12 also bound to the atypical receptor ACKR1 (previously known as Duffy antigen/receptor for chemokines or DARC). In vitro nuclear magnetic resonance spectroscopy and isothermal titration calorimetry revealed that dimeric CXCL12 bound to the extracellular N terminus of ACKR1 with low nanomolar affinity, whereas the binding affinity of monomeric CXCL12 was orders of magnitude lower. In transfected MDCK cells and primary human Duffy-positive erythrocytes, a dimeric, but not a monomeric, construct of CXCL12 efficiently bound to and internalized with ACKR1. This interaction between CXCL12 and ACKR1 provides another layer of regulation of the multiple biological functions of CXCL12. The findings also raise the possibility that ACKR1 can bind other dimeric chemokines, thus potentially further expanding the role of ACKR1 in chemokine retention and presentation.
Collapse
Affiliation(s)
- Julia C Gutjahr
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Kyler S Crawford
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Davin R Jensen
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Prachi Naik
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Francis C Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Guerric P B Samson
- Biotechnology Institute Thurgau (BITg), University of Konstanz, 8280 Kreuzlingen, Switzerland
| | - Daniel F Legler
- Biotechnology Institute Thurgau (BITg), University of Konstanz, 8280 Kreuzlingen, Switzerland.,Theodor Kocher Institute, University of Bern, 3012 Bern, Switzerland
| | - Johan Duchene
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, 80336 Munich, Germany
| | | | - Antal Rot
- Centre for Microvascular Research, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK. .,Institute for Cardiovascular Prevention, Ludwig-Maximilians University, 80336 Munich, Germany.,Centre for Inflammation and Therapeutic Innovation, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Brian F Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA.
| |
Collapse
|
63
|
Hofmann J, Keppler SJ. Tissue clearing and 3D imaging - putting immune cells into context. J Cell Sci 2021; 134:271108. [PMID: 34342351 DOI: 10.1242/jcs.258494] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
A better understanding of cell-cell and cell-niche interactions is crucial to comprehend the complexity of inflammatory or pathophysiological scenarios such as tissue damage during viral infections, the tumour microenvironment and neuroinflammation. Optical clearing and 3D volumetric imaging of large tissue pieces or whole organs is a rapidly developing methodology that holds great promise for the in-depth study of cells in their natural surroundings. These methods have mostly been applied to image structural components such as endothelial cells and neuronal architecture. Recent work now highlights the possibility of studying immune cells in detail within their respective immune niches. This Review summarizes recent developments in tissue clearing methods and 3D imaging, with a focus on the localization and quantification of immune cells. We first provide background to the optical challenges involved and their solutions before discussing published protocols for tissue clearing, the limitations of 3D imaging of immune cells and image analysis. Furthermore, we highlight possible applications for tissue clearing and propose future developments for the analysis of immune cells within homeostatic or inflammatory immune niches.
Collapse
Affiliation(s)
- Julian Hofmann
- Institute for Clinical Chemistry and Pathobiochemistry, München rechts der Isar (MRI), Technical University Munich, 81675 Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University Munich, 81675 Munich, Germany
| | - Selina J Keppler
- Institute for Clinical Chemistry and Pathobiochemistry, München rechts der Isar (MRI), Technical University Munich, 81675 Munich, Germany.,TranslaTUM, Center for Translational Cancer Research, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
64
|
Black S, Phillips D, Hickey JW, Kennedy-Darling J, Venkataraaman VG, Samusik N, Goltsev Y, Schürch CM, Nolan GP. CODEX multiplexed tissue imaging with DNA-conjugated antibodies. Nat Protoc 2021; 16:3802-3835. [PMID: 34215862 PMCID: PMC8647621 DOI: 10.1038/s41596-021-00556-8] [Citation(s) in RCA: 351] [Impact Index Per Article: 87.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 04/14/2021] [Indexed: 02/06/2023]
Abstract
Advances in multiplexed imaging technologies have drastically improved our ability to characterize healthy and diseased tissues at the single-cell level. Co-detection by indexing (CODEX) relies on DNA-conjugated antibodies and the cyclic addition and removal of complementary fluorescently labeled DNA probes and has been used so far to simultaneously visualize up to 60 markers in situ. CODEX enables a deep view into the single-cell spatial relationships in tissues and is intended to spur discovery in developmental biology, disease and therapeutic design. Herein, we provide optimized protocols for conjugating purified antibodies to DNA oligonucleotides, validating the conjugation by CODEX staining and executing the CODEX multicycle imaging procedure for both formalin-fixed, paraffin-embedded (FFPE) and fresh-frozen tissues. In addition, we describe basic image processing and data analysis procedures. We apply this approach to an FFPE human tonsil multicycle experiment. The hands-on experimental time for antibody conjugation is ~4.5 h, validation of DNA-conjugated antibodies with CODEX staining takes ~6.5 h and preparation for a CODEX multicycle experiment takes ~8 h. The multicycle imaging and data analysis time depends on the tissue size, number of markers in the panel and computational complexity.
Collapse
Affiliation(s)
- Sarah Black
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Darci Phillips
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - John W Hickey
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Julia Kennedy-Darling
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Akoya Biosciences, Menlo Park, CA, USA
| | - Vishal G Venkataraaman
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nikolay Samusik
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Becton Dickinson, San Jose, CA, USA
| | - Yury Goltsev
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Christian M Schürch
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany.
| | - Garry P Nolan
- Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
65
|
The Impact of Sedentary Lifestyle, High-fat Diet, Tobacco Smoke, and Alcohol Intake on the Hematopoietic Stem Cell Niches. Hemasphere 2021; 5:e615. [PMID: 34291194 PMCID: PMC8288907 DOI: 10.1097/hs9.0000000000000615] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 06/07/2021] [Indexed: 11/25/2022] Open
Abstract
Hematopoietic stem and progenitor cells maintain hematopoiesis throughout life by generating all major blood cell lineages through the process of self-renewal and differentiation. In adult mammals, hematopoietic stem cells (HSCs) primarily reside in the bone marrow (BM) at special microenvironments called “niches.” Niches are thought to extrinsically orchestrate the HSC fate including their quiescence and proliferation. Insight into the HSC niches mainly comes from studies in mice using surface marker identification and imaging to visualize HSC localization and association with niche cells. The advantage of mouse models is the possibility to study the 3-dimensional BM architecture and cell interactions in an intact traceable system. However, this may not be directly translational to human BM. Sedentary lifestyle, unhealthy diet, excessive alcohol intake, and smoking are all known risk factors for various diseases including hematological disorders and cancer, but how do lifestyle factors impact hematopoiesis and the associated niches? Here, we review current knowledge about the HSC niches and how unhealthy lifestyle may affect it. In addition, we summarize epidemiological data concerning the influence of lifestyle factors on hematological disorders and malignancies.
Collapse
|
66
|
Tratwal J, Rojas-Sutterlin S, Bataclan C, Blum S, Naveiras O. Bone marrow adiposity and the hematopoietic niche: A historical perspective of reciprocity, heterogeneity, and lineage commitment. Best Pract Res Clin Endocrinol Metab 2021; 35:101564. [PMID: 34417114 DOI: 10.1016/j.beem.2021.101564] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
PURPOSE Here we review the current knowledge on bone marrow adipocytes (BMAds) as active contributors to the regulation of the hematopoietic niche, and as potentially pivotal players in the progression of hematological malignancies. We highlight the hierarchical and functional heterogeneity of the adipocyte lineage within the bone marrow, and how potentially different contexts dictate their interactions with hematopoietic populations. RECENT FINDINGS Growing evidence associates the adipocyte lineage with important functions in hematopoietic regulation within the BM niche. Initially proposed to serve as negative regulators of the hematopoietic microenvironment, studies have also demonstrated that BMAds positively influence the survival and maintenance of hematopoietic stem cells (HSCs). These seemingly incongruous findings may at least be partially explained by stage-specificity across the adipocytic differentiation axis and by BMAds subtypes, suggesting that the heterogeneity of these populations allows for differential context-based interactions. One such distinction relies on the location of adipocytes. Constitutive bone marrow adipose tissue (cBMAT) historically associates to the "yellow" marrow containing so-called "stable" BMAs larger in size, less responsive to stimuli, and linked to HSC quiescence. On the other hand, regulated bone marrow adipose tissue (rBMAT)-associated adipocytes, also referred to as "labile" are smaller, more responsive to hematopoietic demand and strategically situated in hematopoietically active regions of the skeleton. Here we propose a model where the effect of distinct BM stromal cell populations (BMSC) in hematopoiesis is structured along the BMSC-BMAd differentiation axis, and where the effects on HSC maintenance versus hematopoietic proliferation are segregated. In doing so, it is possible to explain how recently identified, adipocyte-primed leptin receptor-expressing, CXCL12-high adventitial reticular cells (AdipoCARs) and marrow adipose lineage precursor cells (MALPs) best support active hematopoietic cell proliferation, while adipose progenitor cells (APCs) and maturing BMAd gradually lose the capacity to support active hematopoiesis, favoring HSC quiescence. Implicated soluble mediators include MCP-1, PAI-1, NRP1, possibly DPP4 and limiting availability of CXCL12 and SCF. How remodeling occurs within the BMSC-BMAd differentiation axis is yet to be elucidated and will likely unravel a three-way regulation of the hematopoietic, bone, and adipocytic compartments orchestrated by vascular elements. The interaction of malignant hematopoietic cells with BMAds is precisely contributing to unravel specific mechanisms of remodeling. SUMMARY BMAds are important operative components of the hematopoietic microenvironment. Their heterogeneity directs their ability to exert a range of regulatory capacities in a manner dependent on their hierarchical, spatial, and biological context. This complexity highlights the importance of (i) developing experimental tools and nomenclature adapted to address stage-specificity and heterogeneity across the BMSC-BMAd differentiation axis when reporting effects in hematopoiesis, (ii) interpreting gene reporter studies within this framework, and (iii) quantifying changes in all three compartments (hematopoiesis, adiposity and bone) when addressing interdependency.
