1
|
Dacoba TG, Nabar N, Hammond PT. Modular Layer-by-Layer Nanoparticle Platform for Hematopoietic Progenitor and Stem Cell Targeting. ACS NANO 2025; 19:11333-11347. [PMID: 40080677 DOI: 10.1021/acsnano.5c00388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Effective delivery of drug and gene cargos to hematopoietic stem and progenitor cells (HSPCs) is a major challenge. Current therapeutic strategies in genetic disorders or hematological malignancies are hindered by high costs, low accessibility, and high off-target toxicities. Layer-by-layer nanoparticles (LbL NPs) are modular systems with tunable surface properties to enable highly specific targeting. In this work, we developed LbL NPs that target HSPCs via antibody functionalization with reduced off-target uptake by circulating myeloid cells. NPs layered with poly(acrylic acid), a bioinert polymer, provided more stealth properties in vivo than other tested bioactive polyanions. The additional conjugation of anti-cKit and anti-CD90 antibodies improved NP uptake by 2- to 3-fold in nondifferentiated bone marrow cells in vitro. By contrast, anti-CD105 functionalized NPs showed the highest association to HSPCs in vivo, ranging from 3.0 to 8.5% in progenitor subpopulations. This LbL NP platform was then adapted to target human HSPC receptors, with similar targeting trends in healthy CD34+ human cells. By contrast, anti-CXCR4 functionalization demonstrated the greatest targeting to human B-cell lymphoma and leukemia cells. Taken together, these results underscore the therapeutic potential of this modular LbL NP platform with the capacity to target HSPCs in a disease-dependent context.
Collapse
Affiliation(s)
- Tamara G Dacoba
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Namita Nabar
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
| | - Paula T Hammond
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, United States
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
- Institute for Soldier Nanotechnologies, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
2
|
Xiong M, Xiu Y, Long J, Zhao X, Wang Q, Yang H, Yu H, Bian L, Ju Y, Yin H, Hou Q, Liang F, Liu N, Chen F, Fan R, Sun Y, Zeng Y. Proteomics reveals dynamic metabolic changes in human hematopoietic stem progenitor cells from fetal to adulthood. Stem Cell Res Ther 2024; 15:303. [PMID: 39278906 PMCID: PMC11403967 DOI: 10.1186/s13287-024-03930-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: 07/23/2024] [Accepted: 09/06/2024] [Indexed: 09/18/2024] Open
Abstract
BACKGROUND Hematopoietic stem progenitor cells (HSPCs) undergo phenotypical and functional changes during their emergence and development. Although the molecular programs governing the development of human hematopoietic stem cells (HSCs) have been investigated broadly, the relationships between dynamic metabolic alterations and their functions remain poorly characterized. METHODS In this study, we comprehensively described the proteomics of HSPCs in the human fetal liver (FL), umbilical cord blood (UCB), and adult bone marrow (aBM). The metabolic state of human HSPCs was assessed via a Seahorse assay, RT‒PCR, and flow cytometry-based metabolic-related analysis. To investigate whether perturbing glutathione metabolism affects reactive oxygen species (ROS) production, the metabolic state, and the expansion of human HSPCs, HSPCs were treated with buthionine sulfoximine (BSO), an inhibitor of glutathione synthetase, and N-acetyl-L-cysteine (NAC). RESULTS We investigated the metabolomic landscape of human HSPCs from the fetal, perinatal, and adult developmental stages by in-depth quantitative proteomics and predicted a metabolic switch from the oxidative state to the glycolytic state during human HSPC development. Seahorse assays, mitochondrial activity, ROS level, glucose uptake, and protein synthesis rate analysis supported our findings. In addition, immune-related pathways and antigen presentation were upregulated in UCB or aBM HSPCs, indicating their functional maturation upon development. Glutathione-related metabolic perturbations resulted in distinct responses in human HSPCs and progenitors. Furthermore, the molecular and immunophenotypic differences between human HSPCs at different developmental stages were revealed at the protein level for the first time. CONCLUSION The metabolic landscape of human HSPCs at three developmental stages (FL, UCB, and aBM), combined with proteomics and functional validations, substantially extends our understanding of HSC metabolic regulation. These findings provide valuable resources for understanding human HSC function and development during fetal and adult life.
Collapse
Affiliation(s)
- Mingfang Xiong
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Yanyu Xiu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Juan Long
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Xiao Zhao
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Qianqian Wang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China
| | - Haoyu Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Hang Yu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Lihong Bian
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Yan Ju
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Hongyu Yin
- Department of Gynecology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Qingxiang Hou
- Department of Obstetrics and Gynecology, PLA Rocket Force Characteristic Medical Center, Beijing, 100088, China
| | - Fei Liang
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Nan Liu
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China
| | - Fudong Chen
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China
| | - Ruiwen Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu, 030801, Shanxi, China
| | - Yuying Sun
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
| | - Yang Zeng
- Senior Department of Hematology, the Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100071, China.
- Medical School of the Chinese PLA General Hospital, Beijing, 100039, China.
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, 311399, China.
| |
Collapse
|
3
|
Frenz-Wiessner S, Fairley SD, Buser M, Goek I, Salewskij K, Jonsson G, Illig D, Zu Putlitz B, Petersheim D, Li Y, Chen PH, Kalauz M, Conca R, Sterr M, Geuder J, Mizoguchi Y, Megens RTA, Linder MI, Kotlarz D, Rudelius M, Penninger JM, Marr C, Klein C. Generation of complex bone marrow organoids from human induced pluripotent stem cells. Nat Methods 2024; 21:868-881. [PMID: 38374263 PMCID: PMC11093744 DOI: 10.1038/s41592-024-02172-2] [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: 01/17/2023] [Accepted: 01/09/2024] [Indexed: 02/21/2024]
Abstract
The human bone marrow (BM) niche sustains hematopoiesis throughout life. We present a method for generating complex BM-like organoids (BMOs) from human induced pluripotent stem cells (iPSCs). BMOs consist of key cell types that self-organize into spatially defined three-dimensional structures mimicking cellular, structural and molecular characteristics of the hematopoietic microenvironment. Functional properties of BMOs include the presence of an in vivo-like vascular network, the presence of multipotent mesenchymal stem/progenitor cells, the support of neutrophil differentiation and responsiveness to inflammatory stimuli. Single-cell RNA sequencing revealed a heterocellular composition including the presence of a hematopoietic stem/progenitor (HSPC) cluster expressing genes of fetal HSCs. BMO-derived HSPCs also exhibited lymphoid potential and a subset demonstrated transient engraftment potential upon xenotransplantation in mice. We show that the BMOs could enable the modeling of hematopoietic developmental aspects and inborn errors of hematopoiesis, as shown for human VPS45 deficiency. Thus, iPSC-derived BMOs serve as a physiologically relevant in vitro model of the human BM microenvironment to study hematopoietic development and BM diseases.
Collapse
Affiliation(s)
- Stephanie Frenz-Wiessner
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Savannah D Fairley
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
- Institute of Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
| | - Maximilian Buser
- Institute of AI for Health, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Isabel Goek
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Kirill Salewskij
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - Gustav Jonsson
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Vienna, Austria
| | - David Illig
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Benedicta Zu Putlitz
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Daniel Petersheim
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Yue Li
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Pin-Hsuan Chen
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martina Kalauz
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Raffaele Conca
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Michael Sterr
- Institute of Diabetes and Regeneration Research, Helmholtz Diabetes Center, Helmholtz Center Munich, Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- Technical University of Munich, Munich, Germany
| | - Johanna Geuder
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-University Munich, Martinsried, Germany
| | - Yoko Mizoguchi
- Department of Pediatrics, Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Remco T A Megens
- Institute of Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, Munich, Germany
- Department of Biomedical Engineering (BME), Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht, The Netherlands
- German Center for Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany
| | - Monika I Linder
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Daniel Kotlarz
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Martina Rudelius
- Institute of Pathology, Faculty of Medicine, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Josef M Penninger
- Institute of Molecular Biotechnology of the Austrian Academy of Sciences (IMBA), Vienna BioCenter (VBC), Vienna, Austria
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Helmholtz Centre for Infection Research, Braunschweig, Germany
- Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, Canada
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Zentrum München-German Research Center for Environmental Health, Neuherberg, Germany
| | - Christoph Klein
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-University Munich, Munich, Germany.
- Gene Center, Ludwig-Maximilians-University Munich, Munich, Germany.
| |
Collapse
|
4
|
Sun H, Wang H, Zhang W, Mao H, Li B. Single-cell RNA sequencing reveals resident progenitor and vascularization-associated cell subpopulations in rat annulus fibrosus. J Orthop Translat 2022; 38:256-267. [PMID: 36568849 PMCID: PMC9758498 DOI: 10.1016/j.jot.2022.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Revised: 11/08/2022] [Accepted: 11/15/2022] [Indexed: 12/14/2022] Open
Abstract
Background One of the main causes of low back pain is intervertebral disc degeneration (IDD). Annulus fibrosus (AF) is important for the integrity and functions of the intervertebral disc (IVD). However, the resident functional cell components such as progenitors and vascularization-associated cells in AF are yet to be fully identified. Purpose Identification of functional AF cell subpopulations including resident progenitors and vascularization-associated cells. Methods In this study, the single-cell RNA sequencing data of rat IVDs from a public database were analyzed using Seurat for cell clustering, gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) for functional analysis, StemID for stem cell identification, Monocle and RNA velocity for pseudotime differentiation trajectory validation, single-cell regulatory network inference and clustering (SCENIC) for gene regulatory network (GRN) analysis, and CellChat for cell-cell interaction analysis. Immunostaining on normal and degenerated rat IVDs, as well as human AF, was used for validations. Results From the data analysis, seven AF cell clusters were identified, including two newly discovered functional clusters, the Grem1 + subpopulation and the Lum + subpopulation. The Grem1 + subpopulation had progenitor characteristics, while the Lum + subpopulation was associated with vascularization during IDD. The GRN analysis showed that Sox9 and Id1 were among the key regulators in the Grem1 + subpopulation, and Nr2f2 and Creb5 could be responsible for the vascularization function in the Lum + subpopulation. Cell-cell interaction analysis revealed highly regulated cellular communications between these cells, and multiple signaling networks including PDGF and MIF signaling pathways were involved in the interactions. Conclusions Our results revealed two new functional AF cell subpopulations, with stemness and vascularization induction potential, respectively. The Translational potential of this article These findings complement our knowledge about IVDs, especially the AF, and in return provide potential cell source and regulation targets for IDD treatment and tissue repair. The existence of the cell subpopulations was also validated in human AF, which strengthen the clinical relevance of the findings.
