1
|
Boueya IL, Sandhow L, Albuquerque JRP, Znaidi R, Passaro D. Endothelial heterogeneity in bone marrow: insights across development, adult life and leukemia. Leukemia 2025; 39:8-24. [PMID: 39528790 PMCID: PMC11717709 DOI: 10.1038/s41375-024-02453-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 10/04/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
The central role of the endothelial microenvironment in orchestrating bone marrow (BM) homeostasis and hematopoietic support has been confirmed at various developmental stages and in adult life. The BM vasculature is crucial in mediating communication between BM parenchyma and circulating blood, displaying remarkable heterogeneity in structure and function. While vascular cell diversity in other tissues has long been recognized, the molecular basis of this phenomenon in BM is just now emerging. Over the past decade, single-cell approaches and microscopic observations have expanded our understanding of BM vasculature. While solely characterized for their paracrine properties in the past, recent advances have revolutionized our perception of endothelial function, revealing distinct anatomical locations associated with diverse endothelial cell states. The identification of phenotypic differences between normal and pathological conditions has therefore deepened our understanding of vascular dynamics and their impact on hematopoiesis in health and disease. In this review, we highlight key milestones and recent advances in understanding vascular heterogeneity within BM microenvironment during development, adulthood and aging. We also explore how leukemia affects this heterogeneity and how we can take this knowledge forward to improve clinical practices. By synthesizing existing literature, we aim to address unresolved questions and outline future research directions.
Collapse
Affiliation(s)
- I L Boueya
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - L Sandhow
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - J R P Albuquerque
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - R Znaidi
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France
| | - D Passaro
- Leukemia and Niche Dynamics laboratory, Institut Cochin, Université Paris Cité UMR-S1016, INSERM U1016, CNRS UMR8104, Paris, France.
| |
Collapse
|
2
|
Koh BI, Mohanakrishnan V, Jeong HW, Park H, Kruse K, Choi YJ, Nieminen-Kelhä M, Kumar R, Pereira RS, Adams S, Lee HJ, Bixel MG, Vajkoczy P, Krause DS, Adams RH. Adult skull bone marrow is an expanding and resilient haematopoietic reservoir. Nature 2024; 636:172-181. [PMID: 39537918 PMCID: PMC11618084 DOI: 10.1038/s41586-024-08163-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 10/07/2024] [Indexed: 11/16/2024]
Abstract
The bone marrow microenvironment is a critical regulator of haematopoietic stem cell self-renewal and fate1. Although it is appreciated that ageing, chronic inflammation and other insults compromise bone marrow function and thereby negatively affect haematopoiesis2, it is not known whether different bone compartments exhibit distinct microenvironmental properties and functional resilience. Here we use imaging, pharmacological approaches and mouse genetics to uncover specialized properties of bone marrow in adult and ageing skull. Specifically, we show that the skull bone marrow undergoes lifelong expansion involving vascular growth, which results in an increasing contribution to total haematopoietic output. Furthermore, skull is largely protected against major hallmarks of ageing, including upregulation of pro-inflammatory cytokines, adipogenesis and loss of vascular integrity. Conspicuous rapid and dynamic changes to the skull vasculature and bone marrow are induced by physiological alterations, namely pregnancy, but also pathological challenges, such as stroke and experimental chronic myeloid leukaemia. These responses are highly distinct from femur, the most extensively studied bone marrow compartment. We propose that skull harbours a protected and dynamically expanding bone marrow microenvironment, which is relevant for experimental studies and, potentially, for clinical treatments in humans.
Collapse
Affiliation(s)
- Bong Ihn Koh
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
| | - Vishal Mohanakrishnan
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hyun-Woo Jeong
- Sequencing Core Facility, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hongryeol Park
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Kai Kruse
- Bioinformatics Service Unit, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Young Jun Choi
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Melina Nieminen-Kelhä
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Rahul Kumar
- Institute of Transfusion Medicine, Transfusion Center, University Medicine Mainz, Mainz, Germany
| | - Raquel S Pereira
- Georg-Speyer-Haus Institute for Tumor Biology and Experimental Medicine and Goethe University Frankfurt, Frankfurt, Germany
| | - Susanne Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Hyuek Jong Lee
- Center for Vascular Research, Institute for Basic Science, Daejeon, Republic of Korea
| | - M Gabriele Bixel
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Peter Vajkoczy
- Department of Neurosurgery, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Daniela S Krause
- Institute of Transfusion Medicine, Transfusion Center, University Medicine Mainz, Mainz, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
| |
Collapse
|
3
|
Pereira AL, Galli S, Nombela‐Arrieta C. Bone marrow niches for hematopoietic stem cells. Hemasphere 2024; 8:e133. [PMID: 39086665 PMCID: PMC11289431 DOI: 10.1002/hem3.133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/05/2024] [Accepted: 05/06/2024] [Indexed: 08/02/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are the cornerstone of the hematopoietic system. HSCs sustain the continuous generation of mature blood derivatives while self-renewing to preserve a relatively constant pool of progenitors throughout life. Yet, long-term maintenance of functional HSCs exclusively takes place in association with their native tissue microenvironment of the bone marrow (BM). HSCs have been long proposed to reside in fixed and identifiable anatomical units found in the complex BM tissue landscape, which control their identity and fate in a deterministic manner. In the last decades, tremendous progress has been made in the dissection of the cellular and molecular fabric of the BM, the structural organization governing tissue function, and the plethora of interactions established by HSCs. Nonetheless, a holistic model of the mechanisms controlling HSC regulation in their niche is lacking to date. Here, we provide an overview of our current understanding of BM anatomy, HSC localization, and crosstalk within local cellular neighborhoods in murine and human tissues, and highlight fundamental open questions on how HSCs functionally integrate in the BM microenvironment.
Collapse
Affiliation(s)
- Ana Luísa Pereira
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - Serena Galli
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| | - César Nombela‐Arrieta
- Department of Medical Oncology and HematologyUniversity Hospital and University of ZurichZurichSwitzerland
| |
Collapse
|
4
|
Grammer C, Komorowska JA, Swann JB. Vhl safeguards thymic epithelial cell identity and thymopoietic capacity by constraining Hif1a activity during development. iScience 2024; 27:110258. [PMID: 39040069 PMCID: PMC11261450 DOI: 10.1016/j.isci.2024.110258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 03/15/2024] [Accepted: 06/10/2024] [Indexed: 07/24/2024] Open
Abstract
The thymus is a physiologically hypoxic organ and fulfills its role of generating T cells under low-oxygen conditions. We have therefore investigated how thymic epithelial cells (TECs) cope with physiological hypoxia by focusing on the role of the Hif1a-Vhl axis. In most cell types, the oxygen-labile transcriptional regulator Hif1a is a central player in co-ordinating responses to low oxygen: under normoxic conditions Hif1a is rapidly degraded in a Vhl-guided manner; however, under hypoxic conditions Hif1a is stabilized and can execute its transcriptional functions. Unexpectedly, we find that, although TECs reside in a hypoxic microenvironment, they express little Hif1a protein and do not require Hif1a for their development or function. Instead, we find that Vhl function in TECs is vital to constrain Hif1a activity, as loss of Vhl results in dramatic defects in TEC differentiation and thymopoiesis, which can be rescued by Hif1a co-depletion.
Collapse
Affiliation(s)
- Christiane Grammer
- Department of Developmental Immunology, Max Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| | - Julia A. Komorowska
- Department of Developmental Immunology, Max Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
- Albert Ludwig University, Faculty of Biology, Freiburg, Germany
| | - Jeremy B. Swann
- Department of Developmental Immunology, Max Planck-Institute of Immunobiology and Epigenetics, Freiburg, Germany
| |
Collapse
|
5
|
Bhattacharyya ND, Kyaw W, McDonald MM, Dhenni R, Grootveld AK, Xiao Y, Chai R, Khoo WH, Danserau LC, Sergio CM, Timpson P, Lee WM, Croucher PI, Phan TG. Minimally invasive longitudinal intravital imaging of cellular dynamics in intact long bone. Nat Protoc 2023; 18:3856-3880. [PMID: 37857852 DOI: 10.1038/s41596-023-00894-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 07/28/2023] [Indexed: 10/21/2023]
Abstract
Intravital two-photon microscopy enables deep-tissue imaging at high temporospatial resolution in live animals. However, the endosteal bone compartment and underlying bone marrow pose unique challenges to optical imaging as light is absorbed, scattered and dispersed by thick mineralized bone matrix and the adipose-rich bone marrow. Early bone intravital imaging methods exploited gaps in the cranial sutures to bypass the need to penetrate through cortical bone. More recently, investigators have developed invasive methods to thin the cortical bone or implant imaging windows to image cellular dynamics in weight-bearing long bones. Here, we provide a step-by-step procedure for the preparation of animals for minimally invasive, nondestructive, longitudinal intravital imaging of the murine tibia. This method involves the use of mixed bone marrow radiation chimeras to unambiguously double-label osteoclasts and osteomorphs. The tibia is exposed by a simple skin incision and an imaging chamber constructed using thermoconductive T-putty. Imaging sessions up to 12 h long can be repeated over multiple timepoints to provide a longitudinal time window into the endosteal and marrow niches. The approach can be used to investigate cellular dynamics in bone remodeling, cancer cell life cycle and hematopoiesis, as well as long-lived humoral and cellular immunity. The procedure requires an hour to complete and is suitable for users with minimal prior expertise in small animal surgery.
Collapse
Affiliation(s)
- Nayan Deger Bhattacharyya
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Wunna Kyaw
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Michelle M McDonald
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Rama Dhenni
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Abigail K Grootveld
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ya Xiao
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Ryan Chai
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Weng Hua Khoo
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
| | - Linda C Danserau
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - C Marcelo Sergio
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Paul Timpson
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - Woei Ming Lee
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, New South Wales, Australia
| | - Peter I Croucher
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia
| | - Tri Giang Phan
- Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW Sydney, Sydney, New South Wales, Australia.
- ACRF INCITe Centre for Intravital Imaging of Niches for Cancer Immune Therapy, Sydney, New South Wales, Australia.
| |
Collapse
|
6
|
Canalis E, Mocarska M, Schilling L, Jafar-Nejad P, Carrer M. Antisense oligonucleotides targeting a NOTCH3 mutation in male mice ameliorate the cortical osteopenia of lateral meningocele syndrome. Bone 2023; 177:116898. [PMID: 37704069 PMCID: PMC10591917 DOI: 10.1016/j.bone.2023.116898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023]
Abstract
Lateral Meningocele Syndrome (LMS) is a monogenic disorder associated with NOTCH3 pathogenic variants that result in the stabilization of NOTCH3 and a gain-of-function. A mouse model (Notch3em1Ecan) harboring a 6691-TAATGA mutation in the Notch3 locus that results in a functional outcome analogous to LMS exhibits cancellous and cortical bone osteopenia. We tested Notch3 antisense oligonucleotides (ASOs) specific to the Notch36691-TAATGA mutation for their effects on Notch3 downregulation and on the osteopenia of Notch3em1Ecan mice. Twenty-four mouse Notch3 mutant ASOs were designed and tested for toxic effects in vivo, and 12 safe ASOs were tested for their impact on the downregulation of Notch36691-TAATGA and Notch3 mRNA in osteoblast cultures from Notch3em1Ecan mice. Three ASOs downregulated Notch3 mutant transcripts specifically and were tested in vivo for their effects on the bone microarchitecture of Notch3em1Ecan mice. All three ASOs were well tolerated. One of these ASOs had more consistent effects in vivo and was studied in detail. The Notch3 mutant ASO downregulated Notch3 mutant transcripts in osteoblasts and bone marrow stromal cells and had no effect on other Notch receptors. The subcutaneous administration of Notch3 mutant ASO at 50 mg/Kg decreased Notch36691-TAATGA mRNA in bone without apparent toxicity; microcomputed tomography demonstrated that the ASO ameliorated the cortical osteopenia of Notch3em1Ecan mice but not the cancellous bone osteopenia. In conclusion, a Notch3 ASO that downregulates Notch3 mutant expression specifically ameliorates the cortical osteopenia in Notch3em1Ecan mice. ASOs may become useful strategies in the management of monogenic disorders affecting the skeleton.