Collapse
Affiliation(s)
- Josefine Tratwal
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Shanti Rojas-Sutterlin
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Charles Bataclan
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland
| | - Sabine Blum
- Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL) & Department of Biomedical Sciences, University of Lausanne (UNIL), Lausanne, Switzerland; Hematology Service, Departments of Oncology and Laboratory Medicine, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
67
|
Allenby MC, Okutsu N, Brailey K, Guasch J, Zhang Q, Panoskaltsis N, Mantalaris A. A spatiotemporal microenvironment model to improve design of a 3D bioreactor for red cell production. Tissue Eng Part A 2021; 28:38-53. [PMID: 34130508 DOI: 10.1089/ten.tea.2021.0028] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cellular microenvironments provide stimuli including paracrine and autocrine growth factors and physico-chemical cues, which support efficient in vivo cell production unmatched by current in vitro biomanufacturing platforms. While three-dimensional (3D) culture systems aim to recapitulate niche architecture and function of the target tissue/organ, they are limited in accessing spatiotemporal information to evaluate and optimize in situ cell/tissue process development. Herein, a mathematical modelling framework is parameterized by single-cell phenotypic imaging and multiplexed biochemical assays to simulate the non-uniform tissue distribution of nutrients/metabolites and growth factors in cell niche environments. This model is applied to a bone marrow mimicry 3D perfusion bioreactor containing dense stromal and hematopoietic tissue with limited red blood cell (RBC) egress. The model characterized an imbalance between endogenous cytokine production and nutrient starvation within the microenvironmental niches, and recommended increased cell inoculum density and enhanced medium exchange, guiding the development of a miniaturized prototype bioreactor. The second-generation prototype improved the distribution of nutrients and growth factors and supported a 50-fold increase in RBC production efficiency. This image-informed bioprocess modelling framework leverages spatiotemporal niche information to enhance biochemical factor utilization and improve cell manufacturing in 3D systems.
Collapse
Affiliation(s)
- Mark Colin Allenby
- Queensland University of Technology, 1969, Institute of Health and Biomedical Innovation (IHBI), Kelvin Grove, Queensland, Australia.,Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Naoki Okutsu
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Kate Brailey
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Joana Guasch
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Qiming Zhang
- Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Nicki Panoskaltsis
- Emory University, 1371, Winship Cancer Institute, Department of Hematology & Medical Oncology, Atlanta, Georgia, United States.,Imperial College London, 4615, Department of Haematology, London, London, United Kingdom of Great Britain and Northern Ireland;
| | - Athanasios Mantalaris
- Georgia Institute of Technology, 1372, BME, Atlanta, Georgia, United States.,Imperial College London, 4615, Department of Chemical Engineering, London, London, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
68
|
Diversity, localization, and (patho)physiology of mature lymphocyte populations in the bone marrow. Blood 2021; 137:3015-3026. [PMID: 33684935 DOI: 10.1182/blood.2020007592] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 02/25/2021] [Indexed: 02/07/2023] Open
Abstract
The bone marrow (BM) is responsible for generating and maintaining lifelong output of blood and immune cells. In addition to its key hematopoietic function, the BM acts as an important lymphoid organ, hosting a large variety of mature lymphocyte populations, including B cells, T cells, natural killer T cells, and innate lymphoid cells. Many of these cell types are thought to visit the BM only transiently, but for others, like plasma cells and memory T cells, the BM provides supportive niches that promote their long-term survival. Interestingly, accumulating evidence points toward an important role for mature lymphocytes in the regulation of hematopoietic stem cells (HSCs) and hematopoiesis in health and disease. In this review, we describe the diversity, migration, localization, and function of mature lymphocyte populations in murine and human BM, focusing on their role in immunity and hematopoiesis. We also address how various BM lymphocyte subsets contribute to the development of aplastic anemia and immune thrombocytopenia, illustrating the complexity of these BM disorders and the underlying similarities and differences in their disease pathophysiology. Finally, we summarize the interactions between mature lymphocytes and BM resident cells in HSC transplantation and graft-versus-host disease. A better understanding of the mechanisms by which mature lymphocyte populations regulate BM function will likely improve future therapies for patients with benign and malignant hematologic disorders.
Collapse
|
69
|
Kwang D, Tjin G, Purton LE. Regulation of murine B lymphopoiesis by stromal cells. Immunol Rev 2021; 302:47-67. [PMID: 34002391 DOI: 10.1111/imr.12973] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/21/2021] [Accepted: 04/30/2021] [Indexed: 12/21/2022]
Abstract
B lymphocytes are crucial for the body's humoral immune response, secreting antibodies generated against foreign antigens to fight infection. Adult murine B lymphopoiesis is initiated in the bone marrow and additional maturation occurs in the spleen. In both these organs, B lymphopoiesis involves interactions with numerous different non-hematopoietic cells, also known as stromal or microenvironment cells, which provide migratory, maturation, and survival signals. A variety of conditional knockout and transgenic mouse models have been used to identify the roles of distinct microenvironment cell types in the regulation of B lymphopoiesis. These studies have revealed that mesenchymal lineage cells and endothelial cells comprise the non-hematopoietic microenvironment cell types that support B lymphopoiesis in the bone marrow. In the spleen, various types of stromal cells and endothelial cells contribute to B lymphocyte maturation. More recently, comprehensive single cell RNA-seq studies have also been used to identify clusters of stromal cell types in the bone marrow and spleen, which will aid in further identifying key regulators of B lymphopoiesis. Here, we review the different types of microenvironment cells and key extrinsic regulators that are known to be involved in the regulation of murine B lymphopoiesis in the bone marrow and spleen.
Collapse
Affiliation(s)
- Diannita Kwang
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Gavin Tjin
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia
| | - Louise E Purton
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, Vic., Australia.,Department of Medicine at St. Vincent's Hospital, The University of Melbourne, Fitzroy, Vic., Australia
| |
Collapse
|
70
|
Cell interactions in the bone marrow microenvironment affecting myeloid malignancies. Blood Adv 2021; 4:3795-3803. [PMID: 32780848 DOI: 10.1182/bloodadvances.2020002127] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/30/2020] [Indexed: 02/06/2023] Open
Abstract
The bone marrow is a complex tissue in which heterogeneous populations of stromal cells interact with hematopoietic cells to dynamically respond to organismal needs in defense, hemostasis, and oxygen delivery. Physiologic challenges modify stromal/hematopoietic cell interactions to generate changes in blood cell production. When either stroma or hematopoietic cells are impaired, the system distorts. The distortions associated with myeloid malignancy are reviewed here and may provide opportunities for therapeutic intervention.
Collapse
|
71
|
Wagner N, Mott K, Upcin B, Stegner D, Schulze H, Ergün S. CXCL12-Abundant Reticular (CAR) Cells Direct Megakaryocyte Protrusions across the Bone Marrow Sinusoid Wall. Cells 2021; 10:722. [PMID: 33804965 PMCID: PMC8063926 DOI: 10.3390/cells10040722] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/16/2021] [Accepted: 03/22/2021] [Indexed: 11/17/2022] Open
Abstract
Megakaryocytes (MKs) release platelets into the lumen of bone marrow (BM) sinusoids while remaining to reside within the BM. The morphogenetic events of this complex process are still not fully understood. We combined confocal laser scanning microscopy with transmission and serial block-face scanning electron microscopy followed by 3D-reconstruction on mouse BM tissue sections. These analyses revealed that MKs in close vicinity to BM sinusoid (BMS) wall first induce the lateral retraction of CXCL12-abundant reticular (CAR) cells (CAR), followed by basal lamina (BL) degradation enabling direct MK-sinusoidal endothelial cells (SECs) interaction. Subsequently, an endothelial engulfment starts that contains a large MK protrusion. Then, MK protrusions penetrate the SEC, transmigrate into the BMS lumen and form proplatelets that are in direct contact to the SEC surface. Furthermore, such processes are induced on several sites, as observed by 3D reconstructions. Our data demonstrate that MKs in interaction with CAR-cells actively induce BMS wall alterations, including CAR-cell retraction, BL degradation, and SEC engulfment containing a large MK protrusion. This results in SEC penetration enabling the migration of MK protrusion into the BMS lumen where proplatelets that are adherent to the luminal SEC surface are formed and contribute to platelet release into the blood circulation.