Collapse
Affiliation(s)
- Heng Sun
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Huan Wang
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Weidong Zhang
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Haijiao Mao
- The Affiliated Hospital of Medical School, Ningbo University, Ningbo, Zhejiang, China,Corresponding author.
| | - Bin Li
- Department of Orthopaedic Surgery, Orthopedic Institute, The First Affiliated Hospital, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China,Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China,Corresponding author. 178 Ganjiang Rd, Rm 201 Bldg 18, Soochow University (North Campus), Suzhou, Jiangsu, 215007, China.
| |
Collapse
|
5
|
Tomášková V, Mýtniková A, Hortová Kohoutková M, Mrkva O, Skotáková M, Šitina M, Helánová K, Frič J, Pařenica J, Šrámek V, Helán M. Prognostic value of soluble endoglin in patients with septic shock and severe COVID-19. Front Med (Lausanne) 2022; 9:972040. [PMID: 36117974 PMCID: PMC9470754 DOI: 10.3389/fmed.2022.972040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/15/2022] [Indexed: 12/01/2022] Open
Abstract
Sepsis is a clinical syndrome characterized by a dysregulated response to infection. It represents a leading cause of mortality in ICU patients worldwide. Although sepsis is in the point of interest of research for several decades, its clinical management and patient survival are improving slowly. Monitoring of the biomarkers and their combinations could help in early diagnosis, estimation of prognosis and patient's stratification and response to the treatment. Circulating soluble endoglin (sEng) is the cleaved extracellular part of transmembrane glycoprotein endoglin. As a biomarker, sEng has been tested in several pathologic conditions where its elevation was associated with endothelial dysfunction. In this study we have tested the ability of sEng to predict mortality and its correlation with other clinical characteristics in the cohort of septic shock patients (n = 37) and patients with severe COVID-19 (n = 40). In patients with COVID-19 sEng did not predict mortality or correlate with markers of organ dysfunction. In contrast, in septic shock the level of sEng was significantly higher in patients with early mortality (p = 0.019; AUC = 0.801). Moreover, sEng levels correlated with signs of circulatory failure (required dose of noradrenalin and lactate levels; p = 0.002 and 0.016, respectively). The predominant clinical problem in patients with COVID-19 was ARDS, and although they often showed signs of other organ dysfunction, circulatory failure was exceptional. This potentially explains the difference between sEng levels in COVID-19 and septic shock. In conclusion, we have confirmed that sEng may reflect the extent of the circulatory failure in septic shock patients and thus could be potentially used for the early identification of patients with the highest degree of endothelial dysfunction who would benefit from endothelium-targeted individualized therapy.
Collapse
Affiliation(s)
- Veronika Tomášková
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Alexandra Mýtniková
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | | | - Ondřej Mrkva
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Monika Skotáková
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Michal Šitina
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
| | - Kateřina Helánová
- Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Cardiology, University Hospital Brno, Brno, Czechia
| | - Jan Frič
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
- Department of Modern Immunotherapy Research, Institute of Hematology and Blood Transfusion, Prague, Czechia
| | - Jiří Pařenica
- Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Cardiology, University Hospital Brno, Brno, Czechia
| | - Vladimír Šrámek
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Martin Helán
- Department of Anesthesiology and Intensive Care, St. Anne's University Hospital, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
- International Clinical Research Center, St. Anne's University Hospital, Brno, Czechia
- *Correspondence: Martin Helán
| |
Collapse
|
6
|
Konturek-Ciesla A, Bryder D. Stem Cells, Hematopoiesis and Lineage Tracing: Transplantation-Centric Views and Beyond. Front Cell Dev Biol 2022; 10:903528. [PMID: 35573680 PMCID: PMC9091331 DOI: 10.3389/fcell.2022.903528] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 04/12/2022] [Indexed: 12/26/2022] Open
Abstract
An appropriate production of mature blood cells, or hematopoiesis, is essential for organismal health and homeostasis. In this developmental cascade, hematopoietic stem cells (HSCs) differentiate into intermediate progenitor types, that subsequently give rise to the many distinct blood cell lineages. Here, we describe tools and methods that permit for temporal and native clonal-level HSC lineage tracing in the mouse, and that can now be combined with emerging single-cell molecular analyses. We integrate new insights derived from such experimental paradigms with past knowledge, which has predominantly been derived from transplantation-based approaches. Finally, we outline current knowledge and novel strategies derived from studies aimed to trace human HSC-derived hematopoiesis.
Collapse
|
7
|
Greiner SM, Märklin M, Holzmayer S, Kaban K, Meyer S, Hinterleitner C, Tandler C, Hagelstein I, Jung G, Salih HR, Heitmann JS, Kauer J. Identification of CD105 (endoglin) as novel risk marker in CLL. Ann Hematol 2022; 101:773-780. [PMID: 35044512 PMCID: PMC8913466 DOI: 10.1007/s00277-022-04756-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 01/03/2022] [Indexed: 12/21/2022]
Abstract
Several genetic and clinical markers are established as prognostic factors in chronic lymphocytic leukemia (CLL). However, additional markers are needed for risk stratification. Flow cytometric analysis is a mainstay of CLL diagnostics, thus identification of novel prognostic surface markers can improve risk assessment without increasing burden for patients and physicians. Furthermore, surface molecules preferentially expressed in high-risk cases could serve as therapeutic targets for immunotherapy. CD105 (endoglin) is a TGF-beta coreceptor and activates endothelial cells in healthy tissues and cancer. In addition, it is expressed on healthy hematopoietic precursors as well as lymphoid and myeloid leukemias. In acute myeloid leukemia (AML), a CD105 antibody is successfully applied in clinical studies. In CLL, mRNA expression of the CD105 gene ENG reportedly correlates with other risk factors but failed to show significant correlation with overall survival. However, CD105 protein expression in CLL has never been studied. We here analyzed CD105 surface expression on CLL cells from 71 patients by flow cytometry and report for the first time that substantial levels of CD105 are detectable on CLL cells in 70.4% of patients. Using receiver operating characteristics, we established a cutoff of 5.99% positive cells to distinguish between low and high CD105 levels, the latter correlating with decreased time to first treatment and overall survival. High CD105 expression further correlates with CD38 expression. Our study identified membrane expression of CD105 as a potential risk marker and therapeutic target in high-risk CLL. However, multivariant analyses of large cohorts should be performed in confirmatory studies.
Collapse
Affiliation(s)
- Sarah M Greiner
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Melanie Märklin
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Samuel Holzmayer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Kübra Kaban
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Sophie Meyer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Clemens Hinterleitner
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.,Department of Medical Oncology & Pneumology, University Hospital Tübingen, Tübingen, Germany
| | - Claudia Tandler
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Ilona Hagelstein
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Gundram Jung
- Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.,Interfaculty Institute for Cell Biology, Department of Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, University of Tübingen, Tübingen, Germany
| | - Helmut R Salih
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany
| | - Jonas S Heitmann
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany. .,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.
| | - Joseph Kauer
- Clinical Collaboration Unit Translational Immunology, German Cancer Consortium (DKTK), Department of Internal Medicine, University Hospital Tübingen, Otfried-Müller-Str. 10, 72076, Tübingen, Germany.,Cluster of Excellence iFIT (EXC 2180) "Image-Guided and Functionally Instructed Tumor Therapies", Eberhard Karls University, Tübingen, Germany.,Interfaculty Institute for Cell Biology, Department of Immunology, German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner Site Tübingen, University of Tübingen, Tübingen, Germany.,Department of Internal Medicine V. Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
8
|
Silva WN, Costa AC, Picoli CC, Rocha BGS, Santos GSP, Costa PAC, Azimnasab-sorkhabi P, Soltani-asl M, da Silva RA, Amorim JH, Resende RR, Mintz A, Birbrair A. Hematopoietic stem cell stretches and moves in its bone marrow niche. Crit Rev Oncol Hematol 2021; 163:103368. [PMID: 34051302 PMCID: PMC8277710 DOI: 10.1016/j.critrevonc.2021.103368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 04/28/2021] [Accepted: 05/03/2021] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem cells are the most illustrious inhabitants of the bone marrow. Direct visualization of endogenous hematopoietic stem cells in this niche is essential to study their functions. Until recently this was not possible in live animals. Recent studies, using state-of-the-art technologies, including sophisticated in vivo inducible genetic approaches in combination with two-photon laser scanning microscopy, allow the follow-up of endogenous hematopoietic stem cells' behavior in their habitat. Strikingly, the new findings reveal that quiescent hematopoietic stem cells are more mobile than previously thought, and link their retained steady state within the niche to a mobile behavior. The arising knowledge from this research will be critical for the therapy of several hematological diseases. Here, we review recent progress in our understanding of hematopoietic stem cell biology in their niches.
Collapse
Affiliation(s)
- Walison N. Silva
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alinne C. Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Caroline C. Picoli
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Beatriz G. S. Rocha
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Pedro A. C. Costa
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Maryam Soltani-asl
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | | | - Rodrigo R. Resende
- Center of Biological Sciences and Health, Federal University of West Bahia, Barreiras, BA, Brazil
| | - Akiva Mintz
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Pathology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
- Department of Biochemistry and Immunology, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
9
|
Gautam DK, Chimata AV, Gutti RK, Paddibhatla I. Comparative hematopoiesis and signal transduction in model organisms. J Cell Physiol 2021; 236:5592-5619. [PMID: 33492678 DOI: 10.1002/jcp.30287] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 12/24/2020] [Accepted: 01/08/2021] [Indexed: 12/21/2022]
Abstract
Hematopoiesis is a continuous phenomenon involving the formation of hematopoietic stem cells (HSCs) giving rise to diverse functional blood cells. This developmental process of hematopoiesis is evolutionarily conserved, yet comparably different in various model organisms. Vertebrate HSCs give rise to all types of mature cells of both the myeloid and the lymphoid lineages sequentially colonizing in different anatomical tissues. Signal transduction in HSCs facilitates their potency and specifies branching of lineages. Understanding the hematopoietic signaling pathways is crucial to gain insights into their deregulation in several blood-related disorders. The focus of the review is on hematopoiesis corresponding to different model organisms and pivotal role of indispensable hematopoietic pathways. We summarize and discuss the fundamentals of blood formation in both invertebrate and vertebrates, examining the requirement of key signaling nexus in hematopoiesis. Knowledge obtained from such comparative studies associated with developmental dynamics of hematopoiesis is beneficial to explore the therapeutic options for hematopoietic diseases.
Collapse
Affiliation(s)
- Dushyant Kumar Gautam
- Department of Biochemistry, School of Life Sciences (SLS), University of Hyderabad, Hyderabad, Telangana, India
| | | | - Ravi Kumar Gutti
- Department of Biochemistry, School of Life Sciences (SLS), University of Hyderabad, Hyderabad, Telangana, India
| | - Indira Paddibhatla
- Department of Biochemistry, School of Life Sciences (SLS), University of Hyderabad, Hyderabad, Telangana, India
| |
Collapse
|
10
|
Endoglin: An 'Accessory' Receptor Regulating Blood Cell Development and Inflammation. Int J Mol Sci 2020; 21:ijms21239247. [PMID: 33287465 PMCID: PMC7729465 DOI: 10.3390/ijms21239247] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/01/2020] [Accepted: 12/02/2020] [Indexed: 12/13/2022] Open
Abstract
Transforming growth factor-β1 (TGF-β1) is a pleiotropic factor sensed by most cells. It regulates a broad spectrum of cellular responses including hematopoiesis. In order to process TGF-β1-responses in time and space in an appropriate manner, there is a tight regulation of its signaling at diverse steps. The downstream signaling is mediated by type I and type II receptors and modulated by the ‘accessory’ receptor Endoglin also termed cluster of differentiation 105 (CD105). Endoglin was initially identified on pre-B leukemia cells but has received most attention due to its high expression on activated endothelial cells. In turn, Endoglin has been figured out as the causative factor for diseases associated with vascular dysfunction like hereditary hemorrhagic telangiectasia-1 (HHT-1), pre-eclampsia, and intrauterine growth restriction (IUPR). Because HHT patients often show signs of inflammation at vascular lesions, and loss of Endoglin in the myeloid lineage leads to spontaneous inflammation, it is speculated that Endoglin impacts inflammatory processes. In line, Endoglin is expressed on progenitor/precursor cells during hematopoiesis as well as on mature, differentiated cells of the innate and adaptive immune system. However, so far only pro-monocytes and macrophages have been in the focus of research, although Endoglin has been identified in many other immune system cell subsets. These findings imply a functional role of Endoglin in the maturation and function of immune cells. Aside the functional relevance of Endoglin in endothelial cells, CD105 is differentially expressed during hematopoiesis, arguing for a role of this receptor in the development of individual cell lineages. In addition, Endoglin expression is present on mature immune cells of the innate (i.e., macrophages and mast cells) and the adaptive (i.e., T-cells) immune system, further suggesting Endoglin as a factor that shapes immune responses. In this review, we summarize current knowledge on Endoglin expression and function in hematopoietic precursors and mature hematopoietic cells of different lineages.
