1
|
Treichel S, Filippi MD. Linking cell cycle to hematopoietic stem cell fate decisions. Front Cell Dev Biol 2023; 11:1231735. [PMID: 37645247 PMCID: PMC10461445 DOI: 10.3389/fcell.2023.1231735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 07/26/2023] [Indexed: 08/31/2023] Open
Abstract
Hematopoietic stem cells (HSCs) have the properties to self-renew and/or differentiate into any blood cell lineages. In order to balance the maintenance of the stem cell pool with supporting mature blood cell production, the fate decisions to self-renew or to commit to differentiation must be tightly controlled, as dysregulation of this process can lead to bone marrow failure or leukemogenesis. The contribution of the cell cycle to cell fate decisions has been well established in numerous types of stem cells, including pluripotent stem cells. Cell cycle length is an integral component of hematopoietic stem cell fate. Hematopoietic stem cells must remain quiescent to prevent premature replicative exhaustion. Yet, hematopoietic stem cells must be activated into cycle in order to produce daughter cells that will either retain stem cell properties or commit to differentiation. How the cell cycle contributes to hematopoietic stem cell fate decisions is emerging from recent studies. Hematopoietic stem cell functions can be stratified based on cell cycle kinetics and divisional history, suggesting a link between Hematopoietic stem cells activity and cell cycle length. Hematopoietic stem cell fate decisions are also regulated by asymmetric cell divisions and recent studies have implicated metabolic and organelle activity in regulating hematopoietic stem cell fate. In this review, we discuss the current understanding of the mechanisms underlying hematopoietic stem cell fate decisions and how they are linked to the cell cycle.
Collapse
Affiliation(s)
- Sydney Treichel
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH, United States
- University of Cincinnati College of Medicine, Cincinnati, OH, United States
- Molecular and Development Biology Graduate Program, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Marie-Dominique Filippi
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children’s Hospital Research Foundation, Cincinnati, OH, United States
- University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
2
|
Radtke S, Kiem HP. Identification of Nonhuman Primate Hematopoietic Stem and Progenitor Cells. Methods Mol Biol 2023; 2567:87-98. [PMID: 36255696 DOI: 10.1007/978-1-0716-2679-5_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The preclinical development of hematopoietic stem cell (HSC) gene therapy/editing and transplantation protocols is frequently performed in large animal models such as nonhuman primates (NHPs). Similarity in physiology, size, and life expectation as well as cross-reactivity of most reagents and medications allows for the development of treatment strategies with rapid translation to clinical applications. Especially after the adverse events of HSC gene therapy observed in the late 1990s, the ability to perform autologous transplants and follow the animals long-term make the NHP a very attractive model to test the efficiency, feasibility, and safety of new HSC-mediated gene-transfer/editing and transplantation approaches.This protocol describes a method to phenotypically characterize functionally distinct NHP HSPC subsets within specimens or stem cell products from three different NHP species. Procedures are based on the flow-cytometric assessment of cell surface markers that are cross-reactive in between human and NHP to allow for immediate clinical translation. This protocol has been successfully used for the quality control of enriched, cultured, and gene-modified NHP CD34+ hematopoietic stem and progenitor cells (HSPCs) as well as sort-purified CD34 subsets for transplantation in the pig-tailed, cynomolgus, and rhesus macaque. It further allows the longitudinal assessment of primary specimens taken during the long-term follow-up post-transplantation in order to monitor homing, engraftment, and reconstitution of the bone marrow stem cell compartment.