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, CT, USA; Department of Medicine, UConn Health, Farmington, CT, USA; UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA.
| | - Magda Mocarska
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | - Lauren Schilling
- UConn Musculoskeletal Institute, UConn Health, Farmington, CT, USA
| | | | | |
Collapse
|
7
|
Capp JP, Bataille R. The Ins and Outs of Endosteal Niche Disruption in the Bone Marrow: Relevance for Myeloma Oncogenesis. BIOLOGY 2023; 12:990. [PMID: 37508420 PMCID: PMC10376322 DOI: 10.3390/biology12070990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/30/2023]
Abstract
Multiple Myeloma (MM) and its preexisting stage, termed Monoclonal Gammopathy of Undetermined Significance (MGUS), have long been considered mainly as genomic diseases. However, the bone changes observed in both conditions have led to a reassessment of the role of the bone microenvironment, mainly the endosteal niche in their genesis. Here, we consider the disruption of the endosteal niche in the bone marrow, that is, the shift of the endosteal niche from an osteoblastic to an osteoclastic profile produced by bone senescence and inflammaging, as the key element. Thus, this disrupted endosteal niche is proposed to represent the permissive microenvironment necessary not only for the emergence of MM from MGUS but also for the emergence and maintenance of MGUS. Moreover, the excess of osteoclasts would favor the presentation of antigens (Ag) into the endosteal niche because osteoclasts are Ag-presenting cells. As such, they could significantly stimulate the presentation of some specific Ag and the clonal expansion of the stimulated cells as well as favor the expansion of such selected clones because osteoclasts are immunosuppressive. We also discuss this scenario in the Gaucher disease, in which the high incidence of MGUS and MM makes it a good model both at the bone level and the immunological level. Finally, we envisage that this endosteal niche disruption would increase the stochasticity (epigenetic and genetic instability) in the selected clones, according to our Tissue Disruption-induced cell Stochasticity (TiDiS) theory.
Collapse
Affiliation(s)
- Jean-Pascal Capp
- Toulouse Biotechnology Institute, INSA/University of Toulouse, CNRS, INRAE, 31077 Toulouse, France
| | - Régis Bataille
- School of Medicine, University of Angers, 49045 Angers, France
| |
Collapse
|
8
|
Abstract
The bone marrow (BM) is home to numerous cell types arising from hematopoietic stem cells (HSCs) and nonhematopoietic mesenchymal stem cells, as well as stromal cell components. Together they form the BM microenvironment or HSC niche. HSCs critically depend on signaling from these niches to function and survive in the long term. Significant advances in imaging technologies over the past decade have permitted the study of the BM microenvironment in mice, particularly with the development of intravital microscopy (IVM), which provides a powerful method to study these cells in vivo and in real time. Still, there is a lot to be learnt about the interactions of individual HSCs with their environment - at steady state and under various stresses - and whether specific niches exist for distinct developing hematopoietic lineages. Here, we describe our protocol and techniques used to visualize transplanted HSCs in the mouse calvarium, using combined confocal and two-photon IVM.
Collapse
Affiliation(s)
- Myriam L R Haltalli
- Imperial College London, London, UK
- The Francis Crick Institute, London, UK
- Wellcome - Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Cristina Lo Celso
- Imperial College London, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
9
|
Taïeb HM, Herment G, Robinson T, Cipitria A. Effect of capillary fluid flow on single cancer cell cycle dynamics, motility, volume and morphology. LAB ON A CHIP 2022; 23:92-105. [PMID: 36448429 DOI: 10.1039/d2lc00322h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
From primary tumours and disseminating to secondary organs, cancer cells experience a wide variety of fluid flow profiles when passing through blood vessels or the lymphatic system before extravasation. Sinusoidal capillaries are a common site for extravasation. Therefore, we aim to investigate how metastatic cancer cells react to a biophysical cue such as capillary fluid flow by quantifying its effect on metastatic cell cycle progression, motility, cell and nuclear volume, and morphology. We use MDA-MB-231 breast cancer cells genetically modified with fluorescent ubiquitination-based cell cycle indicator 2 (FUCCI2) as a model system. Single cells are trapped using a microfluidic device and exposed to different laminar flows. Quantitative time-lapse imaging in both 2D epifluorescence and 3D confocal microscopy is performed using in-house software FUCCItrack. In addition, 3D time-lapse with cell and nuclear segmentation is performed with a deep learning approach to streamline the image processing of big datasets. We show that at a single cell level, faster fluid flow leads to a shorter S/G2/M phase and an overall shorter cell cycle, as well as increase in cell motility irrespective of the flow direction. 3D time-lapse confocal imaging of MDA-FUCCI2 single cells reveals the evolution of cell and nuclear volume and morphology as a function of a specific cell cycle phase. Both cell and nuclear volume increase linearly over time. Cell morphology elongates more strongly during the S/G2/M phase, whereas the nuclear shape remains constant. Under the highest flow conditions, only during the S/G2/M phase can we observe a more elongated nucleus, while the cell sphericity remains similar to the control. Collectively, this data, together with the deep learning approach, provides new insights into the potential impact of fluid flow at a single cell level.
Collapse
Affiliation(s)
- Hubert M Taïeb
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany.
| | - Guillaume Herment
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany.
| | - Tom Robinson
- Department of Theory and Bio-Systems, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany.
| | - Amaia Cipitria
- Department of Biomaterials, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany.
- Group of Bioengineering in Regeneration and Cancer, Biodonostia Health Research Institute, 20014 San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
10
|
Intravital microscopy for real-time monitoring of drug delivery and nanobiological processes. Adv Drug Deliv Rev 2022; 189:114528. [PMID: 36067968 DOI: 10.1016/j.addr.2022.114528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 08/10/2022] [Accepted: 08/30/2022] [Indexed: 01/24/2023]
Abstract
Intravital microscopy (IVM) expands our understanding of cellular and molecular processes, with applications ranging from fundamental biology to (patho)physiology and immunology, as well as from drug delivery to drug processing and drug efficacy testing. In this review, we highlight modalities, methods and model organisms that make up today's IVM landscape, and we present how IVM - via its high spatiotemporal resolution - enables analysis of metabolites, small molecules, nanoparticles, immune cells, and the (tumor) tissue microenvironment. We furthermore present examples of how IVM facilitates the elucidation of nanomedicine kinetics and targeting mechanisms, as well as of biological processes such as immune cell death, host-pathogen interactions, metabolic states, and disease progression. We conclude by discussing the prospects of IVM clinical translation and examining the integration of machine learning in future IVM practice.
Collapse
|
11
|
Magliulo D, Bernardi R. Hypoxic stress and hypoxia-inducible factors in leukemias. Front Oncol 2022; 12:973978. [PMID: 36059690 PMCID: PMC9435438 DOI: 10.3389/fonc.2022.973978] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/21/2022] [Indexed: 11/17/2022] Open
Abstract
To cope with hypoxic stress, ancient organisms have developed evolutionally conserved programs centered on hypoxia-inducible transcriptional factors (HIFs). HIFs and their regulatory proteins have evolved as rheostats to adapt cellular metabolism to atmospheric oxygen fluctuations, but the amplitude of their transcriptional programs has tremendously increased along evolution to include a wide spectrum of physiological and pathological processes. The bone marrow represents a notable example of an organ that is physiologically exposed to low oxygen levels and where basal activation of hypoxia signaling appears to be intrinsically wired within normal and neoplastic hematopoietic cells. HIF-mediated responses are mainly piloted by the oxygen-labile α subunits HIF1α and HIF2α, and current literature suggests that these genes have a functional specification that remains to be fully defined. Since their identification in the mid 90s, HIF factors have been extensively studied in solid tumors, while their implication in leukemia has lagged behind. In the last decades however, many laboratories have addressed the function of hypoxia signaling in leukemia and obtained somewhat contradictory results. Suppression of HIFs expression in different types of leukemia has unveiled common leukemia-promoting functions such as stimulation of bone marrow neoangiogenesis, maintenance of leukemia stem cells and chemoresistance. However, genetic studies are revealing that a definition of HIF factors as bona fide tumor promoters is overly simplistic, and, depending on the leukemia subtype, the specific oncogenic event, or the stage of leukemia development, activation of hypoxia-inducible genes may lead to opposite consequences. With this article we will provide an updated summary of the studies describing the regulation and function of HIF1α and HIF2α in blood malignancies, spanning from acute to chronic, lymphoid to myeloid leukemias. In discussing these data, we will attempt to provide plausible explanations to contradictory findings and point at what we believe are areas of weakness in which further investigations are urgently needed. Gaining additional knowledge into the role of hypoxia signaling in leukemia appears especially timely nowadays, as new inhibitors of HIF factors are entering the clinical arena for specific types of solid tumors but their utility for patients with leukemia is yet to be determined.
Collapse
Affiliation(s)
| | - Rosa Bernardi
- Laboratory of Preclinical Models of Cancer, Division of Experimental Oncology, San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
12
|
Takihara Y, Higaki T, Yokomizo T, Umemoto T, Ariyoshi K, Hashimoto M, Sezaki M, Takizawa H, Inoue T, Suda T, Mizuno H. Bone marrow imaging reveals the migration dynamics of neonatal hematopoietic stem cells. Commun Biol 2022; 5:776. [PMID: 35918480 PMCID: PMC9346000 DOI: 10.1038/s42003-022-03733-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 07/15/2022] [Indexed: 12/03/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are produced from the blood vessel walls and circulate in the blood during the perinatal period. However, the migration dynamics of how HSCs enter the bone marrow remain elusive. To observe the dynamics of HSCs over time, the present study develops an intravital imaging method to visualize bone marrow in neonatal long bones formed by endochondral ossification which is essential for HSC niche formation. Endogenous HSCs are labeled with tdTomato under the control of an HSC marker gene Hlf, and a customized imaging system with a bone penetrating laser is developed for intravital imaging of tdTomato-labeled neonatal HSCs in undrilled tibia, which is essential to avoid bleeding from fragile neonatal tibia by bone drilling. The migration speed of neonatal HSCs is higher than that of adult HSCs. Neonatal HSCs migrate from outside to inside the tibia via the blood vessels that penetrate the bone, which is a transient structure during the neonatal period, and settle on the blood vessel wall in the bone marrow. The results obtained from direct observations in vivo reveal the motile dynamics and colonization process of neonatal HSCs during bone marrow formation. An intravital imaging method reveals the in vivo motile dynamics and colonization process of neonatal hematopoietic stem cells during bone marrow formation.