Collapse
|
11
|
Heck AM, Ishida T, Hadland B. Location, Location, Location: How Vascular Specialization Influences Hematopoietic Fates During Development. Front Cell Dev Biol 2020; 8:602617. [PMID: 33282876 PMCID: PMC7691428 DOI: 10.3389/fcell.2020.602617] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/30/2020] [Indexed: 01/22/2023] Open
Abstract
During embryonic development, sequential waves of hematopoiesis give rise to blood-forming cells with diverse lineage potentials and self-renewal properties. This process must accomplish two important yet divergent goals: the rapid generation of differentiated blood cells to meet the needs of the developing embryo and the production of a reservoir of hematopoietic stem cells to provide for life-long hematopoiesis in the adult. Vascular beds in distinct anatomical sites of extraembryonic tissues and the embryo proper provide the necessary conditions to support these divergent objectives, suggesting a critical role for specialized vascular niche cells in regulating disparate blood cell fates during development. In this review, we will examine the current understanding of how organ- and stage-specific vascular niche specialization contributes to the development of the hematopoietic system.
Collapse
Affiliation(s)
- Adam M. Heck
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Takashi Ishida
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
| | - Brandon Hadland
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, United States
- Department of Pediatrics, University of Washington School of Medicine, Seattle, WA, United States
| |
Collapse
|
12
|
Solomon M, DeLay M, Reynaud D. Phenotypic Analysis of the Mouse Hematopoietic Hierarchy Using Spectral Cytometry: From Stem Cell Subsets to Early Progenitor Compartments. Cytometry A 2020; 97:1057-1065. [PMID: 32449586 DOI: 10.1002/cyto.a.24041] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/25/2020] [Accepted: 04/29/2020] [Indexed: 12/28/2022]
Abstract
Phenotypic analysis by flow cytometry is one of the most utilized primary tools to study the hematopoietic system. Here, we present a complex panel designed for spectral flow cytometry that allows for the in-depth analysis of the mouse hematopoietic stem and progenitor compartments. The developed panel encompasses the hematopoietic stem cell (HSC) compartment, an array of multipotent progenitors with early marks of lineage specification and a series of progenitors associated with lymphoid, granulo-macrophagic, megakaryocytic and erythroid lineage commitment. It has a built-in redundancy for key markers known to decipher the fine architecture of the HSC compartment by segregating subsets with different functional potential. As a resource, we used this panel to provide a snapshot view of the evolution of these phenotypically defined hematopoietic compartments during the life of the animals. We show that by using a spectral cytometer, this panel is compatible with the analysis of GFP-expressing gene-reporter mice across the hematopoietic system. We leverage this tool to determine how previously described markers such as CD150, CD34, CD105, CD41, ECPR, and CD49b define specific HSC subsets and confirm that high expression of the transcription factor Gfi1 is a hallmark of the most primitive HSC compartment. Altogether, our results provide a convenient protocol to obtain in one analysis a more extensive view of the hematopoietic architecture in mouse models. Our results could also serve as a base for further development of high-end panels leveraging spectral flow cytometry beyond the 15-fluorochrome panel presented in this report. © 2020 International Society for Advancement of Cytometry.
Collapse
Affiliation(s)
- Michael Solomon
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Monica DeLay
- Cytek Biosciences, Fremont, California, 94538, USA
| | - Damien Reynaud
- Stem Cell Program, Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229, USA
| |
Collapse
|
13
|
CD105 (endoglin) as risk marker in AML patients undergoing stem cell transplantation. Int J Hematol 2020; 112:57-64. [PMID: 32266669 DOI: 10.1007/s12185-020-02875-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 03/27/2020] [Accepted: 03/29/2020] [Indexed: 10/24/2022]
Abstract
Several genetic and molecular markers are general predictors of outcome in acute myeloid leukemia (AML), but only few are predictive of outcomes after allogeneic hematopoietic stem cell transplantation (HSCT). Novel markers are needed to improve treatment decisions regarding HSCT. CD105 (endoglin) is a type I transmembrane protein capable of activating endothelial cells. Moreover, CD105 mediates stem cell properties of hematopoietic stem cells and enables repopulation within the bone marrow. Expression of CD105 on vessels of solid tumors and on AML blasts is correlated with poor prognosis. We recently demonstrated that CD105 expression is an independent predictor of overall survival in AML. However, its role in patients receiving intensive treatment with consecutive allogenic transplantation has not been assessed. Using flow cytometry, we analyzed primary samples of 41 patients who underwent HSCT. Using the previously defined SFI cut-off of 5.2, we identified differences in CD105 expression regarding FAB classification and NCCN risk score. Moreover, we detected differences regarding transplant indication and WHO classification with regards to CD105 surface levels. In patients undergoing HSCT high CD105 expression correlated significantly with poor overall survival. We identify CD105 expression in AML as prognostic marker for outcome after HSCT in AML.
Collapse
|
14
|
Schoonderwoerd MJA, Goumans MJTH, Hawinkels LJAC. Endoglin: Beyond the Endothelium. Biomolecules 2020; 10:biom10020289. [PMID: 32059544 PMCID: PMC7072477 DOI: 10.3390/biom10020289] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 02/06/2023] Open
Abstract
Keywords: endoglin; CD105 TGF-β; BMP9; ALK-1; TRC105; tumor microenvironment.
Collapse
Affiliation(s)
- Mark J. A. Schoonderwoerd
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | - Lukas J. A. C. Hawinkels
- Department of Gastrenterology-Hepatology, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
- Correspondence: ; Tel.: +31-71-526-6736
| |
Collapse
|
15
|
Abstract
While several genetic and morphological markers are established and serve to guide therapy of acute myeloid leukaemia (AML), there is still profound need to identify additional markers to better stratify patients. CD105 (Endoglin) is a type I transmembrane protein reported to induce activation and proliferation of endothelial cells. In addition, CD105 is expressed in haematological malignancies and the vessels of solid tumours. Here, CD105 associates with unfavourable disease course, but so far no data are available on the prognostic relevance of CD105 in haematological malignancies. We here generated a novel CD105 antibody for analysis of expression and prognostic relevance of CD105 in a cohort of 62 AML patients. Flow cytometric analysis revealed substantial expression in the various AML FAB types, with FAB M3 type displaying significantly lower surface levels. Next we established a cut-off specific fluorescence level of 5.22 using receiver-operating characteristics, which allowed to group patients in cases with CD105lo and CD105hi surface expression and revealed that high CD105 expression correlated significantly with poor overall and progression free survival. In conclusion, we here identify CD105 expression as a novel prognostic marker in AML, which may serve to optimize follow up and treatment decisions for AML patients.
Collapse
|
16
|
Zhang Y, Clay D, Mitjavila-Garcia MT, Alama A, Mennesson B, Berseneff H, Louache F, Bennaceur-Griscelli A, Oberlin E. VE-Cadherin and ACE Co-Expression Marks Highly Proliferative Hematopoietic Stem Cells in Human Embryonic Liver. Stem Cells Dev 2019; 28:165-185. [PMID: 30426841 DOI: 10.1089/scd.2018.0154] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Despite advances to engineer transplantable hematopoietic stem and progenitor cells (HSPCs) for research and therapy, an in-depth characterization of the developing human hematopoietic system is still lacking. The human embryonic liver is at the crossroad of several hematopoietic sites and harbors a complex hematopoietic hierarchy, including the first actively dividing HSPCs that will further seed the definitive hematopoietic organs. However, few are known about the phenotypic and functional HSPC organization operating at these stages of development. In this study, using a combination of four endothelial and hematopoietic surface markers, that is, the endothelial-specific marker vascular endothelial-cadherin (Cdh5, CD144), the pan-leukocyte antigen CD45, the hemato-endothelial marker CD34, and the angiotensin-converting enzyme (ACE, CD143), we identified distinct HSPC subsets, and among them, a population co-expressing the four markers that uniquely harbored an outstanding proliferation potential both ex vivo and in vivo. Moreover, we traced back this population to the yolk sac (YS) and aorta-gonad-mesonephros (AGM) sites of hematopoietic emergence. Taken together, our data will help to identify human HSPC self-renewal and amplification mechanisms for future cell therapies.
Collapse
Affiliation(s)
- Yanyan Zhang
- 1 Inserm, UMR 1170, Villejuif, France.,2 Paris-Saclay University, Villejuif, France.,3 Gustave Roussy, Villejuif, France
| | - Denis Clay
- 4 Inserm UMS 33, Villejuif, France.,5 André Lwoff Institute (IFR89), Villejuif, France.,6 Paris-Saclay University, Villejuif, France
| | - Maria Teresa Mitjavila-Garcia
- 5 André Lwoff Institute (IFR89), Villejuif, France.,6 Paris-Saclay University, Villejuif, France.,7 Inserm UMR 935, Villejuif, France
| | - Aurélie Alama
- 5 André Lwoff Institute (IFR89), Villejuif, France.,6 Paris-Saclay University, Villejuif, France.,7 Inserm UMR 935, Villejuif, France
| | - Benoit Mennesson
- 8 Obstetrics and Gynecology Department, René-Dubos Hospital, Pontoise, France
| | - Helene Berseneff
- 8 Obstetrics and Gynecology Department, René-Dubos Hospital, Pontoise, France
| | - Fawzia Louache
- 1 Inserm, UMR 1170, Villejuif, France.,2 Paris-Saclay University, Villejuif, France.,3 Gustave Roussy, Villejuif, France
| | - Annelise Bennaceur-Griscelli
- 5 André Lwoff Institute (IFR89), Villejuif, France.,6 Paris-Saclay University, Villejuif, France.,7 Inserm UMR 935, Villejuif, France
| | - Estelle Oberlin
- 5 André Lwoff Institute (IFR89), Villejuif, France.,6 Paris-Saclay University, Villejuif, France.,7 Inserm UMR 935, Villejuif, France
| |
Collapse
|
17
|
Serial transplantation reveals a critical role for endoglin in hematopoietic stem cell quiescence. Blood 2018; 133:688-696. [PMID: 30593445 DOI: 10.1182/blood-2018-09-874677] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 12/21/2018] [Indexed: 12/11/2022] Open
Abstract
Transforming growth factor β (TGF-β) is well known for its important function in hematopoietic stem cell (HSC) quiescence. However, the molecular mechanism underlining this function remains obscure. Endoglin (Eng), a type III receptor for the TGF-β superfamily, has been shown to selectively mark long-term HSCs; however, its necessity in adult HSCs is unknown due to embryonic lethality. Using conditional deletion of Eng combined with serial transplantation, we show that this TGF-β receptor is critical to maintain the HSC pool. Transplantation of Eng-deleted whole bone marrow or purified HSCs into lethally irradiated mice results in a profound engraftment defect in tertiary and quaternary recipients. Cell cycle analysis of primary grafts revealed decreased frequency of HSCs in G0, suggesting that lack of Eng impairs reentry of HSCs to quiescence. Using cytometry by time of flight (CyTOF) to evaluate the activity of signaling pathways in individual HSCs, we find that Eng is required within the Lin-Sca+Kit+-CD48- CD150+ fraction for canonical and noncanonical TGF-β signaling, as indicated by decreased phosphorylation of SMAD2/3 and the p38 MAPK-activated protein kinase 2, respectively. These findings support an essential role for Eng in positively modulating TGF-β signaling to ensure maintenance of HSC quiescence.