Collapse
Affiliation(s)
- Nicole Wagner
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany; (N.W.); (B.U.)
| | - Kristina Mott
- Institute of Experimental Biomedicine, Chair I, University Hospital Würzburg, 97080 Würzburg, Germany; (K.M.); (D.S.)
| | - Berin Upcin
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany; (N.W.); (B.U.)
| | - David Stegner
- Institute of Experimental Biomedicine, Chair I, University Hospital Würzburg, 97080 Würzburg, Germany; (K.M.); (D.S.)
| | - Harald Schulze
- Institute of Experimental Biomedicine, Chair I, University Hospital Würzburg, 97080 Würzburg, Germany; (K.M.); (D.S.)
| | - Süleyman Ergün
- Institute of Anatomy and Cell Biology, Julius-Maximilians-University Würzburg, 97070 Würzburg, Germany; (N.W.); (B.U.)
| |
Collapse
|
72
|
Novkovic M, Onder L, Bocharov G, Ludewig B. Topological Structure and Robustness of the Lymph Node Conduit System. Cell Rep 2021; 30:893-904.e6. [PMID: 31968261 DOI: 10.1016/j.celrep.2019.12.070] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 11/26/2019] [Accepted: 12/18/2019] [Indexed: 02/07/2023] Open
Abstract
Fibroblastic reticular cells (FRCs) form a road-like cellular network in lymph nodes (LNs) that provides essential chemotactic, survival, and regulatory signals for immune cells. While the topological characteristics of the FRC network have been elaborated, the network properties of the micro-tubular conduit system generated by FRCs, which drains lymph fluid through a pipeline-like system to distribute small molecules and antigens, has remained unexplored. Here, we quantify the crucial 3D morphometric parameters and determine the topological properties governing the structural organization of the intertwined networks. We find that the conduit system exhibits lesser small-worldness and lower resilience to perturbation compared to the FRC network, while the robust topological organization of both networks is maintained in a lymphotoxin-β-receptor-independent manner. Overall, the high-resolution topological analysis of the "roads-and-pipes" networks highlights essential parameters underlying the functional organization of LN micro-environments and will, hence, advance the development of multi-scale LN models.
Collapse
Affiliation(s)
- Mario Novkovic
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Lucas Onder
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland
| | - Gennady Bocharov
- Marchuk Institute of Numerical Mathematics, Russian Academy of Sciences, Moscow 119333, Russia; Institute for Personalized Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia
| | - Burkhard Ludewig
- Institute of Immunobiology, Kantonsspital St. Gallen, St. Gallen 9007, Switzerland.
| |
Collapse
|
73
|
Distinct Expression Patterns of Cxcl12 in Mesenchymal Stem Cell Niches of Intact and Injured Rodent Teeth. Int J Mol Sci 2021; 22:ijms22063024. [PMID: 33809663 PMCID: PMC8002260 DOI: 10.3390/ijms22063024] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/13/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
Specific stem cell populations within dental mesenchymal tissues guarantee tooth homeostasis and regeneration throughout life. The decision between renewal and differentiation of stem cells is greatly influenced by interactions with stromal cells and extracellular matrix molecules that form the tissue specific stem cell niches. The Cxcl12 chemokine is a general marker of stromal cells and plays fundamental roles in the maintenance, mobilization and migration of stem cells. The aim of this study was to exploit Cxcl12-GFP transgenic mice to study the expression patterns of Cxcl12 in putative dental niches of intact and injured teeth. We showed that endothelial and stromal cells expressed Cxcl12 in the dental pulp tissue of both intact molars and incisors. Isolated non-endothelial Cxcl12+ dental pulp cells cultured in different conditions in vitro exhibited expression of both adipogenic and osteogenic markers, thus suggesting that these cells possess multipotent fates. Taken together, our results show that Cxcl12 is widely expressed in intact and injured teeth and highlight its importance as a key component of the various dental mesenchymal stem cell niches.
Collapse
|
74
|
Dolgalev I, Tikhonova AN. Connecting the Dots: Resolving the Bone Marrow Niche Heterogeneity. Front Cell Dev Biol 2021; 9:622519. [PMID: 33777933 PMCID: PMC7994602 DOI: 10.3389/fcell.2021.622519] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
Single-cell sequencing approaches have transformed our understanding of stem cell systems, including hematopoiesis and its niche within the bone marrow. Recent reports examined the bone marrow microenvironment at single-cell resolution at steady state, following chemotherapy treatment, leukemic onset, and aging. These rapid advancements significantly informed our understanding of bone marrow niche heterogeneity. However, inconsistent representation and nomenclature among the studies hinder a comprehensive interpretation of this body of work. Here, we review recent reports interrogating bone marrow niche architecture and present an integrated overview of the published datasets.
Collapse
Affiliation(s)
- Igor Dolgalev
- Applied Bioinformatics Laboratories, NYU School of Medicine, New York, NY, United States
| | | |
Collapse
|
75
|
Agas D, Sabbieti MG. Archetypal autophagic players through new lenses for bone marrow stem/mature cells regulation. J Cell Physiol 2021; 236:6101-6114. [PMID: 33492700 DOI: 10.1002/jcp.30296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 01/04/2021] [Accepted: 01/11/2021] [Indexed: 12/11/2022]
Abstract
The bone marrow landscape consists of specialized and stem/progenitor cells, which coordinate important tissue-related and systemic physiological features. Within the marrow cavity, stem/progenitor and differentiated hematopoietic and skeletal cells congregate into dynamic functional assemblies throughout specific anatomical regions, termed niches. There is a need for better understanding of the bone marrow microareas, through exploration of the intramural physical and molecular interactions of the distinctive cell populations. The elective liaisons established among the mesenchymal/stromal stem cell and hematopoietic stem cell lineage trees play a key role in orchestrating the stem/mature cell behavior and customized hierarchies within bone marrow cell populations. Recently, the autophagic apparatus has been discovered to be an important feature of bone marrow homeostasis. Autophagy-related factors involved in the labyrinthic and highly dynamic bone marrow workshop redesign the niche framework by coordinating the operational schedule of pluripotent stem and mature cells. The following report summarizes the most recent breakthroughs in our understanding of the intramural relationships between bone marrow cells and key autophagic mediators. Doubtless, the consideration of the autophagy-related and unrelated functions of main players, such as p62, Atg7, Atg5, and Beclin-1 remains a compelling task to thoroughly understand the complex relations between the heterogenic cell types that populate bone marrow.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| | - Maria Giovanna Sabbieti
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Macerata, Italy
| |
Collapse
|
76
|
Banjanin B, Schneider RK. Mesenchymal Stromal Cells as a Cellular Target in Myeloid Malignancy: Chances and Challenges in the Genome Editing of Stromal Alterations. Front Genome Ed 2021; 2:618308. [PMID: 34713241 PMCID: PMC8525402 DOI: 10.3389/fgeed.2020.618308] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/22/2020] [Indexed: 11/13/2022] Open
Abstract
The contribution of bone marrow stromal cells to the pathogenesis and therapy response of myeloid malignancies has gained significant attention over the last decade. Evidence suggests that the bone marrow stroma should not be neglected in the design of novel, targeted-therapies. In terms of gene-editing, the focus of gene therapies has mainly been on correcting mutations in hematopoietic cells. Here, we outline why alterations in the stroma should also be taken into consideration in the design of novel therapeutic strategies but also outline the challenges in specifically targeting mesenchymal stromal cells in myeloid malignancies caused by somatic and germline mutations.
Collapse
Affiliation(s)
- Bella Banjanin
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
- Oncode Institute, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
| | - Rebekka K. Schneider
- Department of Hematology, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
- Oncode Institute, Erasmus Medical Center Cancer Institute, Rotterdam, Netherlands
- Department of Cell Biology, Faculty of Medicine, Institute for Biomedical Engineering, Rheinisch-Westfälische Technische Hochschule (RWTH) Aachen University, Aachen, Germany
| |
Collapse
|
77
|
Dray EL, Ousley CG, McKim DB. Methodological considerations for the enrichment of bone marrow endothelial and mesenchymal stromal cells. Mol Immunol 2021; 131:127-136. [PMID: 33441247 DOI: 10.1016/j.molimm.2020.12.029] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/16/2020] [Accepted: 12/20/2020] [Indexed: 11/16/2022]
Abstract
Stromal cells are critical regulators of bone marrow hematopoietic niches, but assessment of their regulatory roles has been impeded by difficult and ineffective dissociation methods. Here, we methodically address bone marrow stromal cell dissociation. Yield of bone marrow CD45-/Ter119-/CD31+/CD202b+ endothelial cells (ECs) and CD45-/Ter119-/CD44-/PDGFR+ mesenchymal stromal cells (MSCs) were determined by flow cytometry. Liberase DL, Collagenase D, and Dispase II (all supplemented with DNase) enhanced EC and MSC yields, with Dispase II + DNase proving most effective. Combinations of these enzymes did not exhibit additive benefits, nor did the addition of Elastase, TrypLE, Hyaluronidase, or Accutase. Similarly, common mechanical dissociation approaches also proved ineffective. However, the combination of gentle Dispase II + DNase dissociation with magnetic sorting dramatically enriched both ECs and MSCs. This work methodically addressed common approaches for bone marrow stromal dissociation and established an effective approach for enrichment.
Collapse
Affiliation(s)
- Evan L Dray
- Department of Animal Sciences, University of Illinois Urbana-Champaign, USA
| | - Carey G Ousley
- Department of Animal Sciences, University of Illinois Urbana-Champaign, USA
| | - Daniel B McKim
- Department of Animal Sciences, University of Illinois Urbana-Champaign, USA; Division of Nutritional Sciences, University of Illinois Urbana-Champaign, USA; Neuroscience Program, University of Illinois Urbana-Champaign, USA.
| |
Collapse
|
78
|
Abstract
The soft marrow tissues, which are found disseminated throughout bone cavities, are prime sites for hematopoietic cell production, development, and control of immune responses, and regulation of skeletal metabolism. These essential functions are executed through the concerted and finely tuned interaction of a large variety of cell types of hematopoietic and nonhematopoietic origin, through yet largely unknown sophisticated molecular mechanisms. A fundamental insight of the biological underpinnings of organ function can be gained from the microscopic study of the bone marrow (BM), its complex structural organization and the existence of cell-specific spatial associations. Albeit the application of advanced imaging techniques to the analysis of BM has historically proved challenging, recent technological developments now enable the interrogation of organ-wide regions of marrow tissues in three dimensions at high resolution. Here, we provide a detailed experimental protocol for the generation of thick slices of BM from murine femoral cavities, the immunostaining of cellular and structural components within these samples, and their optical clearing, which enhances the depth at which optical sectioning can be performed with standard confocal microscopes. Collectively, the experimental pipeline here described allows for the rendering of single-cell resolution, multidimensional reconstructions of vast volumes of the complex BM microenvironment.