Collapse
Affiliation(s)
- Yuji Takihara
- Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan.,Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, #12-01, 117599, Singapore, Singapore
| | - Takumi Higaki
- Graduate School of Science and Technology, Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, Japan.,International Research Organization for Advanced Science and Technology (IROAST), Kumamoto University, 2-39-1 Kurokami, Chuo-ku, Kumamoto, Japan
| | - Tomomasa Yokomizo
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Terumasa Umemoto
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Kazunori Ariyoshi
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Michihiro Hashimoto
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Maiko Sezaki
- International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Hitoshi Takizawa
- Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan.,International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Toshihiro Inoue
- Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan
| | - Toshio Suda
- Cancer Science Institute of Singapore, National University of Singapore, 14 Medical Drive, #12-01, 117599, Singapore, Singapore. .,International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan.
| | - Hidenobu Mizuno
- Graduate School of Medical Sciences, Kumamoto University, 1-1-1 Honjo, Chuo-ku, Kumamoto, Japan. .,International Research Center for Medical Sciences (IRCMS), Kumamoto University, 2-2-1 Honjo, Chuo-ku, Kumamoto, Japan.
| |
Collapse
|
13
|
Trivanovic D, Harder J, Leucht M, Kreuzahler T, Schlierf B, Holzapfel BM, Rudert M, Jakob F, Herrmann M. Immune and stem cell compartments of acetabular and femoral bone marrow in hip osteoarthritis patients. Osteoarthritis Cartilage 2022; 30:1116-1129. [PMID: 35569800 DOI: 10.1016/j.joca.2022.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/22/2022] [Accepted: 05/02/2022] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Hip osteoarthritis (OA) affects all components of the osteochondral unit, leading to bone marrow (BM) lesions, and unknown consequences on BM cell functionality. We analyzed the cellular composition in OA-affected acetabula compared to proximal femur shafts obtained of hip OA patients to reveal yet not explored immune and stem cell compartments. DESIGN Combining flow cytometry, cellular assays and transcription analyses, we performed extensive ex vivo phenotyping of acetabular BM cells from 18 hip OA patients, comparing them with their counterparts from patient-matched femoral shaft BM samples. Findings were related to differences in skeletal sites and age. RESULTS Acetabular BM had a greater frequency of T-lymphocytes, non-hematopoietic cells and colony-forming units fibroblastic potential than femoral BM. The incidence of acetabular CD45+CD3+ T-lymphocytes increased (95% CI: 0.1770 to 0.0.8416), while clonogenic hematopoietic progenitors declined (95% CI: -0.9023 to -0.2399) with age of patients. On the other side, in femoral BM, we observed higher B-lymphocyte, myeloid and erythroid cell frequencies. Acetabular mesenchymal stromal cells (MSCs) showed a senescent profile associated with the expression of survival and inflammation-related genes. Efficient osteogenic and chondrogenic differentiation was detected in acetabular MSCs, while adipogenesis was more pronounced in their femoral counterparts. CONCLUSION Our results suggest that distinctions in BM cellular compartments and MSCs may be due to the influence of the OA-stressed microenvironment, but also acetabular vs femoral shaft-specific peculiarities cannot be excluded. These results bring new knowledge on acetabular BM cell populations and may be addressed as novel pathogenic mechanisms and therapeutic targets in OA.
Collapse
Affiliation(s)
- D Trivanovic
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - J Harder
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - M Leucht
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - T Kreuzahler
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - B Schlierf
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - B M Holzapfel
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Department of Orthopaedic Surgery, König-Ludwig-Haus, University of Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Department of Orthopaedic Surgery, University Clinics, Ludwig-Maximilians University Munich, Munich, 81377, Germany
| | - M Rudert
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Department of Orthopaedic Surgery, König-Ludwig-Haus, University of Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - F Jakob
- Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany
| | - M Herrmann
- IZKF Group Tissue Regeneration in Musculoskeletal Diseases, University Hospital Wuerzburg, Wuerzburg, Bavaria, 97070, Germany; Bernhard-Heine-Center for Locomotion Research, University Wuerzburg, Wuerzburg, Bavaria, 97070, Germany.
| |
Collapse
|
14
|
Ni R, Chen Z, Deán-Ben XL, Voigt FF, Kirschenbaum D, Shi G, Villois A, Zhou Q, Crimi A, Arosio P, Nitsch RM, Nilsson KPR, Aguzzi A, Helmchen F, Klohs J, Razansky D. Multiscale optical and optoacoustic imaging of amyloid-β deposits in mice. Nat Biomed Eng 2022; 6:1031-1044. [PMID: 35835994 DOI: 10.1038/s41551-022-00906-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 05/27/2022] [Indexed: 12/26/2022]
Abstract
Deposits of amyloid-β (Aβ) in the brains of rodents can be analysed by invasive intravital microscopy on a submillimetre scale, or via whole-brain images from modalities lacking the resolution or molecular specificity to accurately characterize Aβ pathologies. Here we show that large-field multifocal illumination fluorescence microscopy and panoramic volumetric multispectral optoacoustic tomography can be combined to longitudinally assess Aβ deposits in transgenic mouse models of Alzheimer's disease. We used fluorescent Aβ-targeted probes (the luminescent conjugated oligothiophene HS-169 and the oxazine-derivative AOI987) to transcranially detect Aβ deposits in the cortex of APP/PS1 and arcAβ mice with single-plaque resolution (8 μm) and across the whole brain (including the hippocampus and the thalamus, which are inaccessible by conventional intravital microscopy) at sub-150 μm resolutions. Two-photon microscopy, light-sheet microscopy and immunohistochemistry of brain-tissue sections confirmed the specificity and regional distributions of the deposits. High-resolution multiscale optical and optoacoustic imaging of Aβ deposits across the entire brain in rodents thus facilitates the in vivo study of Aβ accumulation by brain region and by animal age and strain.
Collapse
Affiliation(s)
- Ruiqing Ni
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - Zhenyue Chen
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Xosé Luís Deán-Ben
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Fabian F Voigt
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| | | | - Gloria Shi
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland
| | - Alessia Villois
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Quanyu Zhou
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
| | - Alessandro Crimi
- Institute of Neuropathology, Universitätsspital Zurich, Zurich, Switzerland
| | - Paolo Arosio
- Department of Chemistry and Applied Biosciences, ETH Zurich, Zurich, Switzerland
| | - Roger M Nitsch
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute for Regenerative Medicine, University of Zurich, Zurich, Switzerland
| | - K Peter R Nilsson
- Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping University, Linköping, Sweden
| | - Adriano Aguzzi
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Institute of Neuropathology, Universitätsspital Zurich, Zurich, Switzerland
| | - Fritjof Helmchen
- Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.,Brain Research Institute, University of Zurich, Zurich, Switzerland
| | - Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland. .,Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland.
| | - Daniel Razansky
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich, Zurich, Switzerland. .,Zurich Neuroscience Center (ZNZ), University of Zurich and ETH Zurich, Zurich, Switzerland. .,Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
| |
Collapse
|
15
|
Canalis E, Carrer M, Eller T, Schilling L, Yu J. Use of antisense oligonucleotides to target Notch3 in skeletal cells. PLoS One 2022; 17:e0268225. [PMID: 35536858 PMCID: PMC9089911 DOI: 10.1371/journal.pone.0268225] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 04/25/2022] [Indexed: 01/02/2023] Open
Abstract
Notch receptors are determinants of cell fate and function, and play an important role in the regulation of bone development and skeletal remodeling. Lateral Meningocele Syndrome (LMS) is a monogenic disorder associated with NOTCH3 pathogenic variants that result in the stabilization of NOTCH3 and a gain-of-function. LMS presents with neurological developmental abnormalities and bone loss. We created a mouse model (Notch3em1Ecan) harboring a 6691TAATGA mutation in the Notch3 locus, and heterozygous Notch3em1Ecan mice exhibit cancellous and cortical bone osteopenia. In the present work, we explored whether Notch3 antisense oligonucleotides (ASO) downregulate Notch3 and have the potential to ameliorate the osteopenia of Notch3em1Ecan mice. Notch3 ASOs decreased the expression of Notch3 wild type and Notch36691-TAATGA mutant mRNA expressed by Notch3em1Ecan mice in osteoblast cultures without evidence of cellular toxicity. The effect was specific since ASOs did not downregulate Notch1, Notch2 or Notch4. The expression of Notch3 wild type and Notch36691-TAATGA mutant transcripts also was decreased in bone marrow stromal cells and osteocytes following exposure to Notch3 ASOs. In vivo, the subcutaneous administration of Notch3 ASOs at 25 to 50 mg/Kg decreased Notch3 mRNA in the liver, heart and bone. Microcomputed tomography demonstrated that the administration of Notch3 ASOs ameliorates the cortical osteopenia of Notch3em1Ecan mice, and ASOs decreased femoral cortical porosity and increased cortical thickness and bone volume. However, the administration of Notch3 ASOs did not ameliorate the cancellous bone osteopenia of Notchem1Ecan mice. In conclusion, Notch3 ASOs downregulate Notch3 expression in skeletal cells and their systemic administration ameliorates cortical osteopenia in Notch3em1Ecan mice; as such ASOs may become useful strategies in the management of skeletal diseases affected by Notch gain-of-function.
Collapse
MESH Headings
- Abnormalities, Multiple
- Animals
- Bone Diseases, Metabolic/genetics
- Bone Diseases, Metabolic/metabolism
- Bone and Bones/diagnostic imaging
- Bone and Bones/metabolism
- Meningocele
- Mice
- Oligonucleotides, Antisense
- RNA, Messenger
- Receptor, Notch2/genetics
- Receptor, Notch2/metabolism
- Receptor, Notch3/genetics
- Receptor, Notch3/metabolism
- Receptors, Notch/genetics
- X-Ray Microtomography
Collapse
Affiliation(s)
- Ernesto Canalis
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
- Department of Medicine, UConn Health, Farmington, Connecticut, United States of America
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| | - Michele Carrer
- Ionis Pharmaceuticals, Inc., Carlsbad, California, United States of America
| | - Tabitha Eller
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| | - Lauren Schilling
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| | - Jungeun Yu
- Department of Orthopaedic Surgery, UConn Health, Farmington, Connecticut, United States of America
- The UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut, United States of America
| |
Collapse
|
16
|
Li W, Liu YH, Estrada H, Rebling J, Reiss M, Galli S, Nombela-Arrieta C, Razansky D. Tracking Strain-Specific Morphogenesis and Angiogenesis of Murine Calvaria with Large-Scale Optoacoustic and Ultrasound Microscopy. J Bone Miner Res 2022; 37:1032-1043. [PMID: 35220594 PMCID: PMC9311448 DOI: 10.1002/jbmr.4533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 01/30/2022] [Accepted: 02/20/2022] [Indexed: 11/25/2022]
Abstract
Skull bone development is a dynamic and well-coordinated process playing a key role in maturation and maintenance of the bone marrow (BM), fracture healing, and progression of diseases such as osteoarthritis or osteoporosis. At present, dynamic transformation of the growing bone (osteogenesis) as well as its vascularization (angiogenesis) remain largely unexplored due to the lack of suitable in vivo imaging techniques capable of noninvasive visualization of the whole developing calvaria at capillary-level resolution. We present a longitudinal study on skull bone development using ultrasound-aided large-scale optoacoustic microscopy (U-LSOM). Skull bone morphogenesis and microvascular growth patterns were monitored in three common mouse strains (C57BL/6J, CD-1, and Athymic Nude-Foxn1nu) at the whole-calvaria scale over a 3-month period. Strain-specific differences in skull development were revealed by quantitative analysis of bone and vessel parameters, indicating the coupling between angiogenesis and osteogenesis during skull bone growth in a minimally invasive and label-free manner. The method further enabled identifying BM-specific sinusoidal vessels, and superficial skull vessels penetrating into BM compartments. Our approach furnishes a new high-throughput longitudinal in vivo imaging platform to study morphological and vascular skull alterations in health and disease, shedding light on the critical links between blood vessel formation, skull growth, and regeneration. © 2022 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Weiye Li
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Yu-Hang Liu
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Héctor Estrada
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Johannes Rebling
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Michael Reiss
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| | - Serena Galli
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - César Nombela-Arrieta
- Department of Medical Oncology and Hematology, University and University Hospital Zurich, Zurich, Switzerland
| | - Daniel Razansky
- Institute for Biomedical Engineering and Institute of Pharmacology and Toxicology, Faculty of Medicine, University of Zurich, Zurich, Switzerland.,Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Dausinas Ni P, Basile C, Junge C, Hartman M, O’Leary HA. Hypoxia and Hematopoiesis. CURRENT STEM CELL REPORTS 2022. [DOI: 10.1007/s40778-021-00203-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
18
|
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
|
19
|
Myneni VD, Szalayova I, Mezey E. Differences in Steady-State Erythropoiesis in Different Mouse Bones and Postnatal Spleen. Front Cell Dev Biol 2021; 9:646646. [PMID: 34055777 PMCID: PMC8155546 DOI: 10.3389/fcell.2021.646646] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 04/22/2021] [Indexed: 11/13/2022] Open
Abstract
Adult erythropoiesis is a highly controlled sequential differentiation of hematopoietic stem cells (HSCs) to mature red blood cells in the bone marrow (BM). The bones which contain BM are diverse in their structure, embryonic origin, and mode of ossification. This has created substantial heterogeneity in HSCs function in BM of different bones, however, it is not known if this heterogeneity influences erythropoiesis in different bones and different regions of the same bone. In this study, we examined steady state BM erythroid progenitors and precursors from different bones - the femur, tibia, pelvis, sternum, vertebrae, radius, humerus, frontal, parietal bone, and compared all to the femur. Trabecular and cortical regions of the femur were also compared for differences in erythropoiesis. In addition, mouse spleen was studied to determine at which age erythropoietic support by the spleen was lost postnatally. We report that total erythroid cells, and erythroid precursors in the femur are comparable to tibia, pelvis, humerus and sternum, but are significantly reduced in the vertebrae, radius, frontal, and parietal bones. Erythroid progenitors and multipotential progenitor numbers are comparable in all the bones except for reduced number in the parietal bone. In the femur, the epiphysis and metaphysis have significantly reduced number of erythroid precursors and progenitors, multipotential progenitors and myeloid progenitors compared to the diaphysis region. These results show that analysis of erythroid precursors from diaphysis region of the femur is representative of tibia, pelvis, humerus and sternum and have significant implications on the interpretation of the steady-state erythropoiesis finding from adult BM. Postnatal spleen supports erythroid precursors until 6 weeks of age which coincides with reduced number of red pulp macrophages. The residual erythroid progenitor support reaches the adult level by 3 months of age. In conclusion, our findings provide insights to the differences in erythropoiesis between different bones, between trabecular and cortical regions of the femur, and developmental changes in postnatal spleen erythropoiesis.