Collapse
|
18
|
Puerto-Camacho P, Amaral AT, Lamhamedi-Cherradi SE, Menegaz BA, Castillo-Ecija H, Ordóñez JL, Domínguez S, Jordan-Perez C, Diaz-Martin J, Romero-Pérez L, Lopez-Alvarez M, Civantos-Jubera G, Robles-Frías MJ, Biscuola M, Ferrer C, Mora J, Cuglievan B, Schadler K, Seifert O, Kontermann R, Pfizenmaier K, Simón L, Fabre M, Carcaboso ÁM, Ludwig JA, de Álava E. Preclinical Efficacy of Endoglin-Targeting Antibody-Drug Conjugates for the Treatment of Ewing Sarcoma. Clin Cancer Res 2018; 25:2228-2240. [PMID: 30420447 DOI: 10.1158/1078-0432.ccr-18-0936] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 07/13/2018] [Accepted: 11/06/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE Endoglin (ENG; CD105) is a coreceptor of the TGFβ family that is highly expressed in proliferating endothelial cells. Often coopted by cancer cells, ENG can lead to neo-angiogenesis and vasculogenic mimicry in aggressive malignancies. It exists both as a transmembrane cell surface protein, where it primarily interacts with TGFβ, and as a soluble matricellular protein (sENG) when cleaved by matrix metalloproteinase 14 (MMP14). High ENG expression has been associated with poor prognosis in Ewing sarcoma, an aggressive bone cancer that primarily occurs in adolescents and young adults. However, the therapeutic value of ENG targeting has not been fully explored in this disease. EXPERIMENTAL DESIGN We characterized the expression pattern of transmembrane ENG, sENG, and MMP14 in preclinical and clinical samples. Subsequently, the antineoplastic potential of two novel ENG-targeting monoclonal antibody-drug conjugates (ADC), OMTX503 and OMTX703, which differed only by their drug payload (nigrin-b A chain and cytolysin, respectively), was assessed in cell lines and preclinical animal models of Ewing sarcoma. RESULTS Both ADCs suppressed cell proliferation in proportion to the endogenous levels of ENG observed in vitro. Moreover, the ADCs significantly delayed tumor growth in Ewing sarcoma cell line-derived xenografts and patient-derived xenografts in a dose-dependent manner. CONCLUSIONS Taken together, these studies demonstrate potent preclinical activity of first-in-class anti-ENG ADCs as a nascent strategy to eradicate Ewing sarcoma.
Collapse
Affiliation(s)
- Pilar Puerto-Camacho
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Ana Teresa Amaral
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | | | - Brian A Menegaz
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Helena Castillo-Ecija
- Institut de Recerca Sant Joan de Déu, Pediatric Hematology and Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - José Luis Ordóñez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | | | - Carmen Jordan-Perez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Juan Diaz-Martin
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Laura Romero-Pérez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Maria Lopez-Alvarez
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Gema Civantos-Jubera
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - María José Robles-Frías
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | - Michele Biscuola
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain
| | | | - Jaume Mora
- Institut de Recerca Sant Joan de Déu, Pediatric Hematology and Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - Branko Cuglievan
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | - Keri Schadler
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas
| | | | | | | | | | | | - Ángel M Carcaboso
- Institut de Recerca Sant Joan de Déu, Pediatric Hematology and Oncology, Hospital Sant Joan de Déu Barcelona, Spain
| | - Joseph A Ludwig
- Department of Sarcoma Medical Oncology, MD Anderson Cancer Center, Houston, Texas.
| | - Enrique de Álava
- Institute of Biomedicine of Sevilla (IBiS), Virgen del Rocio University Hospital/CSIC/University of Sevilla/CIBERONC, Seville, Spain.
| |
Collapse
|
19
|
Cosimato V, Scalia G, Raia M, Gentile L, Cerbone V, Visconte F, Statuto T, Valvano L, D'Auria F, Calice G, Graziano D, Musto P, Del Vecchio L. Surface endoglin (CD105) expression on acute leukemia blast cells: an extensive flow cytometry study of 1002 patients. Leuk Lymphoma 2018; 59:2242-2245. [PMID: 29334284 DOI: 10.1080/10428194.2017.1416366] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Affiliation(s)
| | | | | | | | | | | | - Teodora Statuto
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Luciana Valvano
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Fiorella D'Auria
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Giovanni Calice
- b Laboratory of Clinical Research and Advanced Diagnostics , IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Daniela Graziano
- c Antonio Cardarelli Hospital , Unit of Transfusional Medicine , Naples , Italy
| | - Pellegrino Musto
- d Scientific Direction, IRCCS-CROB, Referral Cancer Center of Basilicata , Rionero in Vulture (Pz) , Italy
| | - Luigi Del Vecchio
- a CEINGE Biotecnologie Avanzate , Naples , Italy.,e Department of Molecular Medicine and Medical Biotechnologies (DMMBM) , Federico II University , Naples , Italy
| |
Collapse
|
20
|
Szade K, Zukowska M, Jozkowicz A, Dulak J. Measuring ATP Concentration in a Small Number of Murine Hematopoietic Stem Cells. Methods Mol Biol 2018; 1516:205-217. [PMID: 27138010 DOI: 10.1007/7651_2016_358] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The metabolism of quiescent adult stem cells differs from the metabolism of differentiated cells. The metabolic processes are tightly regulated and their alterations disturb function of stem cells. One of the indicators of metabolic status of cells is the ATP level. While the method of measuring the ATP levels has been known for many years, estimating ATP levels in small population of defined stem cells isolated directly from the tissue has remained challenging. Here, we show our method of measuring the ATP levels in hematopoietic stem cells sorted from murine bone marrow. We used magnetic sorting as well as cell sorter and adopted the commonly used bioluminescence-based detection kits in described protocol. Our strategy allows to measure ATP levels in 1000 highly purified HSC.
Collapse
Affiliation(s)
- Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Monika Zukowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| |
Collapse
|
21
|
Naka K, Hirao A. Regulation of Hematopoiesis and Hematological Disease by TGF-β Family Signaling Molecules. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a027987. [PMID: 28193723 DOI: 10.1101/cshperspect.a027987] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Throughout the lifetime of an individual, hematopoietic stem cells (HSCs) maintain the homeostasis of normal hematopoiesis through the precise generation of mature blood cells. Numerous genetic studies in mice have shown that stem-cell quiescence is critical for sustaining primitive long-term HSCs in vivo. In this review, we first examine the crucial roles of transforming growth factor β (TGF-β) and related signaling molecules in not only regulating the well-known cytostatic effects of these molecules but also governing the self-renewal capacity of HSCs in their in vivo microenvironmental niche. Second, we discuss the current evidence indicating that TGF-β signaling has a dual function in disorders of the hematopoietic system. In particular, we examine the paradox that, although intrinsic TGF-β signaling is essential for regulating the survival and resistance to therapy of chronic myelogenous leukemia (CML) stem cells, genetic changes that abrogate TGF-β signaling can lead to the development of several hematological malignancies.
Collapse
Affiliation(s)
- Kazuhito Naka
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Minami-ku, Hiroshima 734-8553, Japan
| | - Atsushi Hirao
- Division of Molecular Genetics, Cancer Research Institute, Kanazawa University, Kanazawa, Ishikawa 920-1192, Japan
| |
Collapse
|
22
|
Endoglin: a novel target for therapeutic intervention in acute leukemias revealed in xenograft mouse models. Blood 2017; 129:2526-2536. [PMID: 28351936 DOI: 10.1182/blood-2017-01-763581] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 03/20/2017] [Indexed: 12/23/2022] Open
Abstract
Endoglin (CD105), a receptor of the transforming growth factor-β superfamily, has been reported to identify functional long-term repopulating hematopoietic stem cells, and has been detected in certain subtypes of acute leukemias. Whether this receptor plays a functional role in leukemogenesis remains unknown. We identified endoglin expression on the majority of blasts from patients with acute myeloid leukemia (AML) and acute B-lymphoblastic leukemia (B-ALL). Using a xenograft model, we find that CD105+ blasts are endowed with superior leukemogenic activity compared with the CD105- population. We test the effect of targeting this receptor using the monoclonal antibody TRC105, and find that in AML, TRC105 prevented the engraftment of primary AML blasts and inhibited leukemia progression following disease establishment, but in B-ALL, TRC105 alone was ineffective due to the shedding of soluble CD105. However, in both B-ALL and AML, TRC105 synergized with reduced intensity myeloablation to inhibit leukemogenesis, indicating that TRC105 may represent a novel therapeutic option for B-ALL and AML.
Collapse
|
23
|
Nasrallah R, Fast EM, Solaimani P, Knezevic K, Eliades A, Patel R, Thambyrajah R, Unnikrishnan A, Thoms J, Beck D, Vink CS, Smith A, Wong J, Shepherd M, Kent D, Roychoudhuri R, Paul F, Klippert J, Hammes A, Willnow T, Göttgens B, Dzierzak E, Zon LI, Lacaud G, Kouskoff V, Pimanda JE. Identification of novel regulators of developmental hematopoiesis using Endoglin regulatory elements as molecular probes. Blood 2016; 128:1928-1939. [PMID: 27554085 PMCID: PMC5064716 DOI: 10.1182/blood-2016-02-697870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Accepted: 08/15/2016] [Indexed: 12/13/2022] Open
Abstract
Enhancers are the primary determinants of cell identity, and specific promoter/enhancer combinations of Endoglin (ENG) have been shown to target blood and endothelium in the embryo. Here, we generated a series of embryonic stem cell lines, each targeted with reporter constructs driven by specific promoter/enhancer combinations of ENG, to evaluate their discriminative potential and value as molecular probes of the corresponding transcriptome. The Eng promoter (P) in combination with the -8/+7/+9-kb enhancers, targeted cells in FLK1 mesoderm that were enriched for blast colony forming potential, whereas the P/-8-kb enhancer targeted TIE2+/c-KIT+/CD41- endothelial cells that were enriched for hematopoietic potential. These fractions were isolated using reporter expression and their transcriptomes profiled by RNA-seq. There was high concordance between our signatures and those from embryos with defects at corresponding stages of hematopoiesis. Of the 6 genes that were upregulated in both hemogenic mesoderm and hemogenic endothelial fractions targeted by the reporters, LRP2, a multiligand receptor, was the only gene that had not previously been associated with hematopoiesis. We show that LRP2 is indeed involved in definitive hematopoiesis and by doing so validate the use of reporter gene-coupled enhancers as probes to gain insights into transcriptional changes that facilitate cell fate transitions.