Collapse
Affiliation(s)
- Stefan Radtke
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| | - Hans-Peter Kiem
- Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
- Department of Medicine, University of Washington School of Medicine, Seattle, WA, USA
- Department of Pathology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
3
|
Butner JD, Dogra P, Chung C, Ruiz-Ramírez J, Nizzero S, Plodinec M, Li X, Pan PY, Chen SH, Cristini V, Ozpolat B, Calin GA, Wang Z. Dedifferentiation-mediated stem cell niche maintenance in early-stage ductal carcinoma in situ progression: insights from a multiscale modeling study. Cell Death Dis 2022; 13:485. [PMID: 35597788 PMCID: PMC9124196 DOI: 10.1038/s41419-022-04939-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 12/14/2022]
Abstract
We present a multiscale agent-based model of ductal carcinoma in situ (DCIS) to study how key phenotypic and signaling pathways are involved in the early stages of disease progression. The model includes a phenotypic hierarchy, and key endocrine and paracrine signaling pathways, and simulates cancer ductal growth in a 3D lattice-free domain. In particular, by considering stochastic cell dedifferentiation plasticity, the model allows for study of how dedifferentiation to a more stem-like phenotype plays key roles in the maintenance of cancer stem cell populations and disease progression. Through extensive parameter perturbation studies, we have quantified and ranked how DCIS is sensitive to perturbations in several key mechanisms that are instrumental to early disease development. Our studies reveal that long-term maintenance of multipotent stem-like cell niches within the tumor are dependent on cell dedifferentiation plasticity, and that disease progression will become arrested due to dilution of the multipotent stem-like population in the absence of dedifferentiation. We have identified dedifferentiation rates necessary to maintain biologically relevant multipotent cell populations, and also explored quantitative relationships between dedifferentiation rates and disease progression rates, which may potentially help to optimize the efficacy of emerging anti-cancer stem cell therapeutics.
Collapse
Affiliation(s)
- Joseph D Butner
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Prashant Dogra
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Javier Ruiz-Ramírez
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Sara Nizzero
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Marija Plodinec
- Biozentrum and the Swiss Nanoscience Institute, University of Basel, Basel, 4056, Switzerland
| | - Xiaoxian Li
- Department of Pathology & Laboratory Medicine, Emory University School of medicine, Atlanta, GA, 30322, USA
| | - Ping-Ying Pan
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Shu-Hsia Chen
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA
- Immunotherapy Research Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Vittorio Cristini
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA
- Department of Imaging Physics, University of Texas MD Anderson Cancer Center, Houston, TX, 77230, USA
- Physiology, Biophysics, and Systems Biology Program, Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, 10065, USA
| | - Bulent Ozpolat
- Department of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - George A Calin
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Zhihui Wang
- Mathematics in Medicine Program, Houston Methodist Research Institute, Houston, TX, 77030, USA.
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, NY, 10065, USA.
- Neal Cancer Center, Houston Methodist Research Institute, Houston, TX, 77030, USA.
| |
Collapse
|
4
|
In Vivo Pre-Instructed HSCs Robustly Execute Asymmetric Cell Divisions In Vitro. Int J Mol Sci 2020; 21:ijms21218225. [PMID: 33153113 PMCID: PMC7663432 DOI: 10.3390/ijms21218225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/31/2020] [Accepted: 10/31/2020] [Indexed: 01/12/2023] Open
Abstract
Hematopoietic stem cells (HSCs) are responsible for life-long production of all mature blood cells. Under homeostasis, HSCs in their native bone marrow niches are believed to undergo asymmetric cell divisions (ACDs), with one daughter cell maintaining HSC identity and the other committing to differentiate into various mature blood cell types. Due to the lack of key niche signals, in vitro HSCs differentiate rapidly, making it challenging to capture and study ACD. To overcome this bottleneck, in this study, we used interferon alpha (IFNα) treatment to "pre-instruct" HSC fate directly in their native niche, and then systematically studied the fate of dividing HSCs in vitro at the single cell level via time-lapse analysis, as well as multigene and protein expression analysis. Triggering HSCs' exit from dormancy via IFNα was found to significantly increase the frequency of asynchronous divisions in paired daughter cells (PDCs). Using single-cell gene expression analyses, we identified 12 asymmetrically expressed genes in PDCs. Subsequent immunocytochemistry analysis showed that at least three of the candidates, i.e., Glut1, JAM3 and HK2, were asymmetrically distributed in PDCs. Functional validation of these observations by colony formation assays highlighted the implication of asymmetric distribution of these markers as hallmarks of HSCs, for example, to reliably discriminate committed and self-renewing daughter cells in dividing HSCs. Our data provided evidence for the importance of in vivo instructions in guiding HSC fate, especially ACD, and shed light on putative molecular players involved in this process. Understanding the mechanisms of cell fate decision making should enable the development of improved HSC expansion protocols for therapeutic applications.