Collapse
|
79
|
Abstract
PURPOSE OF REVIEW The bone marrow is the main site for hematopoiesis. It contains a unique microenvironment that provides niches that support self-renewal and differentiation of hematopoietic stem cells (HSC), multipotent progenitors (MPP), and lineage committed progenitors to produce the large number of blood cells required to sustain life. The bone marrow is notoriously difficult to image; because of this the anatomy of blood cell production -- and how local signals spatially organize hematopoiesis -- are not well defined. Here we review our current understanding of the spatial organization of the mouse bone marrow with a special focus in recent advances that are transforming our understanding of this tissue. RECENT FINDINGS Imaging studies of HSC and their interaction with candidate niches have relied on ex-vivo imaging of fixed tissue. Two recent manuscripts demonstrating live imaging of subsets of HSC in unperturbed bone marrow have revealed unexpected HSC behavior and open the door to examine HSC regulation, in situ, over time. We also discuss recent findings showing that the bone marrow contains distinct microenvironments, spatially organized, that regulate unique aspects of hematopoiesis. SUMMARY Defining the spatial architecture of hematopoiesis in the bone marrow is indispensable to understand how this tissue ensures stepwise, balanced, differentiation to meet organism demand; for deciphering alterations to hematopoiesis during disease; and for designing organ systems for blood cell production ex vivo.
Collapse
Affiliation(s)
- Daniel Lucas
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Medical center
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, USA
| |
Collapse
|
80
|
Panvini FM, Pacini S, Montali M, Barachini S, Mazzoni S, Morganti R, Ciancia EM, Carnicelli V, Petrini M. High NESTIN Expression Marks the Endosteal Capillary Network in Human Bone Marrow. Front Cell Dev Biol 2020; 8:596452. [PMID: 33364234 PMCID: PMC7753038 DOI: 10.3389/fcell.2020.596452] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 11/11/2020] [Indexed: 12/20/2022] Open
Abstract
Hematopoiesis is hosted, supported and regulated by a special bone marrow (BM) microenvironment known as "niche." BM niches have been classified based on micro-anatomic distance from the bone surface into "endosteal" and "central" niches. Whilst different blood vessels have been found in both BM niches in mice, our knowledge of the human BM architecture is much more limited. Here, we have used a combination of markers including NESTIN, CD146, and αSMA labeling different blood vessels in benign human BM. Applying immunohistochemical/immunofluorescence techniques on BM trephines and performing image analysis on almost 300 microphotographs, we detected high NESTIN expression in BM endothelial cells (BMECs) of small arteries (A) and endosteal arterioles (EA), and also in very small vessels we named NESTIN+ capillary-like tubes (NCLTs), not surrounded by sub-endothelial perivascular cells that occasionally reported low levels of NESTIN expression. Statistically, NCLTs were detected within 40 μm from bone trabecula, frequently found in direct contact to the bone line and spatially correlated with hematopoietic stem/progenitor cells. Our results support the expression of NESTIN in human BMECs of EA and A in accordance with the updated classification of murine BM micro-vessels. NCLTs for their peculiar characteristics and micro-anatomical localization have been here proposed as transitional vessels possibly involved in regulating human hematopoiesis.
Collapse
Affiliation(s)
- Francesca M. Panvini
- Institute of Life Sciences, Sant’Anna School of Advanced Studies, Pisa, Italy
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Simone Pacini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Marina Montali
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Stefano Mazzoni
- Department of Translational Research and New Technology in Medicine, University of Pisa, Pisa, Italy
| | - Riccardo Morganti
- Statistical Support to Clinical Trials Department, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy
| | - Eugenio M. Ciancia
- Department of Pathology, Azienda Ospedsaliero Universitaria Pisana, Pisa, Italy
| | - Vittoria Carnicelli
- Department of Surgical, Medical and Molecular Pathology and Critical Care Medicine, University of Pisa, Pisa, Italy
| | - Mario Petrini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
81
|
Deguchi T, Bianchini P, Palazzolo G, Oneto M, Diaspro A, Duocastella M. Volumetric Lissajous confocal microscopy with tunable spatiotemporal resolution. BIOMEDICAL OPTICS EXPRESS 2020; 11:6293-6310. [PMID: 33282491 PMCID: PMC7687945 DOI: 10.1364/boe.400777] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 10/06/2020] [Accepted: 10/06/2020] [Indexed: 05/29/2023]
Abstract
Dynamic biological systems present challenges to existing three-dimensional (3D) optical microscopes because of their continuous temporal and spatial changes. Most techniques are rigid in adapting the acquisition parameters over time, as in confocal microscopy, where a laser beam is sequentially scanned at a predefined spatial sampling rate and pixel dwell time. Such lack of tunability forces a user to provide scan parameters, which may not be optimal, based on the best assumption before an acquisition starts. Here, we developed volumetric Lissajous confocal microscopy to achieve unsurpassed 3D scanning speed with a tunable sampling rate. The system combines an acoustic liquid lens for continuous axial focus translation with a resonant scanning mirror. Accordingly, the excitation beam follows a dynamic Lissajous trajectory enabling sub-millisecond acquisitions of image series containing 3D information at a sub-Nyquist sampling rate. By temporal accumulation and/or advanced interpolation algorithms, the volumetric imaging rate is selectable using a post-processing step at the desired spatiotemporal resolution for events of interest. We demonstrate multicolor and calcium imaging over volumes of tens of cubic microns with 3D acquisition speeds of 30 Hz and frame rates up to 5 kHz.
Collapse
Affiliation(s)
- Takahiro Deguchi
- Nanoscopy & NIC@IIT, Center for Human Technologies, Istituto Italiano di Tecnologia, via E. Melen 83B, 16152 Genoa, Italy
| | - Paolo Bianchini
- Nanoscopy & NIC@IIT, Center for Human Technologies, Istituto Italiano di Tecnologia, via E. Melen 83B, 16152 Genoa, Italy
| | - Gemma Palazzolo
- Enhanced Regenerative Medicine, Istituto Italiano di Tecnologia, via Morego 30, 16163 Genoa, Italy
| | - Michele Oneto
- Nanoscopy & NIC@IIT, Center for Human Technologies, Istituto Italiano di Tecnologia, via E. Melen 83B, 16152 Genoa, Italy
| | - Alberto Diaspro
- Nanoscopy & NIC@IIT, Center for Human Technologies, Istituto Italiano di Tecnologia, via E. Melen 83B, 16152 Genoa, Italy
- Dipartimento di Fisica, Universita di Genova, Via Dodecaneso 33, 16146, Genoa, Italy
| | - Martí Duocastella
- Nanoscopy & NIC@IIT, Center for Human Technologies, Istituto Italiano di Tecnologia, via E. Melen 83B, 16152 Genoa, Italy
- Departament de Física Aplicada, Universitat de Barcelona, C/Marti i Franques 1, 08028 Barcelona, Spain
| |
Collapse
|
82
|
Helbling PM, Piñeiro-Yáñez E, Gerosa R, Boettcher S, Al-Shahrour F, Manz MG, Nombela-Arrieta C. Global Transcriptomic Profiling of the Bone Marrow Stromal Microenvironment during Postnatal Development, Aging, and Inflammation. Cell Rep 2020; 29:3313-3330.e4. [PMID: 31801092 DOI: 10.1016/j.celrep.2019.11.004] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 10/03/2019] [Accepted: 10/31/2019] [Indexed: 12/22/2022] Open
Abstract
Bone marrow (BM) stromal cells provide the regulatory framework for hematopoiesis and contribute to developmental stage-specific niches, such as those preserving hematopoietic stem cells. Despite advances in our understanding of stromal function, little is known about the transcriptional changes that this compartment undergoes throughout lifespan and during adaptation to stress. Using RNA sequencing, we perform transcriptional analyses of four principal stromal subsets, namely CXCL12-abundant reticular, platelet-derived growth factor receptor (PDGFR)-α+Sca1+, sinusoidal, and arterial endothelial cells, from early postnatal, adult, and aged mice. Our data reveal (1) molecular fingerprints defining cell-specific anatomical and functional features, (2) a radical reprogramming of pro-hematopoietic, immune, and matrisomic transcriptional programs during the transition from juvenile stages to adulthood, and (3) the aging-driven progressive upregulation of pro-inflammatory gene expression in stroma. We further demonstrate that transcriptomic pathways elicited in vivo by prototypic microbial molecules are largely recapitulated during aging, thereby supporting the inflammatory basis of age-related adaptations of BM hematopoietic function.