Collapse
Affiliation(s)
- Rabab Nasrallah
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Eva M Fast
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA; Howard Hughes Medical Institute, Stem Cell Program and Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Parham Solaimani
- Erasmus Medical Center Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kathy Knezevic
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Alexia Eliades
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Rahima Patel
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Roshana Thambyrajah
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Ashwin Unnikrishnan
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Julie Thoms
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Dominik Beck
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Chris S Vink
- Erasmus Medical Center Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands; Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Aileen Smith
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Jason Wong
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia
| | - Mairi Shepherd
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - David Kent
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Rahul Roychoudhuri
- The Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Fabian Paul
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Julia Klippert
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Annette Hammes
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Thomas Willnow
- Max Delbrueck Center for Molecular Medicine, Berlin, Germany; and
| | - Bertie Göttgens
- Department of Haematology, Wellcome Trust and Medical Research Council Cambridge Stem Cell Institute and Cambridge Institute for Medical Research, Cambridge University, Cambridge, United Kingdom
| | - Elaine Dzierzak
- Erasmus Medical Center Stem Cell Institute, Department of Cell Biology, Erasmus University Medical Center, Rotterdam, The Netherlands; Medical Research Council Centre for Inflammation Research, University of Edinburgh, Edinburgh, United Kingdom
| | - Leonard I Zon
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA; Howard Hughes Medical Institute, Stem Cell Program and Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - George Lacaud
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - Valerie Kouskoff
- Cancer Research United Kingdom Manchester Institute, The University of Manchester, Manchester, United Kingdom
| | - John E Pimanda
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, University of New South Wales Australia, Sydney, Australia; Department of Haematology, The Prince of Wales Hospital, Sydney, Australia
| |
Collapse
|
24
|
Cañete A, Comaills V, Prados I, Castro AM, Hammad S, Ybot-Gonzalez P, Bockamp E, Hengstler JG, Gottgens B, Sánchez MJ. Characterization of a Fetal Liver Cell Population Endowed with Long-Term Multiorgan Endothelial Reconstitution Potential. Stem Cells 2016; 35:507-521. [PMID: 27615355 PMCID: PMC5298023 DOI: 10.1002/stem.2494] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 07/27/2016] [Accepted: 08/10/2016] [Indexed: 12/26/2022]
Abstract
Stable reconstitution of vascular endothelial beds upon transplantation of progenitor cells represents an important challenge due to the paucity and generally limited integration/expansion potential of most identified vascular related cell subsets. We previously showed that mouse fetal liver (FL) hemato/vascular cells from day 12 of gestation (E12), expressing the Stem Cell Leukaemia (SCL) gene enhancer transgene (SCL‐PLAP+ cells), had robust endothelial engraftment potential when transferred to the blood stream of newborns or adult conditioned recipients, compared to the scarce vascular contribution of adult bone marrow cells. However, the specific SCL‐PLAP+ hematopoietic or endothelial cell subset responsible for the long‐term reconstituting endothelial cell (LTR‐EC) activity and its confinement to FL developmental stages remained unknown. Using a busulfan‐treated newborn transplantation model, we show that LTR‐EC activity is restricted to the SCL‐PLAP+VE‐cadherin+CD45− cell population, devoid of hematopoietic reconstitution activity and largely composed by Lyve1+ endothelial‐committed cells. SCL‐PLAP+ Ve‐cadherin+CD45− cells contributed to the liver sinusoidal endothelium and also to the heart, kidney and lung microvasculature. LTR‐EC activity was detected at different stages of FL development, yet marginal activity was identified in the adult liver, revealing unknown functional differences between fetal and adult liver endothelial/endothelial progenitors. Importantly, the observations that expanding donor‐derived vascular grafts colocalize with proliferating hepatocyte‐like cells and participate in the systemic circulation, support their functional integration into young livers. These findings offer new insights into the engraftment, phonotypical, and developmental characterization of a novel endothelial/endothelial progenitor cell subtype with multiorgan LTR‐EC activity, potentially instrumental for the treatment/genetic correction of vascular diseases. Stem Cells2017;35:507–521
Collapse
Affiliation(s)
- Ana Cañete
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Valentine Comaills
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Isabel Prados
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Ana María Castro
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| | - Seddik Hammad
- Faculty of Veterinary Medicine, Department of Forensic Medicine and Veterinary Toxicology, South Valley University, Qena, Egypt.,Leibniz Research Center for Working Environment and Human Factors (IfADo), TU Dortmund University, Dortmund, Germany
| | - Patricia Ybot-Gonzalez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío, CSIC, Universidad de Sevilla, Seville, Spain
| | - Ernesto Bockamp
- Institute of Translational Immunology, University Medical Center, Johannes Gutenberg University, Mainz, Germany
| | - Jan G Hengstler
- Leibniz Research Center for Working Environment and Human Factors (IfADo), TU Dortmund University, Dortmund, Germany
| | - Bertie Gottgens
- Cambridge Institute for Medical Research & Wellcome Trust and MRC Cambridge Stem Cell Institute, Cambridge University, United Kingdom
| | - María José Sánchez
- Centro Andaluz de Biología del Desarrollo (CABD), Consejo Superior de Investigaciones Científicas (CSIC), Junta de Andalucía (JA), Universidad Pablo de Olavide (UPO), Sevilla, Spain
| |
Collapse
|
25
|
Kent DG, Dykstra BJ, Eaves CJ. Isolation and Assessment of Single Long-Term Reconstituting Hematopoietic Stem Cells from Adult Mouse Bone Marrow. CURRENT PROTOCOLS IN STEM CELL BIOLOGY 2016; 38:2A.4.1-2A.4.24. [PMID: 27532815 DOI: 10.1002/cpsc.10] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Hematopoietic stem cells with long-term repopulating activity can now be routinely obtained at purities of 40% to 50% from suspensions of adult mouse bone marrow. Here we describe robust protocols for both their isolation as CD45(+) EPCR(+) CD150(+) CD48(-) (ESLAM) cells using multiparameter cell sorting and for tracking their clonal growth and differentiation activity in irradiated mice transplanted with single ESLAM cells. The simplicity of these procedures makes them attractive for characterizing the molecular and biological properties of individual hematopoietic stem cells with unprecedented power and precision. © 2016 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- David G Kent
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
- Department of Haematology, University of Cambridge, United Kingdom
| | - Brad J Dykstra
- Department of Dermatology, Brigham and Women's Hospital, Boston, Massachusetts
- Harvard Medical School, Boston, Massachusetts
| | - Connie J Eaves
- Terry Fox Laboratory, BC Cancer Agency, Vancouver, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, Canada
| |
Collapse
|
26
|
Rae DT, Hocum JD, Bii V, Deeg HJ, Trobridge GD. A novel retroviral mutagenesis screen identifies prognostic genes in RUNX1 mediated myeloid leukemogenesis. Oncotarget 2016; 6:30664-74. [PMID: 26384344 PMCID: PMC4741560 DOI: 10.18632/oncotarget.5133] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/31/2015] [Indexed: 12/16/2022] Open
Abstract
Using a novel retroviral shuttle vector approach we identified genes that collaborate with a patient derived RUNX1 (AML1) mutant. RUNX1 mutations occurs in 40% of myelodysplastic syndromes (MDS). MDS are a group of hematopoietic stem cell disorders that are characterized by dysplasia that often progress to acute myeloid leukemia (AML). Our goal was to identify genes dysregulated by vector-mediated genotoxicity that may collaborate with the RUNX1 mutant (D171N). D171N expressing cells have a survival and engraftment disadvantage and require additional genetic lesions to survive and persist. By dysregulating genes near the integrated vector provirus, the shuttle vector can promote transformation of D171N cells and tag the nearby genes that collaborate with D171N. In our approach, a gammaretroviral shuttle vector that expresses D171N is used to transduce CD105+, Sca-1+ mouse bone marrow. Mutagenized cells are expanded in liquid culture and vector integration sites from surviving cells are then identified using a retroviral shuttle vector approach. We repeatedly recovered integrated vector proviruses near genes (Itpkb, Ccdc12, and Nbeal2). To assess the prognostic significance of the genes identified we examined differential expression, overall survival, and relapse free survival of AML patients with alteration in the genes identified using The Cancer Genome Atlas (TCGA) AML data set. We found that ITPKB functions as an independent factor for poor prognoses and RUNX1 mutations in conjunction with ITPKB, CCDC12, and NBEAL2 have prognostic potential in AML.
Collapse
Affiliation(s)
- Dustin T Rae
- Washington State University College of Pharmacy, Spokane, WA, USA
| | - Jonah D Hocum
- Washington State University College of Pharmacy, Spokane, WA, USA
| | - Victor Bii
- Washington State University College of Pharmacy, Spokane, WA, USA
| | - H Joachim Deeg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Grant D Trobridge
- Washington State University College of Pharmacy, Spokane, WA, USA.,School of Molecular Biosciences, Washington State University, Pullman, WA, USA
| |
Collapse
|
27
|
Thomas DD, Sommer AG, Balazs AB, Beerman I, Murphy GJ, Rossi D, Mostoslavsky G. Insulin-like growth factor 2 modulates murine hematopoietic stem cell maintenance through upregulation of p57. Exp Hematol 2016; 44:422-433.e1. [PMID: 26872540 DOI: 10.1016/j.exphem.2016.01.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 01/29/2016] [Accepted: 01/31/2016] [Indexed: 01/12/2023]
Abstract
Hematopoietic stem cells (HSC) rely on a highly regulated molecular network to balance self-renewal and lineage specification to sustain life-long hematopoiesis. Despite a plethora of studies aimed at identifying molecules governing HSC fate, our current knowledge of the genes responsible is limited. We have found insulin-like growth factor 2 (IGF2) to be expressed predominantly within long-term HSCs. This study examines IGF2 expression patterns and the effects of the gene in HSCs. Through the overexpression and knockdown of IGF2 within purified HSCs, we report that IGF2 expression increases HSC-derived multilineage colonies in vitro and enhances hematopoietic contribution in vivo on competitive bone marrow transplantation. The effects of IGF2 are mediated by direct upregulation of the CDKi p57, exclusively within long-term HSCs, via activation of the PI3K-Akt pathway. Increased expression of p57 resulted in a concomitant increase in HSCs in the G0/G1 stage of the cell cycle. Analysis of genomic DNA methylation revealed that HSCs exhibited a hypomethylated state within the promoter region of the CDKN1C (p57) gene, providing a potential mechanism for the exclusive effects of IGF2 within HSCs. Our studies indicate a novel role for IGF2 in regulating HSC cell cycle and illustrate potential novel therapeutic targets for hematologic diseases.
Collapse
Affiliation(s)
- Dolly D Thomas
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, MA; Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA
| | - Andreia Gianotti Sommer
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, MA; Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA
| | | | - Isabel Beerman
- Department of Stem Cell and Regenerative Biology, Harvard Medical School, Boston, MA
| | - George J Murphy
- Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA
| | - Derrick Rossi
- Department of Stem Cell and Regenerative Biology, Harvard Medical School, Boston, MA
| | - Gustavo Mostoslavsky
- Section of Gastroenterology, Department of Medicine, Boston University School of Medicine, Boston, MA; Center for Regenerative Medicine (CReM), Boston University and Boston Medical Center, Boston, MA.
| |
Collapse
|
28
|
Tian C, Zhang Y. Purification of hematopoietic stem cells from bone marrow. Ann Hematol 2016; 95:543-7. [DOI: 10.1007/s00277-016-2608-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 01/31/2016] [Indexed: 12/22/2022]
|
29
|
Regulation of Stem Cell Self-Renewal and Oncogenesis by RNA-Binding Proteins. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 907:153-88. [PMID: 27256386 DOI: 10.1007/978-3-319-29073-7_7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Throughout their life span, multicellular organisms rely on stem cell systems. During development pluripotent embryonic stem cells give rise to all cell types that make up the organism. After birth, tissue stem cells maintain properly functioning tissues and organs under homeostasis as well as promote regeneration after tissue damage or injury. Stem cells are capable of self-renewal, which is the ability to divide indefinitely while retaining the potential of differentiation into multiple cell types. The ability to self-renew, however, is a double-edged sword; the molecular mechanisms of self-renewal can be a target of malignant transformation driving tumor development and progression. Growing lines of evidence have shown that RNA-binding proteins (RBPs) play pivotal roles in the regulation of self-renewal by modulating metabolism of coding and non-coding RNAs both in normal tissues and in cancers. In this review, we discuss our current understanding of tissue stem cell systems and how RBPs regulate stem cell fates as well as how the regulatory functions of RBPs contribute to oncogenesis.
Collapse
|
30
|
Abstract
Hematopoietic stem cells (HSC) act as paradigmatic tissue-specific adult stem cells. While they are quiescent in steady-state conditions, they enter the cell cycle and proliferate in stress conditions and during tissue regeneration. Therefore, analysis of cell cycle status of HSC is crucial for understanding their biology. However, due to low number of HSC in tissue and need to use many surface markers for their identification, analysis of their cycle status is technically complicated. Here, we presented our simple strategy to analyze cell cycle of strictly defined LKS CD48(-)CD150(+)CD34(-) HSC, together with Ki67 and DAPI staining by flow cytometry.