Collapse
|
5
|
Butner JD, Fuentes D, Ozpolat B, Calin GA, Zhou X, Lowengrub J, Cristini V, Wang Z. A Multiscale Agent-Based Model of Ductal Carcinoma In Situ. IEEE Trans Biomed Eng 2020; 67:1450-1461. [PMID: 31603768 PMCID: PMC8445608 DOI: 10.1109/tbme.2019.2938485] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
OBJECTIVE we present a multiscale agent-based model of Ductal Carcinoma in Situ (DCIS) in order to gain a detailed understanding of the cell-scale population dynamics, phenotypic distributions, and the associated interplay of important molecular signaling pathways that are involved in DCIS ductal invasion into the duct cavity (a process we refer to as duct advance rate here). METHODS DCIS is modeled mathematically through a hybridized discrete cell-scale model and a continuum molecular scale model, which are explicitly linked through a bidirectional feedback mechanism. RESULTS we find that duct advance rates occur in two distinct phases, characterized by an early exponential population expansion, followed by a long-term steady linear phase of population expansion, a result that is consistent with other modeling work. We further found that the rates were influenced most strongly by endocrine and paracrine signaling intensity, as well as by the effects of cell density induced quiescence within the DCIS population. CONCLUSION our model analysis identified a complex interplay between phenotypic diversity that may provide a tumor adaptation mechanism to overcome proliferation limiting conditions, allowing for dynamic shifts in phenotypic populations in response to variation in molecular signaling intensity. Further, sensitivity analysis determined DCIS axial advance rates and calcification rates were most sensitive to cell cycle time variation. SIGNIFICANCE this model may serve as a useful tool to study the cell-scale dynamics involved in DCIS initiation and intraductal invasion, and may provide insights into promising areas of future experimental research.
Collapse
|
6
|
Kordelas L, Görgens A, Radtke S, Horn PA, Beelen DW, Giebel B. Allogeneic transplantation of peripheral blood stem cell grafts results in a massive decrease of primitive hematopoietic progenitor frequencies in reconstituted bone marrows. Bone Marrow Transplant 2019; 55:100-109. [PMID: 31435034 DOI: 10.1038/s41409-019-0645-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 06/25/2019] [Accepted: 06/28/2019] [Indexed: 01/22/2023]
Abstract
The success of allogeneic hematopoietic stem cell transplantation (alloSCT) is indicated by the reconstitution of the peripheral blood system of patients after alloSCT and the engraftment of hematopoietic stem and progenitor cells (HSPCs) into their bone marrow (BM). The number of CD34+ cells is commonly used as surrogate for the content of hematopoietic stem cells in the grafts. During the last decade, several antigens (including CD133, CD45RA, CD38, and CD10) were identified allowing discrimination of different HSPC subpopulations within the human CD34+ cell compartment. Although such studies increased our understanding of early human hematopoiesis tremendously, hardly any study dissected the CD34+ compartment in the alloSCT setting. Consequently, we comprehensively analyzed the CD34+ compartment in G-CSF-stimulated peripheral blood stem cell grafts of allogeneic donors, in BM samples of the respective recipients 4 weeks after alloSCT, and in BM samples of healthy donors. We demonstrate that alloSCT is associated with a dramatic shift from primitive to more mature HSPC types. Upon investigating whether the composition of engrafted CD34+ cells has any impact on the incidence and severity of graft-versus-host disease, we did not find any correlation. However, more detailed analyses of the CD34+ compartment may elucidate associations with other transplantation-related complications.