Collapse
Affiliation(s)
- Patrick M Helbling
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Elena Piñeiro-Yáñez
- Bioinformatics Unit, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Rahel Gerosa
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Steffen Boettcher
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Spanish National Cancer Research Center (CNIO), 28029 Madrid, Spain
| | - Markus G Manz
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital and University of Zurich, 8091 Zurich, Switzerland.
| |
Collapse
|
83
|
Single-cell and spatial transcriptomics approaches of the bone marrow microenvironment. Curr Opin Oncol 2020; 32:146-153. [PMID: 31833957 DOI: 10.1097/cco.0000000000000602] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE OF REVIEW The bone marrow is home to hematopoietic stem cells responsible for lifelong blood production, alongside mesenchymal stem cells required for skeletal regeneration. In the bone marrow, a unique combination of signals derived from a multitude of cell types results in the establishment of so-called niches that regulate stem-cell maintenance and differentiation. Recently, single-cell and spatially resolved transcriptomics technologies have been utilized to characterize the murine bone marrow microenvironment during homeostasis, stress and upon cancer-induced remodeling. In this review, we summarize the major findings of these studies. RECENT FINDINGS Single-cell technologies applied to bone marrow provided the first systematic and label-free identification of bone marrow cell types, enabled their molecular and spatial characterization, and clarified the cellular sources of key prohematopoietic factors. Large transcriptional heterogeneity and novel subpopulations were observed in compartments previously thought to be homogenous. For example, Lepr Cxcl12-abundant reticular cells were shown to constitute the major source of prohematopoietic factors, but consist of subpopulations differing in their adipogenic versus osteogenic priming, morphology and localization. These subpopulations were suggested to act as professional cytokine secreting cells, thereby establishing distinct bone marrow niches. SUMMARY Single-cell and spatially resolved transcriptomics approaches have clarified the molecular identity and localization of bone marrow-resident cell types, paving the road for a deeper exploration of bone marrow niches in the mouse and humans.
Collapse
|
84
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) reside in specialized anatomical microenvironments within the bone marrow space, termed HSC niches. Different bone marrow imaging modalities have been utilized to visualize HSCs in situ, and unravel the cellular identity of bone marrow cell types located in their immediate proximity. However, despite extensive research, the exact identity of bone marrow populations that physically associate with HSCs remains controversial. RECENT FINDINGS Recent advances in volumetric imaging enable precise identification of bone marrow populations and their spatial distribution both at tissue-wide scale and single-cell resolution. In addition, single-cell RNA sequencing and mass-cytometry-based approaches dissect the complexity of the bone marrow microenvironment with unprecedented resolution. Here, we review current concepts regarding bone marrow populations that physically associate with HSCs and recent efforts to localize HSCs and their niche populations. SUMMARY Defining the bone marrow cell types in the immediate proximity of HSCs in homeostasis and stress is key to determine the cellular and molecular cues driving HSC maintenance and regeneration.
Collapse
|
85
|
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: 26] [Impact Index Per Article: 5.2] [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
|
86
|
Upadhaya S, Krichevsky O, Akhmetzyanova I, Sawai CM, Fooksman DR, Reizis B. Intravital Imaging Reveals Motility of Adult Hematopoietic Stem Cells in the Bone Marrow Niche. Cell Stem Cell 2020; 27:336-345.e4. [PMID: 32589864 DOI: 10.1016/j.stem.2020.06.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 04/28/2020] [Accepted: 06/02/2020] [Indexed: 01/05/2023]
Abstract
Adult mammalian hematopoietic stem cells (HSCs) reside in the bone marrow (BM) but can be mobilized into blood for use in transplantation. HSCs interact with BM niche cells that produce growth factor c-Kit ligand (Kitl/SCF) and chemokine CXCL12, and were thought to be static and sessile. We used two-photon laser scanning microscopy to visualize genetically labeled HSCs in the BM of live mice for several hours. The majority of HSCs showed a dynamic non-spherical morphology and significant motility, undergoing slow processive motion interrupted by short stretches of confined motion. HSCs moved in the perivascular space and showed intermittent close contacts with SCF-expressing perivascular stromal cells. In contrast, mobilization-inducing blockade of CXCL12 receptor CXCR4 and integrins rapidly abrogated HSC motility and shape dynamics in real time. Our results reveal an unexpectedly dynamic nature of HSC residence in the BM and interaction with the SCF+ stromal niche, which is disrupted during HSC mobilization.
Collapse
Affiliation(s)
- Samik Upadhaya
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Oleg Krichevsky
- Physics Department, Ben Gurion University of the Negev, Beer-Sheva 84105, Israel
| | | | - Catherine M Sawai
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA; INSERM Unit 1218 ACTION Laboratory, University of Bordeaux, Bergonié Cancer Institute, 33076 Bordeaux, France
| | - David R Fooksman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
87
|
Deynoux M, Sunter N, Ducrocq E, Dakik H, Guibon R, Burlaud-Gaillard J, Brisson L, Rouleux-Bonnin F, le Nail LR, Hérault O, Domenech J, Roingeard P, Fromont G, Mazurier F. A comparative study of the capacity of mesenchymal stromal cell lines to form spheroids. PLoS One 2020; 15:e0225485. [PMID: 32484831 PMCID: PMC7266346 DOI: 10.1371/journal.pone.0225485] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 05/17/2020] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSC)-spheroid models favor maintenance of stemness, ex vivo expansion and transplantation efficacy. Spheroids may also be considered as useful surrogate models of the hematopoietic niche. However, accessibility to primary cells, from bone marrow (BM) or adipose tissues, may limit their experimental use and the lack of consistency in methods to form spheroids may affect data interpretation. In this study, we aimed to create a simple model by examining the ability of cell lines, from human (HS-27a and HS-5) and murine (MS-5) BM origins, to form spheroids, compared to primary human MSCs (hMSCs). Our protocol efficiently allowed the spheroid formation from all cell types within 24 hours. Whilst hMSC-spheroids began to shrink after 24 hours, the size of spheroids from cell lines remained constant during three weeks. The difference was partially explained by the balance between proliferation and cell death, which could be triggered by hypoxia and induced oxidative stress. Our results demonstrate that, like hMSCs, MSC cell lines make reproductible spheroids that are easily handled. Thus, this model could help in understanding mechanisms involved in MSC functions and may provide a simple model by which to study cell interactions in the BM niche.
Collapse
Affiliation(s)
- Margaux Deynoux
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Nicola Sunter
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Elfi Ducrocq
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Hassan Dakik
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
| | - Roseline Guibon
- Anatomie et cytologie pathologique, CHRU de Tours, Tours, France
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Julien Burlaud-Gaillard
- Plateforme IBiSA de Microscopie Electronique, Université et CHRU de Tours, Tours, France
- INSERM U1259 MAVIVH, Université et CHRU de Tours, Tours, France
| | - Lucie Brisson
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | | | | | - Olivier Hérault
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- Service d'hématologie biologique, CHRU de Tours, Tours, France
| | - Jorge Domenech
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- Service d'hématologie biologique, CHRU de Tours, Tours, France
| | - Philippe Roingeard
- Plateforme IBiSA de Microscopie Electronique, Université et CHRU de Tours, Tours, France
- INSERM U1259 MAVIVH, Université et CHRU de Tours, Tours, France
| | - Gaëlle Fromont
- Anatomie et cytologie pathologique, CHRU de Tours, Tours, France
- INSERM UMR1069, Nutrition, Croissance et Cancer, Université de Tours, Tours, France
| | - Frédéric Mazurier
- EA 7501 GICC, CNRS ERL 7001 LNOx, Université de Tours, Tours, France
- * E-mail:
| |
Collapse
|
88
|
Ghazi S, Bourgeois S, Gomariz A, Bugarski M, Haenni D, Martins JR, Nombela-Arrieta C, Unwin RJ, Wagner CA, Hall AM, Craigie E. Multiparametric imaging reveals that mitochondria-rich intercalated cells in the kidney collecting duct have a very high glycolytic capacity. FASEB J 2020; 34:8510-8525. [PMID: 32367531 DOI: 10.1096/fj.202000273r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 03/30/2020] [Accepted: 04/16/2020] [Indexed: 02/06/2023]
Abstract
Alpha intercalated cells (αICs) in the kidney collecting duct (CD) belong to a family of mitochondria rich cells (MRCs) and have a crucial role in acidifying the urine via apical V-ATPase pumps. The nature of metabolism in αICs and its relationship to transport was not well-understood. Here, using multiphoton live cell imaging in mouse kidney tissue, FIB-SEM, and other complementary techniques, we provide new insights into mitochondrial structure and function in αICs. We show that αIC mitochondria have a rounded structure and are not located in close proximity to V-ATPase containing vesicles. They display a bright NAD(P)H fluorescence signal and low uptake of voltage-dependent dyes, but are energized by a pH gradient. However, expression of complex V (ATP synthase) is relatively low in αICs, even when stimulated by metabolic acidosis. In contrast, anaerobic glycolytic capacity is surprisingly high, and sufficient to maintain intracellular calcium homeostasis in the presence of complete aerobic inhibition. Moreover, glycolysis is essential for V-ATPase-mediated proton pumping. Key findings were replicated in narrow/clear cells in the epididymis, also part of the MRC family. In summary, using a range of cutting-edge techniques to investigate αIC metabolism in situ, we have discovered that these mitochondria dense cells have a high glycolytic capacity.