Collapse
|
31
|
[Heterogeneity of hematopoietic stem cell]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2015; 36:878-82. [PMID: 26477773 PMCID: PMC7364952 DOI: 10.3760/cma.j.issn.0253-2727.2015.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/14/2022]
|
32
|
Nasrallah R, Knezevic K, Thai T, Thomas SR, Göttgens B, Lacaud G, Kouskoff V, Pimanda JE. Endoglin potentiates nitric oxide synthesis to enhance definitive hematopoiesis. Biol Open 2015; 4:819-29. [PMID: 25979706 PMCID: PMC4571086 DOI: 10.1242/bio.011494] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2014] [Accepted: 04/14/2015] [Indexed: 01/12/2023] Open
Abstract
During embryonic development, hematopoietic cells develop by a process of endothelial-to hematopoietic transition of a specialized population of endothelial cells. These hemogenic endothelium (HE) cells in turn develop from a primitive population of FLK1(+) mesodermal cells. Endoglin (ENG) is an accessory TGF-β receptor that is enriched on the surface of endothelial and hematopoietic stem cells and is also required for the normal development of hemogenic precursors. However, the functional role of ENG during the transition of FLK1(+) mesoderm to hematopoietic cells is ill defined. To address this we used a murine embryonic stem cell model that has been shown to mirror the temporal emergence of these cells in the embryo. We noted that FLK1(+) mesodermal cells expressing ENG generated fewer blast colony-forming cells but had increased hemogenic potential when compared with ENG non-expressing cells. TIE2(+)/CD117(+) HE cells expressing ENG also showed increased hemogenic potential compared with non-expressing cells. To evaluate whether high ENG expression accelerates hematopoiesis, we generated an inducible ENG expressing ES cell line and forced expression in FLK1(+) mesodermal or TIE2(+)/CD117(+) HE cells. High ENG expression at both stages accelerated the emergence of CD45(+) definitive hematopoietic cells. High ENG expression was associated with increased pSMAD2/eNOS expression and NO synthesis in hemogenic precursors. Inhibition of eNOS blunted the ENG induced increase in definitive hematopoiesis. Taken together, these data show that ENG potentiates the emergence of definitive hematopoietic cells by modulating TGF-β/pSMAD2 signalling and increasing eNOS/NO synthesis.
Collapse
Affiliation(s)
- Rabab Nasrallah
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, UNSW Australia, Sydney, NSW 2052, Australia Cancer Research UK Manchester Institute, The University of Manchester, Manchester, M20 4BX, UK
| | - Kathy Knezevic
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, UNSW Australia, Sydney, NSW 2052, Australia
| | - Thuan Thai
- Centre for Vascular Research and School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Shane R Thomas
- Centre for Vascular Research and School of Medical Sciences, UNSW Australia, Sydney, NSW 2052, Australia
| | - Berthold Göttgens
- Department of Haematology, Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 1QR, UK
| | - Georges Lacaud
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, M20 4BX, UK
| | - Valerie Kouskoff
- Cancer Research UK Manchester Institute, The University of Manchester, Manchester, M20 4BX, UK
| | - John E Pimanda
- Lowy Cancer Research Centre and the Prince of Wales Clinical School, UNSW Australia, Sydney, NSW 2052, Australia Department of Haematology, Prince of Wales Hospital, Sydney, NSW 2031, Australia
| |
Collapse
|
33
|
Kays SK, Kaufmann KB, Abel T, Brendel C, Bonig H, Grez M, Buchholz CJ, Kneissl S. CD105 is a surface marker for receptor-targeted gene transfer into human long-term repopulating hematopoietic stem cells. Stem Cells Dev 2015; 24:714-23. [PMID: 25517513 DOI: 10.1089/scd.2014.0455] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are an important target cell population for gene therapy since they can reconstitute the entire hematopoietic system. HSC-enriched cell populations can be recognized based on cell surface marker expression, such as CD34, which is broadly expressed on immature and partially differentiated cells. In mice, co-expression of CD34 and CD105 was previously shown to be relatively more specific for the most immature, long-term repopulating HSCs. Here, we evaluated whether CD105, which is expressed on 30%-80% of CD34(+) cells, is a marker also for human long-term repopulating HSCs. Therefore, we tracked the mature progeny of CD34(+) cells transduced with the CD105-targeted lentiviral vector CD105-LV in xenotolerant mice. Transduction was blocked with soluble CD105 protein confirming specificity. Importantly, CD105-LV transduced human CD34(+) cells engrafted in NOD-scid IL2Rγ(-/-) mice with up to 20% reporter gene-positive cells detected long term in all human hematopoietic lineages in bone marrow (BM), spleen, and blood. In addition, competitive repopulation experiments in mice showed a superior engraftment of CD105-LV transduced CD34(+) cells in BM and spleen compared with cells transduced with a conventional nontargeted lentiviral vector. Thus, human CD34(+)/CD105(+) cells are enriched for early HSCs with high repopulating capacity. Targeting this cell population with CD105-LV offers a novel gene transfer strategy to reach high engraftment rates of transduced cells and highlights the applicability of receptor-targeted vectors to trace cell subsets offering an alternative to prospective isolation by surface markers.
Collapse
Affiliation(s)
- Sarah-Katharina Kays
- 1 Molecular Biotechnology and Gene Therapy, Paul-Ehrlich-Institut , Langen, Germany
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Increase of circulating endothelial cells in patients with Hereditary Hemorrhagic Telangiectasia. Int J Hematol 2014; 101:23-31. [PMID: 25465912 DOI: 10.1007/s12185-014-1698-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2014] [Revised: 10/28/2014] [Accepted: 10/29/2014] [Indexed: 10/24/2022]
Abstract
Hereditary Hemorrhagic Telangiectasia (HHT) is an autosomal dominant disorder characterized by vascular malformations. The genes known to be associated with HHT include ENG (HHT1), ACVRL1 (HHT2) and SMAD4 (JPHT). It has been reported that circulating CD34(+) cell subsets repair damaged vessels. To investigate whether mobilization of these cells is present in the peripheral blood (PB) of HTT patients, we analyzed CD34(+) cells, CD34(+)VEGFR-2(+) progenitor or mature endothelial cells, and CD34(+)CD133(+)VEGFR-2(-) hematopoietic progenitor cells (HPCs). Cytofluorimetric analysis was performed in 150 HTT patients and 43 healthy subjects (CTRLs). In HTT patients, PB CD34(+) cells were significantly increased; the frequency of endothelial cells was higher (P = 0.002), while the frequency of CD34(+)CD133(+)VEGFR-2(-) HPCs was lower (P = 0.00007) than in CTRLs. Results were comparable in patients with ENG or ACVRL1 gene mutation; in patients with ENG mutation, the frequency of the cell subsets inversely correlated with the age of the patients at time of sampling (CD34(+)), disease duration (CD34(+)VEGFR-2(+)), and age at disease onset (CD34(+)CD133(+) VEGFR-2(-)). In conclusion, HHT patients show an increase of circulating endothelial cells and a decrease of HPCs. In patients with ENG mutation, the frequency of CD34(+) endothelial cells correlates with specific clinical characteristics suggesting that their active turnover characterizes the initial phase of the disease.
Collapse
|
35
|
Megakaryocytes maintain homeostatic quiescence and promote post-injury regeneration of hematopoietic stem cells. Nat Med 2014; 20:1321-6. [PMID: 25326798 DOI: 10.1038/nm.3706] [Citation(s) in RCA: 444] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 09/02/2014] [Indexed: 12/15/2022]
Abstract
Multiple bone marrow stromal cell types have been identified as hematopoietic stem cell (HSC)-regulating niche cells. However, whether HSC progeny can serve directly as HSC niche cells has not previously been shown. Here we report a dichotomous role of megakaryocytes (MKs) in both maintaining HSC quiescence during homeostasis and promoting HSC regeneration after chemotherapeutic stress. We show that MKs are physically associated with HSCs in the bone marrow of mice and that MK ablation led to activation of quiescent HSCs and increased HSC proliferation. RNA sequencing (RNA-seq) analysis revealed that transforming growth factor β1 (encoded by Tgfb1) is expressed at higher levels in MKs as compared to other stromal niche cells. MK ablation led to reduced levels of biologically active TGF-β1 protein in the bone marrow and nuclear-localized phosphorylated SMAD2/3 (pSMAD2/3) in HSCs, suggesting that MKs maintain HSC quiescence through TGF-β-SMAD signaling. Indeed, TGF-β1 injection into mice in which MKs had been ablated restored HSC quiescence, and conditional deletion of Tgfb1 in MKs increased HSC activation and proliferation. These data demonstrate that TGF-β1 is a dominant signal emanating from MKs that maintains HSC quiescence. However, under conditions of chemotherapeutic challenge, MK ablation resulted in a severe defect in HSC expansion. In response to stress, fibroblast growth factor 1 (FGF1) signaling from MKs transiently dominates over TGF-β inhibitory signaling to stimulate HSC expansion. Overall, these observations demonstrate that MKs serve as HSC-derived niche cells to dynamically regulate HSC function.
Collapse
|
36
|
Gazit R, Mandal PK, Ebina W, Ben-Zvi A, Nombela-Arrieta C, Silberstein LE, Rossi DJ. Fgd5 identifies hematopoietic stem cells in the murine bone marrow. ACTA ACUST UNITED AC 2014; 211:1315-31. [PMID: 24958848 PMCID: PMC4076584 DOI: 10.1084/jem.20130428] [Citation(s) in RCA: 139] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fdg5 identifies bone marrow cells with potent hematopoietic stem cell activity. Hematopoietic stem cells (HSCs) are the best-characterized tissue-specific stem cells, yet experimental study of HSCs remains challenging, as they are exceedingly rare and methods to purify them are cumbersome. Moreover, genetic tools for specifically investigating HSC biology are lacking. To address this we sought to identify genes uniquely expressed in HSCs within the hematopoietic system and to develop a reporter strain that specifically labels them. Using microarray profiling we identified several genes with HSC-restricted expression. Generation of mice with targeted reporter knock-in/knock-out alleles of one such gene, Fgd5, revealed that though Fgd5 was required for embryonic development, it was not required for definitive hematopoiesis or HSC function. Fgd5 reporter expression near exclusively labeled cells that expressed markers consistent with HSCs. Bone marrow cells isolated based solely on Fgd5 reporter signal showed potent HSC activity that was comparable to stringently purified HSCs. The labeled fraction of the Fgd5 reporter mice contained all HSC activity, and HSC-specific labeling was retained after transplantation. Derivation of next generation mice bearing an Fgd5-CreERT2 allele allowed tamoxifen-inducible deletion of a conditional allele specifically in HSCs. In summary, reporter expression from the Fgd5 locus permits identification and purification of HSCs based on single-color fluorescence.
Collapse
Affiliation(s)
- Roi Gazit
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 Program in Cellular and Molecular Medicine, Division of Hematology/Oncology and Division of Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, MA 02116
| | - Pankaj K Mandal
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 Program in Cellular and Molecular Medicine, Division of Hematology/Oncology and Division of Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, MA 02116
| | - Wataru Ebina
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 Program in Cellular and Molecular Medicine, Division of Hematology/Oncology and Division of Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, MA 02116
| | - Ayal Ben-Zvi
- Department of Pediatrics, Department of Neurobiology, Harvard Medical School, Boston MA 02115
| | - César Nombela-Arrieta
- Program in Cellular and Molecular Medicine, Division of Hematology/Oncology and Division of Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, MA 02116
| | - Leslie E Silberstein
- Program in Cellular and Molecular Medicine, Division of Hematology/Oncology and Division of Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, MA 02116 Program in Cellular and Molecular Medicine, Division of Hematology/Oncology and Division of Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, MA 02116 Harvard Stem Cell Institute, Cambridge, MA 02138
| | - Derrick J Rossi
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 Program in Cellular and Molecular Medicine, Division of Hematology/Oncology and Division of Transfusion Medicine, Department of Laboratory Medicine, Boston Children's Hospital, MA 02116 Department of Pediatrics, Department of Neurobiology, Harvard Medical School, Boston MA 02115 Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
37
|
Grosselin J, Sii-Felice K, Payen E, Chretien S, Tronik-Le Roux D, Leboulch P. Arrayed lentiviral barcoding for quantification analysis of hematopoietic dynamics. Stem Cells 2014; 31:2162-71. [PMID: 23554255 DOI: 10.1002/stem.1383] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/05/2013] [Accepted: 03/13/2013] [Indexed: 11/11/2022]
Abstract
Our understanding of system dynamics of mixed cell populations in whole organisms has benefited from the advent of individual cell marking by nonarrayed DNA barcodes subsequently analyzed by high-throughput DNA sequencing. However, key limitations include statistical biases compromising quantification and the lack of applicability to deconvolute individual cell fate in vivo after pooling single cells differentially exposed to different conditions ex vivo. Here, we have derived an arrayed lentiviral library of DNA barcodes and obtained a proof-of-concept of its resolving capacity by quantifying hematopoietic regeneration after engraftment of mice with genetically modified autologous cells. This method has helped clarify and bridge the seemingly opposed clonal-succession and continuous-recruitment models of hematopoietic stem cell behavior and revealed that myeloid-lymphoid biases are common occurrences in steady-state hematopoiesis. Arrayed lentiviral barcoding should prove a versatile and powerful approach to deconvolute cell dynamics in vivo with applications in hematology, embryology, and cancer biology.