Collapse
Affiliation(s)
- Vamsee D. Myneni
- Adult Stem Cell Section, Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| | | | - Eva Mezey
- Adult Stem Cell Section, Craniofacial and Skeletal Diseases Branch, National Institute of Dental and Craniofacial Research (NIDCR), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
20
|
Bonaud A, Lemos JP, Espéli M, Balabanian K. Hematopoietic Multipotent Progenitors and Plasma Cells: Neighbors or Roommates in the Mouse Bone Marrow Ecosystem? Front Immunol 2021; 12:658535. [PMID: 33936091 PMCID: PMC8083056 DOI: 10.3389/fimmu.2021.658535] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/25/2021] [Indexed: 11/25/2022] Open
Abstract
The bone marrow is a complex ecosystem in which hematopoietic and non-hematopoietic cells reside. In this review, we discuss the bone marrow niches in mice that facilitate the survival, maintenance, and differentiation of cells of hematopoietic origin based on the recent literature. Our review places a special focus on the hematopoietic multipotent progenitors and on plasma cells, corresponding to the last stage of the B-cell lineage, that play a key role in the humoral memory response. We highlight the similarities between the microenvironments necessary for the establishment and the maintenance of these two immune cell subsets, and how the chemokine CXCL12/CXCR4 signaling axis contributes to these processes. Finally, we bring elements to address the following question: are multipotent progenitors and plasma cells neighbors or roommates within the bone marrow?
Collapse
Affiliation(s)
- Amélie Bonaud
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Julia P Lemos
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Marion Espéli
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| | - Karl Balabanian
- Université de Paris, Institut de Recherche Saint-Louis, EMiLy, INSERM U1160, Paris, France.,OPALE Carnot Institute, The Organization for Partnerships in Leukemia, Hôpital Saint-Louis, Paris, France
| |
Collapse
|
21
|
Abstract
Quiescence is a cellular state in which a cell remains out of the cell cycle but retains the capacity to divide. The unique ability of adult stem cells to maintain quiescence is crucial for life-long tissue homeostasis and regenerative capacity. Quiescence has long been viewed as an inactive state but recent studies have shown that it is in fact an actively regulated process and that adult stem cells are highly reactive to extrinsic stimuli. This has fuelled hopes of boosting the reactivation potential of adult stem cells to improve tissue function during ageing. In this Review, we provide a perspective of the quiescent state and discuss how quiescent adult stem cells transition into the cell cycle. We also discuss current challenges in the field, highlighting recent technical advances that could help overcome some of these challenges.
Collapse
Affiliation(s)
- Noelia Urbán
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna Biocenter Campus (VBC), Dr. Bohr Gasse 3, 1030 Vienna, Austria
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, Center of Systems Biology and Human Health, State Key Laboratory of Molecular Neuroscience, and Molecular Neuroscience Center, Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
- Hong Kong Center for Neurodegenerative Diseases, Hong Kong Science Park, Hong Kong, China
- Guangdong Provincial Key Laboratory of Brain Science, Disease and Drug Development, The Hong Kong University of Science and Technology Shenzhen Research Institute, Shenzhen-Hong Kong Institute of Brain Science, Shenzhen, Guangdong 518057, China
| |
Collapse
|
22
|
Haltalli MLR, Lo Celso C. Intravital Imaging of Bone Marrow Niches. Methods Mol Biol 2021; 2308:203-222. [PMID: 34057725 DOI: 10.1007/978-1-0716-1425-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
Haematopoietic stem cells (HSCs) are instrumental in driving the generation of mature blood cells, essential for various functions including immune defense and tissue remodeling. They reside within a specialised bone marrow (BM) microenvironment , or niche, composed of cellular and chemical components that play key roles in regulating long-term HSC function and survival. While flow cytometry methods have significantly advanced studies of hematopoietic cells, enabling their quantification in steady-state and perturbed situations, we are still learning about the specific BM microenvironments that support distinct lineages and how their niches are altered under stress and with age. Major advances in imaging technology over the last decade have permitted in-depth studies of HSC niches in mice. Here, we describe our protocol for visualizing and analyzing the localization, morphology, and function of niche components in the mouse calvarium, using combined confocal and two-photon intravital microscopy, and we present the specific example of measuring vascular permeability.
Collapse
Affiliation(s)
- Myriam L R Haltalli
- Imperial College London, London, UK
- The Francis Crick Institute, London, UK
- Wellcome-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Cristina Lo Celso
- Imperial College London, London, UK.
- The Francis Crick Institute, London, UK.
| |
Collapse
|
23
|
Brooks J, Kumar B, Zuro DM, Raybuck JD, Madabushi SS, Vishwasrao P, Parra LE, Kortylewski M, Armstrong B, Froelich J, Hui SK. Biophysical Characterization of the Leukemic Bone Marrow Vasculature Reveals Benefits of Neoadjuvant Low-Dose Radiation Therapy. Int J Radiat Oncol Biol Phys 2021; 109:60-72. [PMID: 32841681 PMCID: PMC7736317 DOI: 10.1016/j.ijrobp.2020.08.037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 08/04/2020] [Accepted: 08/13/2020] [Indexed: 12/13/2022]
Abstract
PURPOSE Although vascular alterations in solid tumor malignancies are known to decrease therapeutic delivery, the effects of leukemia-induced bone marrow vasculature (BMV) alterations on therapeutic delivery are not well known. Additionally, functional quantitative measurements of the leukemic BMV during chemotherapy and radiation therapy are limited, largely due to a lack of high-resolution imaging techniques available preclinically. This study develops a murine model using compartmental modeling for quantitative multiphoton microscopy (QMPM) to characterize the malignant BMV before and during treatment. METHODS AND MATERIALS Using QMPM, live time-lapsed images of dextran leakage from the local BMV to the surrounding bone marrow of mice bearing acute lymphoblastic leukemia (ALL) were taken and fit to a 2-compartment model to measure the transfer rate (Ktrans), fractional extracellular extravascular space (νec), and vascular permeability parameters, as well as functional single-vessel characteristics. In response to leukemia-induced BMV alterations, the effects of 2 to 4 Gy low-dose radiation therapy (LDRT) on the BMV, drug delivery, and mouse survival were assessed post-treatment to determine whether neoadjuvant LDRT before chemotherapy improves treatment outcome. RESULTS Mice bearing ALL had significantly altered Ktrans, increased νec, and increased permeability compared with healthy mice. Angiogenesis, decreased single-vessel perfusion, and decreased vessel diameter were observed. BMV alterations resulted in disease-dependent reductions in cellular uptake of Hoechst dye. LDRT to mice bearing ALL dilated BMV, increased single-vessel perfusion, and increased daunorubicin uptake by ALL cells. Consequently, LDRT administered to mice before receiving nilotinib significantly increased survival compared with mice receiving LDRT after nilotinib, demonstrating the importance of LDRT conditioning before therapeutic administration. CONCLUSION The developed QMPM enables single-platform assessments of the pharmacokinetics of fluorescent agents and characterization of the BMV. Initial results suggest BMV alterations after neoadjuvant LDRT may contribute to enhanced drug delivery and increased treatment efficacy for ALL. The developed QMPM enables observations of the BMV for use in ALL treatment optimization.
Collapse
Affiliation(s)
- Jamison Brooks
- Department of Radiation Oncology, City of Hope, Duarte, California; Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | - Bijender Kumar
- Department of Radiation Oncology, City of Hope, Duarte, California; Hematology Malignancies and Stem Cell Transplantation Institute, City of Hope National Medical Center, Duarte, California
| | - Darren M Zuro
- Department of Radiation Oncology, City of Hope, Duarte, California; Department of Radiation Oncology, University of Minnesota, Minneapolis, Minnesota
| | | | | | | | | | - Marcin Kortylewski
- Department of Immuno-Oncology, City of Hope, Duarte, California; Beckman Research Institute of City of Hope, Duarte, California
| | - Brian Armstrong
- Beckman Research Institute of City of Hope, Duarte, California; Department of Development and Stem Cell Biology, City of Hope, Duarte, California
| | - Jerry Froelich
- Department of Radiology, University of Minnesota, Minneapolis, Minnesota
| | - Susanta K Hui
- Department of Radiation Oncology, City of Hope, Duarte, California; Beckman Research Institute of City of Hope, Duarte, California.
| |
Collapse
|
24
|
Estrada H, Rebling J, Hofmann U, Razansky D. Discerning calvarian microvascular networks by combined optoacoustic ultrasound microscopy. PHOTOACOUSTICS 2020; 19:100178. [PMID: 32215252 PMCID: PMC7090363 DOI: 10.1016/j.pacs.2020.100178] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 01/21/2020] [Accepted: 03/14/2020] [Indexed: 06/10/2023]
Abstract
Bone microvasculature plays a paramount role in bone marrow maintenance, development, and hematopoiesis. Studies of calvarian vascular patterns within living mammalian skull with the available intravital microscopy techniques are limited to small scale observations. We developed an optical-resolution optoacoustic microscopy method combined with ultrasound biomicroscopy in order to reveal and discern the intricate networks of calvarian and cerebral vasculature over large fields of view covering majority of the murine calvaria. The vasculature segmentation method is based on an angle-corrected homogeneous model of the rodent skull, generated using simultaneously acquired three-dimensional pulse-echo ultrasound images. The hybrid microscopy design along with the appropriate skull segmentation method enable high throughput studies of a living bone while facilitating correct anatomical interpretation of the vasculature images acquired with optical resolution optoacoustic microscopy.