Collapse
Affiliation(s)
- Susan Ghazi
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Soline Bourgeois
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Alvaro Gomariz
- Department of Medical Oncology and Hematology, University of Zurich, Zurich, Switzerland.,Computer Vision Laboratory, ETH Zurich, Zurich, Switzerland
| | - Milica Bugarski
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| | - Dominik Haenni
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - Joana R Martins
- Center for Microscopy and Image Analysis, University of Zurich, Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University of Zurich, Zurich, Switzerland
| | - Robert J Unwin
- Department of Renal Medicine, University College London, UK.,AstraZeneca Biopharmaceuticals R&D, Gothenburg, Sweden
| | - Carsten A Wagner
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | - Andrew M Hall
- Institute of Anatomy, University of Zurich, Zurich, Switzerland.,Department of Nephrology, University Hospital Zurich, Switzerland
| | - Eilidh Craigie
- Institute of Anatomy, University of Zurich, Zurich, Switzerland
| |
Collapse
|
89
|
Al-Sharea A, Lee MKS, Purton LE, Hawkins ED, Murphy AJ. The haematopoietic stem cell niche: a new player in cardiovascular disease? Cardiovasc Res 2020; 115:277-291. [PMID: 30590405 DOI: 10.1093/cvr/cvy308] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
Haematopoiesis, the process of blood production, can be altered during the initiation or progression of many diseases. Cardiovascular disease (CVD) has been shown to be heavily influenced by changes to the haematopoietic system, including the types and abundance of immune cells produced. It is now well established that innate immune cells are increased in people with CVD, and the mechanisms contributing to this can be vastly different depending on the risk factors or comorbidities present. Many of these changes begin at the level of the haematopoietic stem and progenitor cells (HSPCs) that reside in the bone marrow (BM). In general, the HSPCs and downstream myeloid progenitors are expanded via increased proliferation in the setting of atherosclerotic CVD. However, HSPCs can also be encouraged to leave the BM and colonise extramedullary sites (i.e. the spleen). Within the BM, HSPCs reside in specialized microenvironments, often referred to as a niche. To date in depth studies assessing the damage or dysregulation that occurs in the BM niche in varying CVDs are scarce. In this review, we provide a general overview of the complex components and interactions within the BM niche and how they influence the function of HSPCs. Additionally, we discuss the main findings regarding changes in the HSPC niche that influence the progression of CVD. We hypothesize that understanding the influence of the BM niche in CVD will aid in delineating new pathways for therapeutic interventions.
Collapse
Affiliation(s)
- Annas Al-Sharea
- Division of Immunometabolism, Haematopoiesis and Leukocyte Biology, Baker Heart & Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.,Department of Immunology, Monash University, Melbourne, Australia
| | - Man Kit Sam Lee
- Division of Immunometabolism, Haematopoiesis and Leukocyte Biology, Baker Heart & Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.,Department of Immunology, Monash University, Melbourne, Australia
| | | | - Edwin D Hawkins
- Division of Immunology, Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | - Andrew J Murphy
- Division of Immunometabolism, Haematopoiesis and Leukocyte Biology, Baker Heart & Diabetes Institute, 75 Commercial Road, Melbourne, VIC, Australia.,Department of Immunology, Monash University, Melbourne, Australia
| |
Collapse
|
90
|
Dupard SJ, Grigoryan A, Farhat S, Coutu DL, Bourgine PE. Development of Humanized Ossicles: Bridging the Hematopoietic Gap. Trends Mol Med 2020; 26:552-569. [PMID: 32470383 DOI: 10.1016/j.molmed.2020.01.016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Revised: 01/16/2020] [Accepted: 01/31/2020] [Indexed: 02/06/2023]
Abstract
Ectopic 'humanized ossicles' (hOss) are miniaturized, engineered human bone organs in mice displaying a similar structure and function to native mouse bones. However, they are composed of human mesenchymal derived cells forming a humanized bone marrow niche. This in vivo reconstitution of human skeletal and hematopoietic compartments provides an opportunity to investigate the cellular and molecular processes involved in their establishment and functions in a human setting. However, current hOs strategies vary in their engineering methods and their downstream applications, undermining comprehensive exploitation of their potential. This review describes the specificities of the hOs models and highlights their potential and limits. Ultimately, we propose directions for the development of hOss as a technological platform for human hematopoietic studies.
Collapse
Affiliation(s)
- Steven J Dupard
- Laboratory for Cell, Tissue, and Organ engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden
| | - Ani Grigoryan
- Laboratory for Cell, Tissue, and Organ engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden
| | - Stephanie Farhat
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Daniel L Coutu
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, ON, Canada; Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada; Division of Orthopedic Surgery, The Ottawa Hospital, Ottawa, ON, Canada
| | - Paul E Bourgine
- Laboratory for Cell, Tissue, and Organ engineering, Department of Clinical Sciences, Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Wallenberg Center for Molecular Medicine, Lund University, Lund, Sweden; Stem Cell Center, Lund University, Lund, Sweden.
| |
Collapse
|
91
|
Altamirano DE, Noller K, Mihaly E, Grayson WL. Recent advances toward understanding the role of transplanted stem cells in tissue-engineered regeneration of musculoskeletal tissues. F1000Res 2020; 9:F1000 Faculty Rev-118. [PMID: 32117568 PMCID: PMC7029752 DOI: 10.12688/f1000research.21333.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/10/2020] [Indexed: 01/16/2023] Open
Abstract
Stem cell-based tissue engineering is poised to revolutionize the treatment of musculoskeletal injuries. However, in order to overcome scientific, practical, and regulatory obstacles and optimize therapeutic strategies, it is essential to better understand the mechanisms underlying the pro-regenerative effects of stem cells. There has been an attempted paradigm shift within the last decade to think of transplanted stem cells as "medicinal" therapies that orchestrate healing on the basis of their secretome and immunomodulatory profiles rather than acting as bona fide stem cells that proliferate, differentiate, and directly produce matrix to form de novo tissues. Yet the majority of current bone and skeletal muscle tissue engineering strategies are still premised on a direct contribution of stem cells as building blocks to tissue regeneration. Our review of the recent literature finds that researchers continue to focus on the quantification of de novo bone/skeletal muscle tissue following treatment and few studies aim to address this mechanistic conundrum directly. The dichotomy of thought is reflected in the diversity of new advances ranging from in situ three-dimensional bioprinting to a focus on exosomes and extracellular vesicles. However, recent findings elucidating the role of the immune system in tissue regeneration combined with novel imaging platform technologies will have a profound impact on our future understanding of how stem cells promote healing following biomaterial-mediated delivery to defect sites.
Collapse
Affiliation(s)
- Dallas E. Altamirano
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Kathleen Noller
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Duke University Medical School, Duke University, Durham, NC, 27710, USA
| | - Eszter Mihaly
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
| | - Warren L. Grayson
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Translational Tissue Engineering Center, Johns Hopkins University School of Medicine, Baltimore, MD, 21231, USA
- Department of Materials Science & Engineering, Johns Hopkins University School of Engineering, Baltimore, MD, 21231, USA
- Institute for NanoBioTechnology, Johns Hopkins University School of Engineering, Baltimore, MD, 21231, USA
| |
Collapse
|
92
|
Witkowski MT, Kousteni S, Aifantis I. Mapping and targeting of the leukemic microenvironment. J Exp Med 2020; 217:e20190589. [PMID: 31873722 PMCID: PMC7041707 DOI: 10.1084/jem.20190589] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 10/04/2019] [Accepted: 11/13/2019] [Indexed: 12/13/2022] Open
Abstract
Numerous studies support a role of the microenvironment in maintenance of the leukemic clone, as well as in treatment resistance. It is clear that disruption of the normal bone marrow microenvironment is sufficient to promote leukemic transformation and survival in both a cell autonomous and non-cell autonomous manner. In this review, we provide a snapshot of the various cell types shown to contribute to the leukemic microenvironment as well as treatment resistance. Several of these studies suggest that leukemic blasts occupy specific cellular and biochemical "niches." Effective dissection of critical leukemic niche components using single-cell approaches has allowed a more precise and extensive characterization of complexity that underpins both the healthy and malignant bone marrow microenvironment. Knowledge gained from these observations can have an important impact in the development of microenvironment-directed targeted approaches aimed at mitigating disease relapse.
Collapse
Affiliation(s)
- Matthew T. Witkowski
- Department of Pathology, New York University School of Medicine, New York, NY
- Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY
| | - Stavroula Kousteni
- Department of Physiology & Cellular Biophysics, Columbia University Irving Medical Center, New York, NY
| | - Iannis Aifantis
- Department of Pathology, New York University School of Medicine, New York, NY
- Laura & Isaac Perlmutter Cancer Center, New York University School of Medicine, New York, NY
| |
Collapse
|
93
|
Bravenboer N, Bredella MA, Chauveau C, Corsi A, Douni E, Ferris WF, Riminucci M, Robey PG, Rojas-Sutterlin S, Rosen C, Schulz TJ, Cawthorn WP. Standardised Nomenclature, Abbreviations, and Units for the Study of Bone Marrow Adiposity: Report of the Nomenclature Working Group of the International Bone Marrow Adiposity Society. Front Endocrinol (Lausanne) 2020; 10:923. [PMID: 32038486 PMCID: PMC6993042 DOI: 10.3389/fendo.2019.00923] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Accepted: 12/18/2019] [Indexed: 12/16/2022] Open
Abstract
Research into bone marrow adiposity (BMA) has expanded greatly since the late 1990s, leading to development of new methods for the study of bone marrow adipocytes. Simultaneously, research fields interested in BMA have diversified substantially. This increasing interest is revealing fundamental new knowledge of BMA; however, it has also led to a highly variable nomenclature that makes it difficult to interpret and compare results from different studies. A consensus on BMA nomenclature has therefore become indispensable. This article addresses this critical need for standardised terminology and consistent reporting of parameters related to BMA research. The International Bone Marrow Adiposity Society (BMAS) was formed in 2017 to consolidate the growing scientific community interested in BMA. To address the BMA nomenclature challenge, BMAS members from diverse fields established a working group (WG). Based on their broad expertise, the WG first reviewed the existing, unsystematic nomenclature and identified terms, and concepts requiring further discussion. They thereby identified and defined 8 broad concepts and methods central to BMA research. Notably, these had been described using 519 unique combinations of term, abbreviation and unit, many of which were overlapping or redundant. On this foundation a second consensus was reached, with each term classified as "to use" or "not to use." As a result, the WG reached a consensus to craft recommendations for 26 terms related to concepts and methods in BMA research. This was approved by the Scientific Board and Executive Board of BMAS and is the basis for the present recommendations for a formal BMA nomenclature. As an example, several terms or abbreviations have been used to represent "bone marrow adipocytes," including BMAds, BM-As, and BMAs. The WG decided that BMA should refer to "bone marrow adiposity"; that BM-A is too similar to BMA; and noted that "Ad" has previously been recommended to refer to adipocytes. Thus, it was recommended to use BMAds to represent bone marrow adipocytes. In conclusion, the standard nomenclature proposed in this article should be followed for all communications of results related to BMA. This will allow for better interactions both inside and outside of this emerging scientific community.