Collapse
Affiliation(s)
- Jeanne Grosselin
- CEA, Institute of Emerging Diseases and Innovative Therapies (iMETI), F-92265 Fontenay-aux-Roses, Paris, France; Inserm U962 and University Paris Sud 11, CEA-iMETI, F-92265 Fontenay-aux-Roses, Paris, France
| | | | | | | | | | | |
Collapse
|
38
|
Borges L, Iacovino M, Koyano-Nakagawa N, Baik J, Garry DJ, Kyba M, Perlingeiro RCR. Expression levels of endoglin distinctively identify hematopoietic and endothelial progeny at different stages of yolk sac hematopoiesis. Stem Cells 2014; 31:1893-901. [PMID: 23712751 DOI: 10.1002/stem.1434] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2012] [Revised: 04/26/2013] [Accepted: 05/02/2013] [Indexed: 11/11/2022]
Abstract
Endoglin (Eng), an ancillary receptor of the transforming growth factor beta (TGFβ) signaling pathway superfamily, has been well recognized for its important function in vascular development and angiogenesis since its discovery more than a decade ago. Recent studies show that this receptor is also critical for the emergence of blood during embryonic development, and that at E7.5, endoglin together with Flk-1 identifies early mesoderm progenitors that are endowed with hematopoietic and endothelial potential. These two lineages emerge in very close association during embryogenesis, and because they share the expression of the same surface markers, it has been difficult to distinguish the earliest hematopoietic from endothelial cells. Here, we evaluated the function of endoglin in hematopoiesis as development progresses past E7.5, and found that the hematopoietic and endothelial progenitors can be distinguished by the levels of endoglin in E9.5 yolk sacs. Whereas endothelial cells are Eng(bright), hematopoietic activity is primarily restricted to a subset of cells that display dim expression of endoglin (Eng(dim)). Molecular characterization of these subfractions showed that endoglin-mediated induction of hematopoiesis occurs in concert with BMP2/BMP4 signaling. This pathway is highly active in Eng(dim) cells but significantly downregulated in the Eng knockout. Taken together, our findings show an important function for endoglin in mediating BMP2/BMP4 signaling during yolk sac hematopoietic development and suggest that the levels of this receptor modulate TGFβ versus bone morphogenetic protein (BMP) signaling.
Collapse
Affiliation(s)
- Luciene Borges
- Department of Medicine, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Ivanovs A, Rybtsov S, Anderson R, Turner M, Medvinsky A. Identification of the niche and phenotype of the first human hematopoietic stem cells. Stem Cell Reports 2014; 2:449-56. [PMID: 24749070 PMCID: PMC3986508 DOI: 10.1016/j.stemcr.2014.02.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 02/13/2014] [Accepted: 02/14/2014] [Indexed: 11/16/2022] Open
Abstract
In various vertebrate species, the dorsal aorta (Ao) is the site of specification of adult hematopoietic stem cells (HSCs). It has been observed that the upregulation of essential hematopoietic transcription factors and the formation of specific intra-aortic hematopoietic cell clusters occur predominantly in the ventral domain of the Ao (AoV). In the mouse, the first HSCs emerge in the AoV. Here, we demonstrate that in the human embryo the first definitive HSCs also emerge asymmetrically and are localized to the AoV, which thus identifies a functional niche for developing human HSCs. Using magnetic cell separation and xenotransplantations, we show that the first human HSCs are CD34(+)VE-cadherin(+)CD45(+)C-KIT(+)THY-1(+)Endoglin(+)RUNX1(+)CD38(-/lo)CD45RA(-). This population harbors practically all committed hematopoietic progenitors and is underrepresented in the dorsal domain of the Ao (AoD) and urogenital ridges (UGRs). The present study provides a foundation for analysis of molecular mechanisms underpinning embryonic specification of human HSCs.
Collapse
Affiliation(s)
- Andrejs Ivanovs
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, Scotland, UK
| | - Stanislav Rybtsov
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, Scotland, UK
| | - Richard A. Anderson
- MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh EH16 4TJ, Scotland, UK
| | - Marc L. Turner
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, Scotland, UK
- Scottish National Blood Transfusion Service, Edinburgh EH17 7QT, Scotland, UK
| | - Alexander Medvinsky
- MRC Centre for Regenerative Medicine, University of Edinburgh, Edinburgh EH16 4UU, Scotland, UK
- Corresponding author
| |
Collapse
|
40
|
Donor hematopoiesis in mice following total lymphoid irradiation requires host T-regulatory cells for durable engraftment. Blood 2014; 123:2882-92. [PMID: 24591203 DOI: 10.1182/blood-2013-10-530212] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Total lymphoid irradiation (TLI) with antithymocyte globulin (ATG) is a unique regimen that prepares recipients for allogeneic hematopoietic cell transplantation by targeting lymph nodes, while sparing large areas of the bone marrow. TLI is reported to increase the frequency of CD4(+)CD25(+)FoxP3(+) T-regulatory cells (Treg) relative to conventional T cells. In this study, barriers to hematopoietic stem cell (HSC) engraftment following this nonmyeloablative conditioning were evaluated. TLI/ATG resulted in profound lymphoablation but endogenous host HSC remained. Initial donor HSC engraftment occurred only in radiation exposed marrow sites, but gradually distributed to bone marrow outside the radiation field. Sustained donor engraftment required host lymphoid cells insofar as lymphocyte deficient Rag2γc(-/-) recipients had unstable engraftment compared with wild-type. TLI/ATG treated wild-type recipients had increased proportions of Treg that were associated with increased HSC frequency and proliferation. In contrast, Rag2γc(-/-) recipients who lacked Treg did not. Adoptive transfer of Treg into Rag2γc(-/-) recipients resulted in increased cell cycling of endogenous HSC. Thus, we hypothesize that Treg influence donor engraftment post-TLI/ATG by increasing HSC cell cycling, thereby promoting the exit of host HSC from the marrow niche. Our study highlights the unique dynamics of donor hematopoiesis following TLI/ATG, and the effect of Treg on HSC activity.
Collapse
|
41
|
Rossi L, Lin KK, Boles NC, Yang L, King KY, Jeong M, Mayle A, Goodell MA. Less is more: unveiling the functional core of hematopoietic stem cells through knockout mice. Cell Stem Cell 2013; 11:302-17. [PMID: 22958929 DOI: 10.1016/j.stem.2012.08.006] [Citation(s) in RCA: 155] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Hematopoietic stem cells (HSCs) represent one of the first recognized somatic stem cell types. As such, nearly 200 genes have been examined for roles in HSC function in knockout mice. In this review, we compile the majority of these reports to provide a broad overview of the functional modules revealed by these genetic analyses and highlight some key regulatory pathways involved, including cell cycle control, Tgf-β signaling, Pten/Akt signaling, Wnt signaling, and cytokine signaling. Finally, we propose recommendations for characterization of HSC function in knockout mice to facilitate cross-study comparisons that would generate a more cohesive picture of HSC biology.
Collapse
Affiliation(s)
- Lara Rossi
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Szade K, Bukowska-Strakova K, Nowak WN, Szade A, Kachamakova-Trojanowska N, Zukowska M, Jozkowicz A, Dulak J. Murine bone marrow Lin⁻Sca⁻1⁺CD45⁻ very small embryonic-like (VSEL) cells are heterogeneous population lacking Oct-4A expression. PLoS One 2013; 8:e63329. [PMID: 23696815 PMCID: PMC3656957 DOI: 10.1371/journal.pone.0063329] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 03/30/2013] [Indexed: 02/06/2023] Open
Abstract
Murine very small embryonic-like (VSEL) cells, defined by the Lin(-)Sca-1(+)CD45(-) phenotype and small size, were described as pluripotent cells and proposed to be the most primitive hematopoietic precursors in adult bone marrow. Although their isolation and potential application rely entirely on flow cytometry, the immunophenotype of VSELs has not been extensively characterized. Our aim was to analyze the possible heterogeneity of Lin(-)Sca(+)CD45(-) population and investigate the extent to which VSELs characteristics may overlap with that of hematopoietic stem cells (HSCs) or endothelial progenitor cells (EPCs). The study evidenced that murine Lin(-)Sca-1(+)CD45(-) population was heterogeneous in terms of c-Kit and KDR expression. Accordingly, the c-Kit(+)KDR(-), c-Kit(-)KDR(+), and c-Kit(-)KDR(-) subpopulations could be distinguished, while c-Kit(+)KDR(+) events were very rare. The c-Kit(+)KDR(-) subset contained almost solely small cells, meeting the size criterion of VSELs, in contrast to relatively bigger c-Kit(-)KDR(+) cells. The c-Kit(-)KDR(-)FSC(low) subset was highly enriched in Annexin V-positive, apoptotic cells, hence omitted from further analysis. Importantly, using qRT-PCR, we evidenced lack of Oct-4A and Oct-4B mRNA expression either in whole adult murine bone marrow or in the sorted of Lin(-)Sca-1(+)CD45(-)FSC(low) population, even by single-cell qRT-PCR. We also found that the Lin(-)Sca-1(+)CD45(-)c-Kit(+) subset did not exhibit hematopoietic potential in a single cell-derived colony in vitro assay, although it comprised the Sca-1(+)c-Kit(+)Lin(-) (SKL) CD34(-)CD45(-)CD105(+) cells, expressing particular HSC markers. Co-culture of Lin(-)Sca-1(+)CD45(-)FSC(low) with OP9 cells did not induce hematopoietic potential. Further investigation revealed that SKL CD45(-)CD105(+) subset consisted of early apoptotic cells with fragmented chromatin, and could be contaminated with nuclei expelled from erythroblasts. Concluding, murine bone marrow Lin(-)Sca-1(+)CD45(-)FSC(low) cells are heterogeneous population, which do not express the pluripotency marker Oct-4A. Despite expression of some hematopoietic markers by a Lin(-)Sca-1(+)CD45(-)c-Kit(+)KDR(-) subset of VSELs, they do not display hematopoietic potential in a clonogenic assay and are enriched in early apoptotic cells.
Collapse
Affiliation(s)
- Krzysztof Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Karolina Bukowska-Strakova
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Witold Norbert Nowak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Agata Szade
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Neli Kachamakova-Trojanowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- Jagiellonian Centre for Experimental Therapeutics, Jagiellonian University, Krakow, Poland
| | - Monika Zukowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- * E-mail: (JD); (AJ)
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
- * E-mail: (JD); (AJ)
| |
Collapse
|
43
|
Abstract
Endoglin (CD105) is a type III auxiliary receptor for the transforming growth factor beta (TGFβ) superfamily. Several lines of evidence suggest that endoglin plays a critical role in maintaining cardiovascular homeostasis. Seemingly disparate disease conditions, including hereditary hemorrhagic telangiectasia, pre-eclampsia, and cardiac fibrosis, have now been associated with endoglin. Given the central role of the TGFβ superfamily in multiple disease conditions, this review provides a detailed update on endoglin as an evolving therapeutic target in the management of cardiovascular disease.