Collapse
Affiliation(s)
- Héctor Estrada
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Johannes Rebling
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Urs Hofmann
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
| | - Daniel Razansky
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland
- Institute for Biomedical Engineering and Department of Information Technology and Electrical Engineering, ETH Zurich, Switzerland
- Institute for Biological and Medical Imaging (IBMI), Helmholtz Center Munich and Technical University of Munich, Germany
| |
Collapse
|
25
|
Ni R, Chen Z, Gerez JA, Shi G, Zhou Q, Riek R, Nilsson KPR, Razansky D, Klohs J. Detection of cerebral tauopathy in P301L mice using high-resolution large-field multifocal illumination fluorescence microscopy. BIOMEDICAL OPTICS EXPRESS 2020; 11:4989-5002. [PMID: 33014595 PMCID: PMC7510859 DOI: 10.1364/boe.395803] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/23/2020] [Accepted: 07/23/2020] [Indexed: 06/11/2023]
Abstract
Current intravital microscopy techniques visualize tauopathy with high-resolution, but have a small field-of-view and depth-of-focus. Herein, we report a transcranial detection of tauopathy over the entire cortex of P301L tauopathy mice using large-field multifocal illumination (LMI) fluorescence microscopy technique and luminescent conjugated oligothiophenes. In vitro assays revealed that fluorescent ligand h-FTAA is optimal for in vivo tau imaging, which was confirmed by observing elevated probe retention in the cortex of P301L mice compared to non-transgenic littermates. Immunohistochemical staining further verified the specificity of h-FTAA to detect tauopathy in P301L mice. The new imaging platform can be leveraged in pre-clinical mechanistic studies of tau spreading and clearance as well as longitudinal monitoring of tau targeting therapeutics.
Collapse
Affiliation(s)
- Ruiqing Ni
- University of Zurich & ETH Zurich, Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, Wolfgang-Pauli-strasse 27 HIT E22.4, 8093, Zurich, Switzerland
- University of Zurich, Zurich Neuroscience Center, Zurich, Switzerland
| | - Zhenyue Chen
- University of Zurich & ETH Zurich, Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, Wolfgang-Pauli-strasse 27 HIT E22.4, 8093, Zurich, Switzerland
- University of Zurich, Faculty of Medicine and Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Juan A. Gerez
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Zurich, Switzerland
| | - Gloria Shi
- University of Zurich & ETH Zurich, Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, Wolfgang-Pauli-strasse 27 HIT E22.4, 8093, Zurich, Switzerland
| | - Quanyu Zhou
- University of Zurich & ETH Zurich, Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, Wolfgang-Pauli-strasse 27 HIT E22.4, 8093, Zurich, Switzerland
- University of Zurich, Faculty of Medicine and Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Roland Riek
- ETH Zurich, Laboratory of Physical Chemistry, Department of Chemistry and Applied Biosciences, Zurich, Switzerland
| | - K. Peter R. Nilsson
- Linköping University, Division of Chemistry, Department of Physics, Chemistry and Biology, Linköping, Sweden
| | - Daniel Razansky
- University of Zurich & ETH Zurich, Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, Wolfgang-Pauli-strasse 27 HIT E22.4, 8093, Zurich, Switzerland
- University of Zurich, Zurich Neuroscience Center, Zurich, Switzerland
- University of Zurich, Faculty of Medicine and Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Jan Klohs
- University of Zurich & ETH Zurich, Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, Wolfgang-Pauli-strasse 27 HIT E22.4, 8093, Zurich, Switzerland
- University of Zurich, Zurich Neuroscience Center, Zurich, Switzerland
| |
Collapse
|
26
|
Abstract
The skeleton is highly vascularized due to the various roles blood vessels play in the homeostasis of bone and marrow. For example, blood vessels provide nutrients, remove metabolic by-products, deliver systemic hormones, and circulate precursor cells to bone and marrow. In addition to these roles, bone blood vessels participate in a variety of other functions. This article provides an overview of the afferent, exchange and efferent vessels in bone and marrow and presents the morphological layout of these blood vessels regarding blood flow dynamics. In addition, this article discusses how bone blood vessels participate in bone development, maintenance, and repair. Further, mechanical loading-induced bone adaptation is presented regarding interstitial fluid flow and pressure, as regulated by the vascular system. The role of the sympathetic nervous system is discussed in relation to blood vessels and bone. Finally, vascular participation in bone accrual with intermittent parathyroid hormone administration, a medication prescribed to combat age-related bone loss, is described and age- and disease-related impairments in blood vessels are discussed in relation to bone and marrow dysfunction. © 2020 American Physiological Society. Compr Physiol 10:1009-1046, 2020.
Collapse
Affiliation(s)
- Rhonda D Prisby
- Bone Vascular and Microcirculation Laboratory, Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
27
|
Stegner D, Heinze KG. Intravital imaging of megakaryocytes. Platelets 2020; 31:599-609. [PMID: 32153253 DOI: 10.1080/09537104.2020.1738366] [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: 10/24/2022]
Abstract
The dynamics of platelet formation could only be investigated since the development of two-photon microscopy in combination with suitable fluorescent labeling strategies. In this review paper, we give an overview of recent advances in fluorescence imaging of the bone marrow that have contributed to our understanding of platelet biogenesis during the last decade. We make a brief survey through the perspectives and limitations of today's intravital imaging, but also discuss complementary methods that may help to piece together the puzzle of megakaryopoiesis and platelet formation.
Collapse
Affiliation(s)
- David Stegner
- Institute of Experimental Biomedicine, University Hospital Würzburg , Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center for Experimental Biomedicine, University of Würzburg , Würzburg, Germany
| |
Collapse
|
28
|
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: 54] [Impact Index Per Article: 10.8] [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
|
29
|
Estrada H, Rebling J, Sievert W, Hladik D, Hofmann U, Gottschalk S, Tapio S, Multhoff G, Razansky D. Intravital optoacoustic and ultrasound bio-microscopy reveal radiation-inhibited skull angiogenesis. Bone 2020; 133:115251. [PMID: 31978616 DOI: 10.1016/j.bone.2020.115251] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 01/17/2020] [Accepted: 01/20/2020] [Indexed: 01/02/2023]
Abstract
Angiogenesis is critical in bone development and growth. Dense, large-scale, and multi-layered vascular networks formed by thin-walled sinusoidal vessels perfuse the plate bones and play an important role in bone repair. Yet, the intricate functional morphology of skull microvasculature remains poorly understood as it is difficult to visualize using existing intravital microscopy techniques. Here we introduced an intravital, fully-transcranial imaging approach based on hybrid optoacoustic and ultrasound bio-microscopy for large-scale observations and quantitative analysis of the vascular morphology, angiogenesis, vessel remodeling, and subsurface roughness in murine skulls. Our approach revealed radiation-inhibited angiogenesis in the skull bone. We also observed previously undocumented sinusoidal vascular networks spanning the entire skullcap, thus opening new vistas for studying the complex interactions between calvarial, pial, and cortical vascular systems.
Collapse
Affiliation(s)
- Héctor Estrada
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH, Zurich, Switzerland
| | - Johannes Rebling
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH, Zurich, Switzerland
| | - Wolfgang Sievert
- Center of Translational Cancer Research (TranslaTUM), Technical University of Munich, Germany; Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany
| | - Daniela Hladik
- Institute of Radiation Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Urs Hofmann
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH, Zurich, Switzerland
| | - Sven Gottschalk
- Institute of Biological and Medical Imaging (IBMI), Helmholtz Center Munich, Neuherberg, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Center Munich, Neuherberg, Germany
| | - Gabriele Multhoff
- Center of Translational Cancer Research (TranslaTUM), Technical University of Munich, Germany; Department of Radiation Oncology, Klinikum rechts der Isar, Munich, Germany; Institute of Pathology, Technical University of Munich, Germany
| | - Daniel Razansky
- Faculty of Medicine and Institute of Pharmacology and Toxicology, University of Zurich, Switzerland; Institute for Biomedical Engineering, Department of Information Technology and Electrical Engineering, ETH, Zurich, Switzerland; Center of Translational Cancer Research (TranslaTUM), Technical University of Munich, Germany; Institute of Biological and Medical Imaging (IBMI), Helmholtz Center Munich, Neuherberg, Germany.
| |
Collapse
|
30
|
Stefanowski J, Fiedler AF, Köhler M, Günther R, Liublin W, Tschaikner M, Rauch A, Reismann D, Matthys R, Nützi R, Bixel MG, Adams RH, Niesner RA, Duda GN, Hauser AE. Limbostomy: Longitudinal Intravital Microendoscopy in Murine Osteotomies. Cytometry A 2020; 97:483-495. [PMID: 32196971 DOI: 10.1002/cyto.a.23997] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Revised: 01/20/2020] [Accepted: 02/24/2020] [Indexed: 12/28/2022]
Abstract
Bone healing involves the interplay of immune cells, mesenchymal cells, and vasculature over the time course of regeneration. Approaches to quantify the spatiotemporal aspects of bone healing at cellular resolution during long bone healing do not yet exist. Here, a novel technique termed Limbostomy is presented, which combines intravital microendoscopy with an osteotomy. This design allows a modular combination of an internal fixator plate with a gradient refractive index (GRIN) lens at various depths in the bone marrow and can be combined with a surgical osteotomy procedure. The field of view (FOV) covers a significant area of the fracture gap and allows monitoring cellular processes in vivo. The GRIN lens causes intrinsic optical aberrations which have to be corrected. The optical system was characterized and a postprocessing algorithm was developed. It corrects for wave front aberration-induced image plane deformation and for background and noise signals, enabling us to observe subcellular processes. Exemplarily, we quantitatively and qualitatively analyze angiogenesis in bone regeneration. We make use of a transgenic reporter mouse strain with nucleargreen fluorescent protein and membrane-bound tdTomato under the Cadherin-5 promoter. We observe two phases of vascularization. First, rapid vessel sprouting pervades the FOV within 3-4 days after osteotomy. Second, the vessel network continues to be dynamically remodeled until the end of our observation time, 14 days after surgery. Limbostomy opens a unique set of opportunities and allows further insight on spatiotemporal aspects of bone marrow biology, for example, hematopoiesis, analysis of cellular niches, immunological memory, and vascularization in the bone marrow during health and disease. © 2020 The Authors. Cytometry Part A published by Wiley Periodicals, Inc. on behalf of International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Jonathan Stefanowski
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Alexander F Fiedler
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Freie Universitat Berlin, Veterinary Medicine, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Markus Köhler
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Freie Universitat Berlin, Veterinary Medicine, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Robert Günther
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Wjatscheslaw Liublin
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Martin Tschaikner
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - Ariana Rauch
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | - David Reismann
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| | | | | | | | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Raluca A Niesner
- Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany.,Freie Universitat Berlin, Veterinary Medicine, Dynamic and Functional in vivo Imaging, Berlin, Germany
| | - Georg N Duda
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin Institute of Health Center for Regenerative Therapies, Berlin, Germany.,Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Julius Wolff Institute, Berlin, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Deutsches Rheuma-Forschungszentrum (DRFZ) Berlin, a Leibniz Institute, Berlin, Germany
| |
Collapse
|
31
|
Boroumand P, Klip A. Bone marrow adipose cells - cellular interactions and changes with obesity. J Cell Sci 2020; 133:133/5/jcs238394. [PMID: 32144195 DOI: 10.1242/jcs.238394] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The bone marrow is a spatially restricted niche, housing cells of the hematopoietic and mesenchymal lineages in various hierarchical commitment states. Although highly localized, cells within this niche are also subject to regulation by environmental and/or circulatory changes through extensive vascularization. Bone marrow adipocytes, derived from mesenchymal stem cells and once known as marrow space fillers, are a heterogeneous population. These cells reside in distinct niches within the bone marrow and interact with proximal cells, such as hematopoietic precursors and lineage-committed cells. In this diverse cellular milieu, bone marrow adipocytes influence commitment decisions and cellular lineage selection by interacting with stem and progenitor cells. In addition, bone marrow adipocytes respond to environmental changes, such as obesity, by undergoing hypertrophy, hyperplasia or adoption of characteristics resembling those of peripheral brown, beige or white adipocytes. Here, we review recent findings and concepts on the influence of bone marrow adipocytes on hematopoietic and other cellular lineages within this niche. We discuss how changes in local, systemic, cellular and secreted signals impact on mesenchymal stem cell expansion, differentiation and lineage commitment. Furthermore, we highlight that bone marrow adipocytes may be intermediaries conveying environmental cues to influence hematopoietic cellular survival, proliferation and preferential differentiation.