Collapse
Affiliation(s)
- Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| | - André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Clinical Research Center, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Stefan Radtke
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,Stem Cell and Gene Therapy Program, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Dietrich W Beelen
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.
| |
Collapse
|
7
|
A hybrid agent-based model of the developing mammary terminal end bud. J Theor Biol 2016; 407:259-270. [PMID: 27475843 DOI: 10.1016/j.jtbi.2016.07.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 07/15/2016] [Accepted: 07/25/2016] [Indexed: 12/13/2022]
Abstract
Mammary gland ductal elongation is spearheaded by terminal end buds (TEBs), where populations of highly proliferative cells are maintained throughout post-pubertal organogenesis in virgin mice until the mammary fat pad is filled by a mature ductal tree. We have developed a hybrid multiscale agent-based model to study how cellular differentiation pathways, cellular proliferation capacity, and endocrine and paracrine signaling play a role during development of the mammary gland. A simplified cellular phenotypic hierarchy that includes stem, progenitor, and fully differentiated cells within the TEB was implemented. Model analysis finds that mammary gland development was highly sensitive to proliferation events within the TEB, with progenitors likely undergoing 2-3 proliferation cycles before transitioning to a non-proliferative phenotype, and this result is in agreement with our previous experimental work. Endocrine and paracrine signaling were found to provide reliable ductal elongation rate regulation, while variations in the probability a new daughter cell will be of a proliferative phenotype were seen to have minimal effects on ductal elongation rates. Moreover, the distribution of cellular phenotypes within the TEB was highly heterogeneous, demonstrating significant allowable plasticity in possible phenotypic distributions while maintaining biologically relevant growth behavior. Finally, simulation results indicate ductal elongation rates due to cellular proliferation within the TEB may have a greater sensitivity to upstream endocrine signaling than endothelial to stromal paracrine signaling within the TEB. This model provides a useful tool to gain quantitative insights into cellular population dynamics and the effects of endocrine and paracrine signaling within the pubertal terminal end bud.
Collapse
|
8
|
Gur-Cohen S, Kollet O, Graf C, Esmon CT, Ruf W, Lapidot T. Regulation of long-term repopulating hematopoietic stem cells by EPCR/PAR1 signaling. Ann N Y Acad Sci 2016; 1370:65-81. [PMID: 26928241 DOI: 10.1111/nyas.13013] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 01/05/2016] [Accepted: 01/11/2016] [Indexed: 01/18/2023]
Abstract
The common developmental origin of endothelial and hematopoietic cells is manifested by coexpression of several cell surface receptors. Adult murine bone marrow (BM) long-term repopulating hematopoietic stem cells (LT-HSCs), endowed with the highest repopulation and self-renewal potential, express endothelial protein C receptor (EPCR), which is used as a marker to isolate them. EPCR/protease-activated receptor-1 (PAR1) signaling in endothelial cells has anticoagulant and anti-inflammatory roles, while thrombin/PAR1 signaling induces coagulation and inflammation. Recent studies define two new PAR1-mediated signaling cascades that regulate EPCR(+) LT-HSC BM retention and egress. EPCR/PAR1 signaling facilitates LT-HSC BM repopulation, retention, survival, and chemotherapy resistance by restricting nitric oxide (NO) production, maintaining NO(low) LT-HSC BM retention with increased VLA4 expression, affinity, and adhesion. Conversely, acute stress and clinical mobilization upregulate thrombin generation and activate different PAR1 signaling that overcomes BM EPCR(+) LT-HSC retention, inducing their recruitment to the bloodstream. Thrombin/PAR1 signaling induces NO generation, TACE-mediated EPCR shedding, and upregulation of CXCR4 and PAR1, leading to CXCL12-mediated stem and progenitor cell mobilization. This review discusses new roles for factors traditionally viewed as coagulation related, which independently act in the BM to regulate PAR1 signaling in bone- and blood-forming progenitor cells, navigating their fate by controlling NO production.