Collapse
Affiliation(s)
- Navin K Kapur
- Molecular Cardiology Research Institute, Tufts Medical Center, Tufts University School of Medicine, Boston, Massachusetts 02111, USA.
| | | | | |
Collapse
|
44
|
Paauwe M, ten Dijke P, Hawinkels LJAC. Endoglin for tumor imaging and targeted cancer therapy. Expert Opin Ther Targets 2013; 17:421-35. [PMID: 23327677 DOI: 10.1517/14728222.2013.758716] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Although cancer treatment has evolved substantially in the past decades, cancer-related mortality rates are still increasing. Therapies targeting tumor angiogenesis, crucial for the growth of solid tumors, mainly target vascular endothelial growth factor (VEGF) and have been clinically applied during the last decade. However, these therapies have not met high expectations, which were based on therapeutic efficacy in animal models. This can partly be explained by the upregulation of alternative angiogenic pathways. Therefore, additional therapies targeting other pro-angiogenic pathways are needed. AREAS COVERED The transforming growth factor (TGF)-β signaling pathway plays an important role in (tumor) angiogenesis. Therefore, components of this pathway are interesting candidates for anti-angiogenic therapy. Endoglin, a co-receptor for various TGF-β family members, is specifically overexpressed in tumor vessels and endoglin expression is associated with metastasis and patient survival. Therefore, endoglin might be a good candidate for anti-angiogenic therapy. In this review, we discuss the potential of using endoglin to target the tumor vasculature for imaging and therapeutic purposes. EXPERT OPINION Considering the promising results from various in vitro studies, in vivo animal models and the first clinical trial targeting endoglin, we are convinced that endoglin is a valuable tool for the diagnosis, visualization and ultimately treatment of solid cancers.
Collapse
Affiliation(s)
- Madelon Paauwe
- Cancer Genomics Centre Netherlands and Centre for BioMedical Genetics, Department of Molecular Cell Biology, Leiden University Medical Center, Building-2, S1-P, PO-box 9600, 2300 RC Leiden, The Netherlands
| | | | | |
Collapse
|
45
|
Mayle A, Luo M, Jeong M, Goodell MA. Flow cytometry analysis of murine hematopoietic stem cells. Cytometry A 2012; 83:27-37. [PMID: 22736515 DOI: 10.1002/cyto.a.22093] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 05/15/2012] [Accepted: 06/03/2012] [Indexed: 12/22/2022]
Abstract
Hematopoietic stem cells (HSCs) remain the most well-characterized adult stem cell population both in terms of markers for purification and assays to assess functional potential. However, despite over 40 years of research, working with HSCs in the mouse remains challenging because of the relative abundance (or lack thereof) of these cells in the bone marrow. The frequency of HSCs in murine bone marrow is about 0.01% of total nucleated cells and ∼5,000 can be isolated from an individual mouse depending on the age, sex, and strain of mice as well as purification scheme utilized. Adding to the challenge is the continual reporting of new markers for HSC purification, which makes it difficult for the uninitiated in the field to know which purification strategies yield the highest proportion of long-term, multilineage HSCs. In this updated version of our review from 2009, we review different strategies for hematopoietic stem and progenitor cell identification and purification. We will also discuss methods for rapid flow cytometric analysis of peripheral blood cell types, and novel strategies for working with rare cell populations such as HSCs in the analysis of cell cycle status by BrdU, Ki-67, and Pyronin Y staining. The purpose of updating this review is to provide insight into some of the recent experimental and technical advances in mouse hematopoietic stem cell biology.
Collapse
Affiliation(s)
- Allison Mayle
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | |
Collapse
|
46
|
Baik J, Borges L, Magli A, Thatava T, Perlingeiro RCR. Effect of endoglin overexpression during embryoid body development. Exp Hematol 2012; 40:837-46. [PMID: 22728030 DOI: 10.1016/j.exphem.2012.06.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Revised: 05/20/2012] [Accepted: 06/12/2012] [Indexed: 10/28/2022]
Abstract
Increasing evidence points to endoglin (Eng), an accessory receptor for the transforming growth factor-β superfamily commonly associated with the endothelial lineage, as an important regulator of the hematopoietic lineage. We have shown that lack of Eng results in reduced numbers of primitive erythroid colonies as well as downregulation of key hematopoietic genes. To determine the effect of Eng overexpression in hematopoietic development, we generated a doxycycline-inducible embryonic stem cell line. Our results demonstrate that induction of Eng during embryoid body differentiation leads to a significant increase in the frequency of hematopoietic progenitors, in particular, the erythroid lineage, which correlated with upregulation of Scl, Gata1, Runx1, and embryonic globin. Interestingly, activation of the hematopoietic program happened at the expense of endothelial and cardiac cells, as differentiation into these mesoderm lineages was compromised. Eng-induced enhanced erythroid activity was accompanied by high levels of Smad1 phosphorylation. This effect was attenuated by addition of a bone morphogenetic protein (BMP) signaling inhibitor to these cultures. Among the BMPs, BMP4 is well known for its role in hematopoietic specification from mesoderm by promoting expression of several hematopoietic genes, including Scl. Because Scl is considered the master regulator of the hematopoietic program, we investigated whether Scl would be capable of rescuing the defective hematopoietic phenotype observed in Eng(-/-) embryonic stem cells. Scl expression in Eng-deficient embryonic stem cells resulted in increased erythroid colony-forming activity and upregulation of Gata1 and Gata2, positioning Eng upstream of Scl. Taken together, these findings support the premise that Eng modulates the hematopoietic transcriptional network, most likely through regulation of BMP4 signaling.
Collapse
Affiliation(s)
- June Baik
- Lillehei Heart Institute, Department of Medicine, University of Minnesota, Minneapolis, MN, USA
| | | | | | | | | |
Collapse
|
47
|
A critical role for endoglin in the emergence of blood during embryonic development. Blood 2012; 119:5417-28. [PMID: 22535663 DOI: 10.1182/blood-2011-11-391896] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Much remains unknown about the signals that induce early mesoderm to initiate hematopoietic differentiation. Here, we show that endoglin (Eng), a receptor for the TGFβ superfamily, identifies all cells with hematopoietic fate in the early embryo. These arise in an Eng(+)Flk1(+) mesodermal precursor population at embryonic day 7.5 (E7.5), a cell fraction also endowed with endothelial potential. In Eng-knockout embryos, hematopoietic colony activity and numbers of CD71(+)Ter119(+) erythroid progenitors were severely reduced. This coincided with severely reduced expression of embryonic globin and key bone morphogenic protein (BMP) target genes, including the hematopoietic regulators Scl, Gata1, Gata2, and Msx-1. To interrogate molecular pathways active in the earliest hematopoietic progenitors, we applied transcriptional profiling to sorted cells from E7.5 embryos. Eng(+)Flk-1(+) progenitors coexpressed TGFβ and BMP receptors and target genes. Furthermore, Eng(+)Flk-1(+) cells presented high levels of phospho-SMAD1/5, indicating active TGFβ and/or BMP signaling. Remarkably, under hematopoietic serum-free culture conditions, hematopoietic outgrowth of Eng-expressing cells was dependent on the TGFβ superfamily ligands BMP4, BMP2, or TGF-β1. These data demonstrate that the E(+)F(+) fraction at E7.5 represents mesodermal cells competent to respond to TGFβ1, BMP4, or BMP2, shaping their hematopoietic development, and that Eng acts as a critical regulator in this process by modulating TGF/BMP signaling.
Collapse
|
48
|
Roques M, Durand C, Gautier R, Canto PY, Petit-Cocault L, Yvernogeau L, Dunon D, Souyri M, Jaffredo T. Endoglin expression level discriminates long-term hematopoietic from short-term clonogenic progenitor cells in the aorta. Haematologica 2012; 97:975-9. [PMID: 22271899 DOI: 10.3324/haematol.2011.046235] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
CD105 is an auxiliary receptor for the transforming growth factor beta superfamily, highly expressed on proliferating endothelial cells and adult hematopoietic stem cells. Because CD105 mRNA expression was reported in the developing aortic region, we further characterized its expression profile in the aorta and examined the hematopoietic potential of CD105(+) cells. Aortic endothelial cells, intra-aortic hematopoietic cell clusters and the purified cell fraction enriched in progenitor/hematopoietic stem cell activity expressed CD105. Aortic hematopoietic short-term clonogenic progenitors were highly enriched in the CD105(intermediate) population whereas more immature long-term progenitors/hematopoietic stem cells are contained within the CD105(high) population. This places CD105 on the short list of molecules discriminating short-term versus long-term progenitors in the aorta. Furthermore, decreasing transforming growth factor beta signaling increases the number of clonogenic progenitors. This suggests that CD105 expression level defines a hierarchy among aortic hematopoietic cells allowing purification of clonogenic versus more immature hematopoietic progenitors, and that the transforming growth factor beta pathway plays a critical role in this process.
Collapse
Affiliation(s)
- Marion Roques
- CNRS UMR7622, Laboratoire de Biologie du Développement, Bat C, 75252 Paris Cedex 05, France
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Purification of adipose tissue mesenchymal stem cells and differentiation toward hepatic-like cells. Methods Mol Biol 2012; 826:61-72. [PMID: 22167640 DOI: 10.1007/978-1-61779-468-1_6] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
There is a great interest in the development of functional hepatocytes in vitro from different types of stem cells. Multipotential mesenchymal stem cells (MSC) compose a great source for stem cell based therapy, especially, because they can be obtain from patients own tissues, sidestepping immunocompatibility and ethical issues. Among MSCs from different sources, adipose-tissue-derived mesenchymal stem cells (AT-MSCs) are very promising because of their high accessibility, proliferation ability, potentiality, and immunocompatibility.AT-MSCs can be easily isolated from stroma vascular fraction (SVF) of adipose tissue. They represent a heterogeneous population of cells. The precise AT-MSCs's marker profile has not been defined yet; therefore, it is still not obvious how to purify these heterogeneous fraction of cells. We postulate that one of the markers defining MSC provenance is CD105 (endoglin).Therefore, we have sorted CD105(+) fraction of AT-MSCs, expanded them, and differentiated toward hepatic-like cells. In order to check their potentiality, we have firstly differentiated sorted CD105(+) AT-MSCs toward mesoderm lineages, using commercialized protocols.We have shown here, that pure CD105(+) AT-MSCs fraction revealed higher homogeneity and differentiation potential toward adipogenic, osteogenic, and chondrogenic lineages and highly inducible into the hepatogenic lineage.Generated (by using our hepatic differentiation protocol) CD105(+) AT-MSCs-derived hepatic-like cells expressed hepatocyte markers, enzymes, and functions.
Collapse
|
50
|
Abstract
Hematopoietic stem and progenitor mobilization has revolutionized the field of hematopoietic transplantation. Currently, hematopoietic grafts acquired from the peripheral blood of patients or donors treated with granulocyte-colony stimulating factor (G-CSF) are the preferred source for transplantation. G-CSF mobilization regimens, however, are associated with known morbidities and a significant number of normal donors and patient populations fail to mobilize sufficient numbers of hematopoietic stem and progenitor cells for transplantation, necessitating the need for non-G-CSF mobilization strategies. Mechanistic studies evaluating hematopoietic bone marrow niche interactions have uncovered novel agents with the capacity for hematopoietic mobilization. This chapter provides a comprehensive overview of mobilizing agents, other than G-CSF, and experimental procedures and technical aspects important to evaluate and define their hematopoietic mobilizing activities alone and in combination.
Collapse
|