Collapse
Affiliation(s)
- Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada.,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario M5G 0A4, Canada .,Department of Biochemistry, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| |
Collapse
|
32
|
Live-animal imaging of native haematopoietic stem and progenitor cells. Nature 2020; 578:278-283. [PMID: 32025033 PMCID: PMC7021587 DOI: 10.1038/s41586-020-1971-z] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2018] [Accepted: 12/06/2019] [Indexed: 12/26/2022]
Abstract
The biology of hematopoietic stem cells (HSCs) has predominantly been studied under transplantation conditions1,2. Particularly challenging has been the study of dynamic HSC behaviors given that live animal HSC visualization in the native niche still represents an elusive goal in the field. Here, we describe a dual genetic strategy in mice that restricts reporter labeling to a subset of the most quiescent long-term HSCs (LT-HSCs) and that is compatible with current intravital imaging approaches in the calvarial bone marrow (BM)3–5. We find that this subset of LT-HSCs resides in close proximity to both sinusoidal blood vessels and the endosteal surface. In contrast, multipotent progenitor cells (MPPs) display a broader distance distribution from the endosteum and are more likely to be associated with transition zone vessels. LT-HSCs are not found in BM niches with the deepest hypoxia and instead are found in similar hypoxic environments as MPPs. In vivo time-lapse imaging reveals that LT-HSCs display limited motility at steady-state. Following activation, LT-HSCs display heterogenous responses, with some cells becoming highly motile and a fraction of HSCs expanding clonally within spatially restricted domains. These domains have defined characteristics, as HSC expansion is found almost exclusively in a subset of BM cavities exhibiting bone-remodeling activities. In contrast, cavities with low bone-resorbing activities do not harbor expanding HSCs. These findings point to a new degree of heterogeneity within the BM microenvironment, imposed by the stages of bone turnover. Overall, our approach enables direct visualization of HSC behaviors and dissection of heterogeneity in HSC niches.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW We reviewed recent progress on the role of sclerostin (SOST) and its effects on the immune system in order to summarize the current state of knowledge in osteoimmunology, in regard to hematopoiesis, lymphopoiesis, and inflammation. RECENT FINDINGS Changes in sclerostin levels affect distinct niches within the bone marrow that support hematopoietic stem cells and B cell development. Sclerostin's regulation of adipogenesis could also be important for immune cell maintenance with age. Surprisingly, B cell development in the bone marrow is influenced by Sost produced by mesenchymal stem cells and osteoblasts, but not by osteocytes. Additionally, extramedullary hematopoiesis in the spleen and increased pro-inflammatory cytokine levels in the bone marrow are observed in global Sost-/- mice. In addition to changes in bone marrow density, sclerostin depletion affects B lymphopoiesis and myelopoiesis, as well as other changes within the bone marrow cavity that could affect hematopoiesis. It is therefore important to monitor for hematopoietic changes in patients receiving sclerostin-depleting therapies.
Collapse
Affiliation(s)
- Cristine Donham
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA
| | - Jennifer O Manilay
- Quantitative and Systems Biology Graduate Program, University of California, Merced, Merced, CA, 95343, USA.
- Dept. of Molecular and Cell Biology, School of Natural Sciences, University of California, Merced, 5200, Lake Road Merced, North, CA, 95343, USA.
| |
Collapse
|
34
|
Wang D, Gilbert JR, Zhang X, Zhao B, Ker DFE, Cooper GM. Calvarial Versus Long Bone: Implications for Tailoring Skeletal Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2019; 26:46-63. [PMID: 31588853 DOI: 10.1089/ten.teb.2018.0353] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Tissue-engineered graft substitutes have shown great potential to treat large bone defects. While we usually assume that therapeutic approaches developed for appendicular bone healing could be similarly translated for application in craniofacial reconstruction and vice versa, this is not necessarily accurate. In addition to those more well-known healing-associated factors, such as age, lifestyle (e.g., nutrition and smoking), preexisting disease (e.g., diabetes), medication, and poor blood supply, the developmental origins and surrounding tissue of the wound sites can largely affect the fracture healing outcome as well as designed treatments. Therefore, the strategies developed for long bone fracture repair might not be suitable or directly applicable to skull bone repair. In this review, we discuss aspects of development, healing process, structure, and tissue engineering considerations between calvarial and long bones to assist in designing the tailored bone repair strategies. Impact Statement We summarized, in this review, the existing body of knowledge between long bone and calvarial bone with regard to their development and healing, tissue structure, and consideration of current tissue engineering strategies. By highlighting their similarities and differences, we propose that tailored tissue engineering strategies, such as scaffold features, growth factor usage, and the source of cells for tissue or region-specific bone repair, are necessary to ensure an optimized healing outcome.
Collapse
Affiliation(s)
- Dan Wang
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - James R Gilbert
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xu Zhang
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Bingkun Zhao
- Department of Stomatology, Tenth People's Hospital of Tongji University, Shanghai, China.,Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Dai Fei Elmer Ker
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Gregory M Cooper
- Department of Plastic Surgery, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Oral Biology, University of Pittsburgh, Pittsburgh, Pennsylvania.,Department of Bioengineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| |
Collapse
|
35
|
Kim J, Bixel MG. Intravital Multiphoton Imaging of the Bone and Bone Marrow Environment. Cytometry A 2019; 97:496-503. [DOI: 10.1002/cyto.a.23937] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 10/30/2019] [Accepted: 11/05/2019] [Indexed: 12/11/2022]
Affiliation(s)
- JungMo Kim
- Department of Tissue MorphogenesisMax Planck Institute for Molecular Biomedicine D‐48149 Münster Germany
| | - Maria Gabriele Bixel
- Department of Tissue MorphogenesisMax Planck Institute for Molecular Biomedicine D‐48149 Münster Germany
| |
Collapse
|
36
|
Dondossola E, Alexander S, Holzapfel BM, Filippini S, Starbuck MW, Hoffman RM, Navone N, De-Juan-Pardo EM, Logothetis CJ, Hutmacher DW, Friedl P. Intravital microscopy of osteolytic progression and therapy response of cancer lesions in the bone. Sci Transl Med 2019; 10:10/452/eaao5726. [PMID: 30068572 DOI: 10.1126/scitranslmed.aao5726] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 01/16/2018] [Accepted: 06/19/2018] [Indexed: 12/12/2022]
Abstract
Intravital multiphoton microscopy (iMPM) in mice provides access to cellular and molecular mechanisms of metastatic progression of cancers and the underlying interactions with the tumor stroma. Whereas iMPM of malignant disease has been performed for soft tissues, noninvasive iMPM of solid tumor in the bone is lacking. We combined miniaturized tissue-engineered bone constructs in nude mice with a skin window to noninvasively and repetitively monitor prostate cancer lesions by three-dimensional iMPM. In vivo ossicles developed large central cavities containing mature bone marrow surrounded by a thin cortex and enabled tumor implantation and longitudinal iMPM over weeks. Tumors grew inside the bone cavity and along the cortical bone interface and induced niches of osteoclast activation (focal osteolysis). Interventional bisphosphonate therapy reduced osteoclast kinetics and osteolysis without perturbing tumor growth, indicating dissociation of the tumor-stroma axis. The ossicle window, with its high cavity-to-cortex ratio and long-term functionality, thus allows for the mechanistic dissection of reciprocal epithelial tumor-bone interactions and therapy response.
Collapse
Affiliation(s)
- Eleonora Dondossola
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Stephanie Alexander
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Boris M Holzapfel
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, Queensland 4059, Australia.,Orthopaedic Center for Musculoskeletal Research, University of Würzburg, Brettreichstraße 11, 97074 Würzburg, Germany
| | - Stefano Filippini
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Michael W Starbuck
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Robert M Hoffman
- Department of Surgery, University of California, San Diego and AntiCancer Inc., 7917 Ostrow Street, San Diego, CA 92111, USA
| | - Nora Navone
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Elena M De-Juan-Pardo
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Christopher J Logothetis
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, Institute of Health and Biomedical Innovation, Queensland University of Technology (QUT), 60 Musk Avenue, Kelvin Grove, Brisbane, Queensland 4059, Australia.,ARC Centre in Additive Biomanufacturing, QUT, 60 Musk Avenue, Kelvin Grove, Brisbane, Queensland 4059, Australia
| | - Peter Friedl
- David H. Koch Center for Applied Research of Genitourinary Cancers, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA. .,Radboud University Nijmegen, Nijmegen, Netherlands.,Cancer Genomics Centre (CGC.nl), 3584 Utrecht, Netherlands
| |
Collapse
|
37
|
Liu Y, Yuan Q, Zhang S. Three-dimensional intravital imaging in bone research. JOURNAL OF BIOPHOTONICS 2019; 12:e201960075. [PMID: 31593614 DOI: 10.1002/jbio.201960075] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 09/11/2019] [Accepted: 09/16/2019] [Indexed: 02/05/2023]
Abstract
Intravital imaging has emerged as a novel and efficient tool for visualization of in situ dynamics of cellular behaviors and cell-microenvironment interactions in live animals, based on desirable microscopy techniques featuring high resolutions, deep imaging and low phototoxicity. Intravital imaging, especially based on multi-photon microscopy, has been used in bone research for dynamics visualization of a variety of physiological and pathological events at the cellular level, such as bone remodeling, hematopoiesis, immune responses and cancer development, thus, providing guidance for elucidating novel cellular mechanisms in bone biology as well as guidance for new therapies. This review is aimed at interpreting development and advantages of intravital imaging in bone research, and related representative discoveries concerning bone matrices, vessels, and various cells types involved in bone physiologies and pathologies. Finally, current limitations, further refinement, and extended application of intravital imaging in bone research are concluded.
Collapse
Affiliation(s)
- Yuhao Liu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Quan Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shiwen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
38
|
Next-generation imaging of the skeletal system and its blood supply. Nat Rev Rheumatol 2019; 15:533-549. [PMID: 31395974 DOI: 10.1038/s41584-019-0274-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/08/2019] [Indexed: 12/16/2022]
Abstract
Bone is organized in a hierarchical 3D architecture. Traditionally, analysis of the skeletal system was based on bone mass assessment by radiographic methods or on the examination of bone structure by 2D histological sections. Advanced imaging technologies and big data analysis now enable the unprecedented examination of bone and provide new insights into its 3D macrostructure and microstructure. These technologies comprise ex vivo and in vivo methods including high-resolution computed tomography (CT), synchrotron-based imaging, X-ray microscopy, ultra-high-field magnetic resonance imaging (MRI), light-sheet fluorescence microscopy, confocal and intravital two-photon imaging. In concert, these techniques have been used to detect and quantify a novel vascular system of trans-cortical vessels in bone. Furthermore, structures such as the lacunar network, which harbours and connects osteocytes, become accessible for 3D imaging and quantification using these methods. Next-generation imaging of the skeletal system and its blood supply are anticipated to contribute to an entirely new understanding of bone tissue composition and function, from macroscale to nanoscale, in health and disease. These insights could provide the basis for early detection and precision-type intervention of bone disorders in the future.