Collapse
Affiliation(s)
- Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam Movement Sciences, Amsterdam University Medical Center, Vrije Universiteit, Amsterdam, Netherlands
| | - Miriam A. Bredella
- Department of Radiology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
| | - Christophe Chauveau
- Univ. Littoral Côte d'Opale, Boulogne-sur-Mer, France
- Univ. Lille, Lille, France
- CHU Lille, Lille, France
- Physiopathologie des Maladies Osseuses Inflammatoires, Boulogne-sur-Mer, France
| | - Alessandro Corsi
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Eleni Douni
- Biological Sciences Research Center “Alexander Fleming”, Athens, Greece
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
| | - William F. Ferris
- Division of Endocrinology, Department of Medicine, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Mara Riminucci
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | - Pamela G. Robey
- Skeletal Biology Section, NIDCR, NIH, DHHS, Bethesda, MD, United States
| | - Shanti Rojas-Sutterlin
- Institute for Research in Immunology and Cancer, Université de Montréal, Montreal, QC, Canada
| | - Clifford Rosen
- Maine Medical Research Center Institute, Scarborough, ME, United States
| | - Tim J. Schulz
- German Institute of Human Nutrition Potsdam-Rehbrücke, Nuthetal, Germany
- German Center for Diabetes Research (DZD), München, Germany
| | - William P. Cawthorn
- BHF Centre for Cardiovascular Science, The Queen's Medical Research Institute, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
94
|
Zhang H, Yarinome K, Kawakami R, Otomo K, Nemoto T, Okamura Y. Nanosheet wrapping-assisted coverslip-free imaging for looking deeper into a tissue at high resolution. PLoS One 2020; 15:e0227650. [PMID: 31923215 PMCID: PMC6953877 DOI: 10.1371/journal.pone.0227650] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/22/2019] [Indexed: 12/19/2022] Open
Abstract
In order to achieve deep tissue imaging, a number of optical clearing agents have been developed. However, in a conventional microscopy setup, an objective lens can only be moved until it is in contact with a coverslip, which restricts the maximum focusing depth into a cleared tissue specimen. Until now, it is still a fact that the working distance of a high magnification objective lens with a high numerical aperture is always about 100 μm. In this study, a polymer thin film (also called as nanosheet) composed of fluoropolymer with a thickness of 130 nm, less than one-thousandth that of a 170 μm thick coverslip, is employed to replace the coverslip. Owing to its excellent characteristics, such as high optical transparency, mechanical robustness, chemical resistance, and water retention ability, nanosheet is uniquely capable of providing a coverslip-free imaging. By wrapping the tissue specimen with a nanosheet, an extra distance of 170 μm for the movement of objective lens is obtained. Results show an equivalently high resolution imaging can be obtained if a homogenous refractive index between immersion liquid and mounting media is adjusted. This method will facilitate a variety of imaging tasks with off-the-shelf high magnification objectives.
Collapse
Affiliation(s)
- Hong Zhang
- Department of Applied Chemistry, School of Engineering, Tokai University, Kanagawa, Japan
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan
| | - Kenji Yarinome
- Course of Applied Science, Graduate School of Engineering, Tokai University, Kanagawa, Japan
| | - Ryosuke Kawakami
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
- Department of Molecular Medicine for Pathogenesis, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Kohei Otomo
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
- Exploratory Research Center on Life and Living Systems, National Institute of Natural Sciences, Aichi, Japan
- National Institute for Physiological Sciences, Aichi, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Aichi, Japan
| | - Tomomi Nemoto
- Research Institute for Electronic Science, Hokkaido University, Sapporo, Japan
- Graduate School of Information Science and Technology, Hokkaido University, Sapporo, Japan
- Exploratory Research Center on Life and Living Systems, National Institute of Natural Sciences, Aichi, Japan
- National Institute for Physiological Sciences, Aichi, Japan
- The Graduate University for Advanced Studies (SOKENDAI), Aichi, Japan
| | - Yosuke Okamura
- Department of Applied Chemistry, School of Engineering, Tokai University, Kanagawa, Japan
- Micro/Nano Technology Center, Tokai University, Kanagawa, Japan
- Course of Applied Science, Graduate School of Engineering, Tokai University, Kanagawa, Japan
| |
Collapse
|
95
|
Tratwal J, Labella R, Bravenboer N, Kerckhofs G, Douni E, Scheller EL, Badr S, Karampinos DC, Beck-Cormier S, Palmisano B, Poloni A, Moreno-Aliaga MJ, Fretz J, Rodeheffer MS, Boroumand P, Rosen CJ, Horowitz MC, van der Eerden BCJ, Veldhuis-Vlug AG, Naveiras O. Reporting Guidelines, Review of Methodological Standards, and Challenges Toward Harmonization in Bone Marrow Adiposity Research. Report of the Methodologies Working Group of the International Bone Marrow Adiposity Society. Front Endocrinol (Lausanne) 2020; 11:65. [PMID: 32180758 PMCID: PMC7059536 DOI: 10.3389/fendo.2020.00065] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 01/31/2020] [Indexed: 12/14/2022] Open
Abstract
The interest in bone marrow adiposity (BMA) has increased over the last decade due to its association with, and potential role, in a range of diseases (osteoporosis, diabetes, anorexia, cancer) as well as treatments (corticosteroid, radiation, chemotherapy, thiazolidinediones). However, to advance the field of BMA research, standardization of methods is desirable to increase comparability of study outcomes and foster collaboration. Therefore, at the 2017 annual BMA meeting, the International Bone Marrow Adiposity Society (BMAS) founded a working group to evaluate methodologies in BMA research. All BMAS members could volunteer to participate. The working group members, who are all active preclinical or clinical BMA researchers, searched the literature for articles investigating BMA and discussed the results during personal and telephone conferences. According to the consensus opinion, both based on the review of the literature and on expert opinion, we describe existing methodologies and discuss the challenges and future directions for (1) histomorphometry of bone marrow adipocytes, (2) ex vivo BMA imaging, (3) in vivo BMA imaging, (4) cell isolation, culture, differentiation and in vitro modulation of primary bone marrow adipocytes and bone marrow stromal cell precursors, (5) lineage tracing and in vivo BMA modulation, and (6) BMA biobanking. We identify as accepted standards in BMA research: manual histomorphometry and osmium tetroxide 3D contrast-enhanced μCT for ex vivo quantification, specific MRI sequences (WFI and H-MRS) for in vivo studies, and RT-qPCR with a minimal four gene panel or lipid-based assays for in vitro quantification of bone marrow adipogenesis. Emerging techniques are described which may soon come to complement or substitute these gold standards. Known confounding factors and minimal reporting standards are presented, and their use is encouraged to facilitate comparison across studies. In conclusion, specific BMA methodologies have been developed. However, important challenges remain. In particular, we advocate for the harmonization of methodologies, the precise reporting of known confounding factors, and the identification of methods to modulate BMA independently from other tissues. Wider use of existing animal models with impaired BMA production (e.g., Pfrt-/-, KitW/W-v) and development of specific BMA deletion models would be highly desirable for this purpose.