Collapse
|
39
|
Calvo J, Fahy L, Uzan B, Pflumio F. Desperately seeking a home marrow niche for T-cell acute lymphoblastic leukaemia. Adv Biol Regul 2019; 74:100640. [PMID: 31378700 DOI: 10.1016/j.jbior.2019.100640] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/19/2019] [Accepted: 07/25/2019] [Indexed: 02/07/2023]
Abstract
T-cell acute leukemia is a hematologic malignancy that results from the progressive acquisition of genomic abnormalities in T-cell progenitors/precursors. T-ALL is commonly thought to originate from the thymus albeit recent literature describes the possible acquisition of the first oncogenic hits in hematopoietic progenitor cells of the bone marrow (BM). The journey of T-ALL from its arising to full blown expansion meets different microenvironments, including the BM in which leukemic cells settle down early after the disease spreading. We take advantage of recent literature to give an overview of important cells and factors that participate in T-ALL, especially in the BM, arguing in favor of a home marrow niche for this rare leukemia.
Collapse
Affiliation(s)
- Julien Calvo
- UMRE008 Stabilité Génétique Cellules Souches et Radiations, U1274 Inserm, Université de Paris, Université Paris-Saclay, CEA, F-92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia, Team Niche and Cancer in Hematopoiesis, U1274, Inserm, CEA, 18 route du panorama, 92260, Fontenay-aux-Roses, France; Laboratoire labellisé par l'Association pour la Recherche sur le Cancer, France
| | - Lucine Fahy
- UMRE008 Stabilité Génétique Cellules Souches et Radiations, U1274 Inserm, Université de Paris, Université Paris-Saclay, CEA, F-92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia, Team Niche and Cancer in Hematopoiesis, U1274, Inserm, CEA, 18 route du panorama, 92260, Fontenay-aux-Roses, France; Laboratoire labellisé par l'Association pour la Recherche sur le Cancer, France
| | - Benjamin Uzan
- UMRE008 Stabilité Génétique Cellules Souches et Radiations, U1274 Inserm, Université de Paris, Université Paris-Saclay, CEA, F-92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia, Team Niche and Cancer in Hematopoiesis, U1274, Inserm, CEA, 18 route du panorama, 92260, Fontenay-aux-Roses, France; Laboratoire labellisé par l'Association pour la Recherche sur le Cancer, France
| | - Françoise Pflumio
- UMRE008 Stabilité Génétique Cellules Souches et Radiations, U1274 Inserm, Université de Paris, Université Paris-Saclay, CEA, F-92260 Fontenay-aux-Roses, France; Laboratory of Hematopoietic Stem Cells and Leukemia, Team Niche and Cancer in Hematopoiesis, U1274, Inserm, CEA, 18 route du panorama, 92260, Fontenay-aux-Roses, France; Laboratoire labellisé par l'Association pour la Recherche sur le Cancer, France.
| |
Collapse
|
40
|
Halvarsson C, Rörby E, Eliasson P, Lang S, Soneji S, Jönsson JI. Putative Role of Nuclear Factor-Kappa B But Not Hypoxia-Inducible Factor-1α in Hypoxia-Dependent Regulation of Oxidative Stress in Hematopoietic Stem and Progenitor Cells. Antioxid Redox Signal 2019; 31:211-226. [PMID: 30827134 PMCID: PMC6590716 DOI: 10.1089/ars.2018.7551] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 03/01/2019] [Accepted: 03/01/2019] [Indexed: 12/14/2022]
Abstract
Aims: Adaptation to low oxygen of hematopoietic stem cells (HSCs) in the bone marrow has been demonstrated to depend on the activation of hypoxia-inducible factor (HIF)-1α as well as the limited production of reactive oxygen species (ROS). In this study, we aimed at determining whether HIF-1α is involved in protecting HSCs from ROS. Results: Oxidative stress was induced by DL-buthionine-(S,R)-sulfoximine (BSO)-treatment, which increases the mitochondrial ROS level. Hypoxia rescued Lineage-Sca-1+c-kit+ (LSK) cells from BSO-induced apoptosis, whereas cells succumbed to apoptosis in normoxia. Apoptosis in normoxia was inhibited with the antioxidant N-acetyl-L-cysteine or by overexpression of anti-apoptotic BCL-2. Moreover, stabilized expression of oxygen-insensitive HIFs could not protect LSK cells from oxidative stress-induced apoptosis at normoxia, neither could short hairpin RNA to Hif-1α inhibit the protective effects by hypoxia in LSK cells. Likewise, BSO treatment of LSK cells from Hif-1α knockout mice did not suppress the effects seen in hypoxia. Microarray analysis identified the nuclear factor-kappa B (NF-κB) pathway as a pathway induced by hypoxia. By using NF-κB lentiviral construct and DNA-binding assay, we found increased NF-κB activity in cells cultured in hypoxia compared with normoxia. Using an inhibitor against NF-κB activation, we could confirm the involvement of NF-κB signaling as BSO-mediated cell death was significantly increased in hypoxia after adding the inhibitor. Innovation: HIF-1α is not involved in protecting HSCs and progenitors to elevated levels of ROS on glutathione depletion during hypoxic conditions. Conclusion: The study proposes a putative role of NF-κB signaling as a hypoxia-induced regulator in early hematopoietic cells.
Collapse
Affiliation(s)
- Camilla Halvarsson
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Emma Rörby
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Pernilla Eliasson
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - Stefan Lang
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Shamit Soneji
- Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Jan-Ingvar Jönsson
- Experimental Hematology Unit, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
41
|
Tjin G, Flores-Figueroa E, Duarte D, Straszkowski L, Scott M, Khorshed RA, Purton LE, Lo Celso C. Imaging methods used to study mouse and human HSC niches: Current and emerging technologies. Bone 2019; 119:19-35. [PMID: 29704697 DOI: 10.1016/j.bone.2018.04.022] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 04/23/2018] [Accepted: 04/23/2018] [Indexed: 12/18/2022]
Abstract
Bone marrow contains numerous different cell types arising from hematopoietic stem cells (HSCs) and non-hematopoietic mesenchymal/skeletal stem cells, in addition to other cell types such as endothelial cells- these non-hematopoietic cells are commonly referred to as stromal cells or microenvironment cells. HSC function is intimately linked to complex signals integrated by their niches, formed by combinations of hematopoietic and stromal cells. Studies of hematopoietic cells have been significantly advanced by flow cytometry methods, enabling the quantitation of each cell type in normal and perturbed situations, in addition to the isolation of these cells for molecular and functional studies. Less is known, however, about the specific niches for distinct developing hematopoietic lineages, or the changes occurring in the niche size and function in these distinct anatomical sites in the bone marrow under stress situations and ageing. Significant advances in imaging technology during the last decade have permitted studies of HSC niches in mice. Additional imaging technologies are emerging that will facilitate the study of human HSC niches in trephine BM biopsies. Here we provide an overview of imaging technologies used to study HSC niches, in addition to highlighting emerging technology that will help us to more precisely identify and characterize HSC niches in normal and diseased states.
Collapse
Affiliation(s)
- Gavin Tjin
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Eugenia Flores-Figueroa
- Oncology Research Unit, Oncology Hospital, National Medical Center Century XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Delfim Duarte
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; The Sir Francis Crick Institute, London, UK
| | - Lenny Straszkowski
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Mark Scott
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Reema A Khorshed
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK
| | - Louise E Purton
- St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia; The University of Melbourne, Department of Medicine at St Vincent's Hospital, Fitzroy, Victoria, Australia.
| | - Cristina Lo Celso
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, London, UK; The Sir Francis Crick Institute, London, UK.
| |
Collapse
|
42
|
Abstract
Two-photon intravital microscopy (2P-IVM) is an advanced imaging technique that allows the visualization of dynamic cellular behavior deeply inside tissues and organs of living animals. Due to the deep tissue penetration, imaging of highly light-scattering tissue as the bone becomes feasible at subcellular resolution.To better understand the influence of blood flow on hematopoietic stem and progenitor cell (HSPC) homing to the bone marrow (BM) microvasculature of the calvarial bone, we analyzed blood flow dynamics and the influence of flow on the early homing behavior of HSPCs during their passage through BM microvessels. Here, we describe a 2P-IVM approach for direct measurements of red blood cell (RBC) velocities in the BM microvasculature using repetitive centerline scans at the level of individual arterial vessels and sinusoidal capillaries to obtain a detailed flow profile map. Furthermore, we explain the isolation and enrichment of HSPCs from long bones and the transplantation of these cells to study the early homing behavior of HSPCs in BM sinusoids at cellular resolution. This is achieved by high-resolution spatiotemporal imaging through a chronic cranial window using transgenic reporter mice.
Collapse
Affiliation(s)
- Jonas Stewen
- Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany
| | - Maria Gabriele Bixel
- Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, Münster, Germany.
| |
Collapse
|
43
|
Abstract
Hematopoietic stem cells are maintained and regulated in spatially confined microenvironments within the bone marrow, in which oxygen availability is hypothesized to be very limited. The hypoxic nature of HSC niches is proposed to play a fundamental role in the preservation of fundamental stem cell properties through the induction of a distinct glycolytic metabolic profile in HSCs. Thus, the capacity to determine oxygen levels or cellular oxygenation status in specific tissue locations is essential to deepen our understanding of HSC biology. We here describe a methodology to indirectly quantify the hypoxic status of individual cells in situ within histological sections of bone marrow tissues. We employ the well-characterized nitroimidazole probe, pimonidazole, which acts as an oxygen mimetic and irreversibly incorporates into cellular proteins only under hypoxic conditions. The use of fluorescently labeled antibodies that recognize pimonidazole epitopes then enables the indirect assessment of the intracellular hypoxic status and its relationship to cell positioning within the complex tissue topography of the bone marrow.
Collapse
|
44
|
Abstract
INTRODUCTION Hypoxia-inducible transcription factors have been identified as regulators of adaptive responses to hypoxia. Over the past 20 years, more than 8000 papers have described their increasingly complex role and regulation in cancer. Presently, it is recognized that hypoxia-inducible factors (HIFs) are regulated by oxygen-dependent and oxygen-independent mechanisms in cancer development; the list of their targets has increased to include more than 500 genes involved in most hallmarks of cancer. Areas covered: Most literature describes the function of HIF factors in solid tumors; however, in the past 10 years, evidence has steadily accumulated to indicate that HIFs are implicated in hematological malignancies. This review summarizes our current understanding of the function and regulation of HIF factors in hematopoiesis and leukemia. Moreover, we provide an update on pharmacological inhibitors of this pathway that have shown promising therapeutic effects in clinical trials or leukemia pre-clinical models. Expert opinion: The inhibition of the function of HIF factors may provide an interesting approach for treating leukemia. We posit that before moving into the clinic, we should (i) fully characterize the outcome of HIF inhibition in specific leukemia contexts (ii) test the possibility of combining HIF-targeting strategies with cytotoxic compounds and (iii) consider patient selection to increase therapeutic efficacy.