Collapse
Affiliation(s)
- Josefine Tratwal
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Rossella Labella
- Tissue and Tumour Microenvironments Lab, The Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam University Medical Centers, Vrije Universiteit, Amsterdam Movement Sciences, Amsterdam, Netherlands
- Section of Endocrinology, Department of Internal Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
| | - Greet Kerckhofs
- Biomechanics Lab, Institute of Mechanics, Materials and Civil Engineering, UCLouvain, Louvain-la-Neuve, Belgium
- Department Materials Engineering, KU Leuven, Leuven, Belgium
| | - Eleni Douni
- Laboratory of Genetics, Department of Biotechnology, Agricultural University of Athens, Athens, Greece
- Institute for Bioinnovation, Biomedical Sciences Research Center Alexander Fleming, Athens, Greece
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, MO, United States
| | - Sammy Badr
- Univ. Lille, EA 4490 - PMOI - Physiopathologie des Maladies Osseuses Inflammatoires, Lille, France
- CHU Lille, Service de Radiologie et Imagerie Musculosquelettique, Lille, France
| | - Dimitrios C. Karampinos
- Department of Diagnostic and Interventional Radiology, Technical University of Munich, Munich, Germany
| | - Sarah Beck-Cormier
- Inserm, UMR 1229, RMeS, Regenerative Medicine and Skeleton, Université de Nantes, ONIRIS, Nantes, France
- Université de Nantes, UFR Odontologie, Nantes, France
| | - Biagio Palmisano
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY, United States
| | - Antonella Poloni
- Hematology, Department of Clinic and Molecular Science, Università Politecnica Marche-AOU Ospedali Riuniti, Ancona, Italy
| | - Maria J. Moreno-Aliaga
- Centre for Nutrition Research and Department of Nutrition, Food Science and Physiology, School of Pharmacy and Nutrition, University of Navarra, Pamplona, Spain
- IdiSNA, Navarra's Health Research Institute, Pamplona, Spain
- CIBERobn Physiopathology of Obesity and Nutrition, Centre of Biomedical Research Network, ISCIII, Madrid, Spain
| | - Jackie Fretz
- Department of Orthopaedics and Rehabilitation, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, CT, United States
| | - Matthew S. Rodeheffer
- Department of Comparative Medicine and Molecular, Cellular and Developmental Biology, Yale University School of Medicine, New Haven, CT, United States
| | - Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Clifford J. Rosen
- Maine Medical Center Research Institute, Center for Clinical and Translational Research, Scarborough, ME, United States
| | - Mark C. Horowitz
- Department of Orthopaedics and Rehabilitation, Yale University School of Medicine, New Haven, CT, United States
| | - Bram C. J. van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Annegreet G. Veldhuis-Vlug
- Section of Endocrinology, Department of Internal Medicine, Center for Bone Quality, Leiden University Medical Center, Leiden, Netherlands
- Maine Medical Center Research Institute, Center for Clinical and Translational Research, Scarborough, ME, United States
- Jan van Goyen Medical Center/OLVG Hospital, Department of Internal Medicine, Amsterdam, Netherlands
- *Correspondence: Annegreet G. Veldhuis-Vlug
| | - Olaia Naveiras
- Laboratory of Regenerative Hematopoiesis, Institute of Bioengineering and Swiss Institute for Experimental Cancer Research, Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
- Hematology Service, Departments of Oncology and Laboratory Medicine, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
- Olaia Naveiras ;
| |
Collapse
|
96
|
Baccin C, Al-Sabah J, Velten L, Helbling PM, Grünschläger F, Hernández-Malmierca P, Nombela-Arrieta C, Steinmetz LM, Trumpp A, Haas S. Combined single-cell and spatial transcriptomics reveal the molecular, cellular and spatial bone marrow niche organization. Nat Cell Biol 2020; 22:38-48. [PMID: 31871321 PMCID: PMC7610809 DOI: 10.1038/s41556-019-0439-6] [Citation(s) in RCA: 548] [Impact Index Per Article: 109.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Accepted: 11/20/2019] [Indexed: 02/07/2023]
Abstract
The bone marrow constitutes the primary site for life-long blood production and skeletal regeneration. However, its cellular and spatial organization remains controversial. Here, we combine single-cell and spatially resolved transcriptomics to systematically map the molecular, cellular and spatial composition of distinct bone marrow niches. This allowed us to transcriptionally profile all major bone-marrow-resident cell types, determine their localization and clarify sources of pro-haematopoietic factors. Our data demonstrate that Cxcl12-abundant-reticular (CAR) cell subsets (Adipo-CAR and Osteo-CAR) differentially localize to sinusoidal and arteriolar surfaces, act locally as 'professional cytokine-secreting cells' and thereby establish peri-vascular micro-niches. Importantly, the three-dimensional bone-marrow organization can be accurately inferred from single-cell transcriptome data using the RNA-Magnet algorithm described here. Together, our study reveals the cellular and spatial organization of bone marrow niches and offers a systematic approach to dissect the complex organization of whole organs.
Collapse
Affiliation(s)
- Chiara Baccin
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
- Faculty of Biosciences, Joint PhD degree from EMBL and Heidelberg University, Heidelberg, Germany
| | - Jude Al-Sabah
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Lars Velten
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Center for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain.
| | - Patrick M Helbling
- Department of Medical Oncology and Hematology, University Hospital and University of Zürich, Zürich, Switzerland
| | - Florian Grünschläger
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Pablo Hernández-Malmierca
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University Hospital and University of Zürich, Zürich, Switzerland
| | - Lars M Steinmetz
- Genome Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany.
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Genome Technology Center, Palo Alto, CA, USA.
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
- German Cancer Consortium (DKTK), Heidelberg, Germany.
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany.
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany.
| |
Collapse
|
97
|
Kunz L, Schroeder T. A 3D Tissue-wide Digital Imaging Pipeline for Quantitation of Secreted Molecules Shows Absence of CXCL12 Gradients in Bone Marrow. Cell Stem Cell 2019; 25:846-854.e4. [DOI: 10.1016/j.stem.2019.10.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 07/15/2019] [Accepted: 10/17/2019] [Indexed: 12/13/2022]
|
98
|
Bourgine PE, Fritsch K, Pigeot S, Takizawa H, Kunz L, Kokkaliaris KD, Coutu DL, Manz MG, Martin I, Schroeder T. Fate Distribution and Regulatory Role of Human Mesenchymal Stromal Cells in Engineered Hematopoietic Bone Organs. iScience 2019; 19:504-513. [PMID: 31442666 PMCID: PMC6710718 DOI: 10.1016/j.isci.2019.08.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 07/01/2019] [Accepted: 08/02/2019] [Indexed: 12/21/2022] Open
Abstract
The generation of humanized ectopic ossicles (hOss) in mice has been proposed as an advanced translational and fundamental model to study the human hematopoietic system. The approach relies on the presence of human bone marrow-derived mesenchymal stromal cells (hMSCs) supporting the engraftment of transplanted human hematopoietic stem and progenitor cells (HSPCs). However, the functional distribution of hMSCs within the humanized microenvironment remains to be investigated. Here, we combined genetic tools and quantitative confocal microscopy to engineer and subsequently analyze hMSCs′ fate and distribution in hOss. Implanted hMSCs reconstituted a humanized environment including osteocytes, osteoblasts, adipocytes, and stromal cells associated with vessels. By imaging full hOss, we identified rare physical interactions between hMSCs and human CD45+/CD34+/CD90+ cells, supporting a functional contact-triggered regulatory role of hMSCs. Our study highlights the importance of compiling quantitative information from humanized organs, to decode the interactions between the hematopoietic and the stromal compartments. Mesenchymal cells can generate human bone organs with tailored molecular signature Mesenchymal cells reconstitute a human niche environment capable of regulating HSPCs
Collapse
Affiliation(s)
- Paul E Bourgine
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland; Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4056 Basel, Switzerland; Department of Clinical Sciences, Lund Stem Cell Center, Lund University, BMC B11, 221 84 Lund, Sweden; Wallenberg Centre for Molecular Medicine, Lund University, Lund, Sweden
| | - Kristin Fritsch
- Department of Hematology, University Hospital Zurich and University of Zurich, 8091 Zurich, Switzerland
| | - Sebastien Pigeot
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4056 Basel, Switzerland
| | - Hitoshi Takizawa
- International Research Center for Medical Sciences, Kumamoto University, Kumamoto 860-0811, Japan
| | - Leo Kunz
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Konstantinos D Kokkaliaris
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Daniel L Coutu
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Markus G Manz
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4056 Basel, Switzerland.
| | - Ivan Martin
- Tissue Engineering, Department of Biomedicine, University of Basel and University Hospital Basel, 4056 Basel, Switzerland.
| | - Timm Schroeder
- Department of Biosystems Science and Engineering (D-BSSE), ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland.
| |
Collapse
|
99
|
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: 2.3] [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
|
100
|
Okuwobi IP, Ji Z, Fan W, Yuan S, Bekalo L, Chen Q. Automated Quantification of Hyperreflective Foci in SD-OCT With Diabetic Retinopathy. IEEE J Biomed Health Inform 2019; 24:1125-1136. [PMID: 31329137 DOI: 10.1109/jbhi.2019.2929842] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The presence of hyperreflective foci (HFs) is related to retinal disease progression, and the quantity has proven to be a prognostic factor of visual and anatomical outcome in various retinal diseases. However, lack of efficient quantitative tools for evaluating the HFs has deprived ophthalmologist of assessing the volume of HFs. For this reason, we propose an automated quantification algorithm to segment and quantify HFs in spectral domain optical coherence tomography (SD-OCT). The proposed algorithm consists of two parallel processes namely: region of interest (ROI) generation and HFs estimation. To generate the ROI, we use morphological reconstruction to obtain the reconstructed image and histogram constructed for data distributions and clustering. In parallel, we estimate the HFs by extracting the extremal regions from the connected regions obtained from a component tree. Finally, both the ROI and the HFs estimation process are merged to obtain the segmented HFs. The proposed algorithm was tested on 40 3D SD-OCT volumes from 40 patients diagnosed with non-proliferative diabetic retinopathy (NPDR), proliferative diabetic retinopathy (PDR), and diabetic macular edema (DME). The average dice similarity coefficient (DSC) and correlation coefficient (r) are 69.70%, 0.99 for NPDR, 70.31%, 0.99 for PDR, and 71.30%, 0.99 for DME, respectively. The proposed algorithm can provide ophthalmologist with good HFs quantitative information, such as volume, size, and location of the HFs.
Collapse
|