Collapse
Affiliation(s)
- Daniela Magliulo
- a Vita-Salute San Raffaele University , Milan , Italy.,b Preclinical Models of Cancer Laboratory, Division of Experimental Oncology , San Raffaele Scientific Institute , Milan , Italy
| | - Rosa Bernardi
- b Preclinical Models of Cancer Laboratory, Division of Experimental Oncology , San Raffaele Scientific Institute , Milan , Italy
| |
Collapse
|
45
|
Magome T, Froelich J, Holtan SG, Takahashi Y, Verneris MR, Brown K, Dusenbery K, Wong J, Hui SK. Whole-Body Distribution of Leukemia and Functional Total Marrow Irradiation Based on FLT-PET and Dual-Energy CT. Mol Imaging 2018; 16:1536012117732203. [PMID: 28948859 PMCID: PMC5624344 DOI: 10.1177/1536012117732203] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
This report describes a multimodal whole-body 3′-deoxy-3′[(18)F]-fluorothymidine positron emission tomography (FLT-PET) and dual-energy computed tomography (DECT) method to identify leukemia distribution within the bone marrow environment (BME) and to develop disease- and/or BME-specific radiation strategies. A control participant and a newly diagnosed patient with acute myeloid leukemia prior to induction chemotherapy were scanned with FLT-PET and DECT. The red marrow (RM) and yellow marrow (YM) of the BME were segmented from DECT using a basis material decomposition method. Functional total marrow irradiation (fTMI) treatment planning simulations were performed combining FLT-PET and DECT imaging to differentially target irradiation to the leukemia niche and the rest of the skeleton. Leukemia colonized both RM and YM regions, adheres to the cortical bone in the spine, and has enhanced activity in the proximal/distal femur, suggesting a potential association of leukemia with the BME. The planning target volume was reduced significantly in fTMI compared with conventional TMI. The dose to active disease (standardized uptake value >4) was increased by 2-fold, while maintaining doses to critical organs similar to those in conventional TMI. In conclusion, a hybrid system of functional–anatomical–physiological imaging can identify the spatial distribution of leukemia and will be useful to both help understand the leukemia niche and develop targeted radiation strategies.
Collapse
Affiliation(s)
- Taiki Magome
- 1 Department of Radiological Sciences, Faculty of Health Sciences, Komazawa University, Tokyo, Japan.,2 Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA
| | - Jerry Froelich
- 3 Department of Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Shernan G Holtan
- 4 Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | - Yutaka Takahashi
- 2 Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,5 Department of Radiation Oncology, Osaka University, Osaka, Japan
| | - Michael R Verneris
- 4 Blood and Marrow Transplant Program, University of Minnesota, Minneapolis, MN, USA
| | | | - Kathryn Dusenbery
- 7 Department of Therapeutic Radiology, University of Minnesota, Minneapolis, MN, USA
| | - Jeffrey Wong
- 8 Department of Radiation Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| | - Susanta K Hui
- 2 Masonic Cancer Center, University of Minnesota, Minneapolis, MN, USA.,7 Department of Therapeutic Radiology, University of Minnesota, Minneapolis, MN, USA.,8 Department of Radiation Oncology, Beckman Research Institute, City of Hope, Duarte, CA, USA
| |
Collapse
|
46
|
van Rhijn-Brouwer FCC, Gremmels H, Fledderus JO, Verhaar MC. Mesenchymal Stromal Cell Characteristics and Regenerative Potential in Cardiovascular Disease: Implications for Cellular Therapy. Cell Transplant 2018; 27:765-785. [PMID: 29895169 PMCID: PMC6047272 DOI: 10.1177/0963689717738257] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Administration of mesenchymal stromal cells (MSCs) is a promising strategy to treat cardiovascular disease (CVD). As progenitor cells may be negatively affected by both age and comorbidity, characterization of MSC function is important to guide decisions regarding use of allogeneic or autologous cells. Definitive answers on which factors affect MSC function can also aid in selecting which MSC donors would yield the most therapeutically efficacious MSCs. Here we provide a narrative review of MSC function in CVD based on a systematic search. A total of 41 studies examining CVD-related MSC (dys)function were identified. These data show that MSC characteristics and regenerative potential are often affected by CVD. However, studies presented conflicting results, and directed assessment of MSC parameters relevant to regenerative medicine applications was lacking in many studies. The predictive ability of in vitro assays for in vivo efficacy was rarely assessed. There was no correlation between quality of study reporting and study findings. Age mismatch was also not associated with study findings or effect size. Future research should focus on assays that assess regenerative potential in MSCs and parameters that relate to clinical success.
Collapse
Affiliation(s)
- F C C van Rhijn-Brouwer
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - H Gremmels
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - J O Fledderus
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| | - M C Verhaar
- 1 Department of Nephrology and Hypertension, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
47
|
Duarte D, Hawkins ED, Akinduro O, Ang H, De Filippo K, Kong IY, Haltalli M, Ruivo N, Straszkowski L, Vervoort SJ, McLean C, Weber TS, Khorshed R, Pirillo C, Wei A, Ramasamy SK, Kusumbe AP, Duffy K, Adams RH, Purton LE, Carlin LM, Lo Celso C. Inhibition of Endosteal Vascular Niche Remodeling Rescues Hematopoietic Stem Cell Loss in AML. Cell Stem Cell 2018; 22:64-77.e6. [PMID: 29276143 PMCID: PMC5766835 DOI: 10.1016/j.stem.2017.11.006] [Citation(s) in RCA: 227] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 09/15/2017] [Accepted: 11/06/2017] [Indexed: 02/07/2023]
Abstract
Bone marrow vascular niches sustain hematopoietic stem cells (HSCs) and are drastically remodeled in leukemia to support pathological functions. Acute myeloid leukemia (AML) cells produce angiogenic factors, which likely contribute to this remodeling, but anti-angiogenic therapies do not improve AML patient outcomes. Using intravital microscopy, we found that AML progression leads to differential remodeling of vasculature in central and endosteal bone marrow regions. Endosteal AML cells produce pro-inflammatory and anti-angiogenic cytokines and gradually degrade endosteal endothelium, stromal cells, and osteoblastic cells, whereas central marrow remains vascularized and splenic vascular niches expand. Remodeled endosteal regions have reduced capacity to support non-leukemic HSCs, correlating with loss of normal hematopoiesis. Preserving endosteal endothelium with the small molecule deferoxamine or a genetic approach rescues HSCs loss, promotes chemotherapeutic efficacy, and enhances survival. These findings suggest that preventing degradation of the endosteal vasculature may improve current paradigms for treating AML.
Collapse
Affiliation(s)
- Delfim Duarte
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK; The Francis Crick Institute, WC2A 3LY London, UK.
| | - Edwin D Hawkins
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK; The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Olufolake Akinduro
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Heather Ang
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Katia De Filippo
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, SW7 2AZ London, UK
| | - Isabella Y Kong
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC 3052, Australia; Department of Medical Biology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Myriam Haltalli
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Nicola Ruivo
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Lenny Straszkowski
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia
| | - Stephin J Vervoort
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, VIC 3052, Australia; Peter MacCallum Cancer Centre, Melbourne, VIC 3000, Australia
| | - Catriona McLean
- Department of Haematology, Alfred Hospital, Melbourne, VIC 3004, Australia
| | - Tom S Weber
- Hamilton Institute, Maynooth University, Maynooth, Ireland
| | - Reema Khorshed
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Chiara Pirillo
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK
| | - Andrew Wei
- Department of Haematology, Alfred Hospital, Melbourne, VIC 3004, Australia
| | | | - Anjali P Kusumbe
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Headington, Oxford OX3 7FY, UK
| | - Ken Duffy
- Hamilton Institute, Maynooth University, Maynooth, Ireland
| | - Ralf H Adams
- Max Planck Institute for Molecular Biomedicine, Department of Tissue Morphogenesis, 48149 Munster, Germany; University of Münster, Faculty of Medicine, 48149 Munster, Germany
| | - Louise E Purton
- Stem Cell Regulation Unit, St. Vincent's Institute of Medical Research, Fitzroy, VIC 3065, Australia; Department of Medicine, The University of Melbourne, Fitzroy, VIC 3065, Australia
| | - Leo M Carlin
- Inflammation, Repair and Development, National Heart and Lung Institute, Imperial College London, SW7 2AZ London, UK; Cancer Research UK Beatson Institute, Glasgow G61 1BD, UK
| | - Cristina Lo Celso
- Department of Life Sciences, Sir Alexander Fleming Building, Imperial College London, SW7 2AZ London, UK; The Francis Crick Institute, WC2A 3LY London, UK.
| |
Collapse
|
48
|
Abstract
Bone marrow fat cells comprise the largest population of cells in the bone marrow cavity, a characteristic that has attracted the attention of scholars from different disciplines. The perception that bone marrow adipocytes are "inert space fillers" has been broken, and currently, bone marrow fat is unanimously considered to be the third largest fat depot, after subcutaneous fat and visceral fat. Bone marrow fat (BMF) acts as a metabolically active organ and plays an active role in energy storage, endocrine function, bone metabolism, and the bone metastasis of tumors. Bone marrow adipocytes (BMAs), as a component of the bone marrow microenvironment, influence hematopoiesis through direct contact with cells and the secretion of adipocyte-derived factors. They also influence the progression of hematologic diseases such as leukemia, multiple myeloma, and aplastic anemia, and may be a novel target when exploring treatments for related diseases in the future. Based on currently available data, this review describes the role of BMF in hematopoiesis as well as in the development of hematologic diseases.
Collapse
|
49
|
Bixel MG, Kusumbe AP, Ramasamy SK, Sivaraj KK, Butz S, Vestweber D, Adams RH. Flow Dynamics and HSPC Homing in Bone Marrow Microvessels. Cell Rep 2017; 18:1804-1816. [PMID: 28199850 PMCID: PMC5318670 DOI: 10.1016/j.celrep.2017.01.042] [Citation(s) in RCA: 83] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 11/28/2016] [Accepted: 01/18/2017] [Indexed: 12/17/2022] Open
Abstract
Measurements of flow velocities at the level of individual arterial vessels and sinusoidal capillaries are crucial for understanding the dynamics of hematopoietic stem and progenitor cell homing in the bone marrow vasculature. We have developed two complementary intravital two-photon imaging approaches to determine blood flow dynamics and velocities in multiple vessel segments by capturing the motion of red blood cells. High-resolution spatiotemporal measurements through a cranial window to determine short-time dynamics of flowing blood cells and repetitive centerline scans were used to obtain a detailed flow-profile map with hemodynamic parameters. In addition, we observed the homing of individual hematopoietic stem and progenitor cells and obtained detailed information on their homing behavior. With our imaging setup, we determined flow patterns at cellular resolution, blood flow velocities and wall shear stress in small arterial vessels and highly branched sinusoidal capillaries, and the cellular dynamics of hematopoietic stem and progenitor cell homing.
Collapse
Affiliation(s)
- M Gabriele Bixel
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; Faculty of Medicine, University of Münster, 48149 Münster, Germany.
| | - Anjali P Kusumbe
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Saravana K Ramasamy
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Kishor K Sivaraj
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Stefan Butz
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Dietmar Vestweber
- Department of Vascular Cell Biology, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany
| | - Ralf H Adams
- Department of Tissue Morphogenesis, Max Planck Institute for Molecular Biomedicine, 48149 Münster, Germany; Faculty of Medicine, University of Münster, 48149 Münster, Germany.
| |
Collapse
|
50
|
Abstract
PURPOSE OF REVIEW Hematopoietic stem cells (HSCs) and progenitors are tasked with maintaining hematopoietic homeostasis in the face of numerous insults and challenges, including infection, inflammation, and exsanguination. HSCs possess the remarkable ability to reconstitute the entire hematopoietic system of an organism whose own hematopoietic system has been ablated. This ability is exploited routinely in the clinic via HSC transplantation (HSCT). Here, we focus on the physiological and molecular bottlenecks overcome by HSCs during transplantation. RECENT FINDINGS During transplantation, HSCs encounter a damaged bone marrow niche, characterized molecularly by increases in oxygen concentrations and an altered cytokine milieu. New mechanisms and pathways have been recently implicated during HSCT, including transplanted HSC-dependent secretion of conditioning molecules that facilitate engraftment and pathways that protect HSCs from perturbed organelle homeostasis. SUMMARY Better understanding the molecular processes HSCs employ to withstand the stress of transplant will illuminate novel targets for further improving conditioning regimens and engraftment during HSCT.
Collapse
|