Collapse
Affiliation(s)
- Shiri Gur-Cohen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Orit Kollet
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Claudine Graf
- Center for Thrombosis and Hemostasis and Johannes Gutenberg University Medical Center, Mainz, Germany.,Third Medical Department, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Charles T Esmon
- Coagulation Biology Laboratory, Oklahoma Medical Research Foundation and Departments of Pathology and Biochemistry & Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Wolfram Ruf
- Center for Thrombosis and Hemostasis and Johannes Gutenberg University Medical Center, Mainz, Germany.,Department of Immunology and Microbial Science, the Scripps Research Institute, La Jolla, California
| | - Tsvee Lapidot
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
9
|
|
10
|
|
11
|
Abstract
Drug resistance is a fundamental problem in the treatment of cancer since cancer that becomes resistant to the available drugs may leave the patient with no therapeutic alternatives. In this chapter, we consider the dynamics of drug resistance in blood cancer and the related issue of the dynamics of cancer stem cells. After describing the main types of chemotherapeutic agents available for cancer treatment, we review the different mechanisms of drug resistance development. Various mathematical models of drug resistance found in the literature are then reviewed. Given the well-known hierarchy of the hematopoietic system, it is critical to focus on those cells that have the ability to self-renew, since these will be the only cells able to induce long-term drug resistance. Thus, a recent mathematical model taking into account the complex dynamics of the leukemic stem-like cells is described. The chapter closes with a few applications of this model to chronic myeloid leukemia.
Collapse
Affiliation(s)
- Cristian Tomasetti
- Johns Hopkins School of Medicine, 550 North Broadway, Suite 1103, Baltimore, MD 21205, USA,
| |
Collapse
|
12
|
Radtke S, Görgens A, Kordelas L, Schmidt M, Kimmig KR, Köninger A, Horn PA, Giebel B. CD133 allows elaborated discrimination and quantification of haematopoietic progenitor subsets in human haematopoietic stem cell transplants. Br J Haematol 2015; 169:868-78. [PMID: 25819405 DOI: 10.1111/bjh.13362] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 01/26/2015] [Indexed: 12/12/2022]
Abstract
The success of haematopoietic stem cell (HSC) transplantation largely depends on numbers of transplanted HSCs, which reside in the CD34(+) populations of bone marrow (BM), peripheral blood stem cells (PBSC) and umbilical cord blood (UCB). More specifically HSCs reside in the CD38(low/-) subpopulation, which cannot be objectively discriminated from mature CD34(+) CD38(+) progenitors. Thus, better marker combinations for the quantification of more primitive haematopoietic stem and progenitor cells in transplants are required. Recently, by combining CD34 and CD133 we could clearly distinguish CD133(+) CD34(+) multipotent and lympho-myeloid from CD133(low) CD34(+) erythro-myeloid progenitors in UCB samples. To qualify the assessment of CD133 for routine quality control of adult HSC sources, we analysed the developmental potentials of CD133(+) and CD133(low) subpopulations in BM and PBSC. Similar to UCB, CD133 expression objectively discriminated functionally distinct subpopulations in adult HSC sources. By implementing anti-CD45RA staining, which separates multipotent (CD133(+) CD34(+) CD45RA(-) ) from lympho-myeloid (CD133(+) CD34(+) CD45RA(+) ) progenitor fractions, UCB was found to contain 2-3 times higher multipotent progenitor frequencies than BM and PBSC. To test for the consistency of CD133 expression, we compared CD133(+) CD34(+) contents of 128 UCB samples with maternal and obstetrical factors and obtained similar correlations to related studies focusing on CD34(+) cell contents. In conclusion, implementation of anti-CD133 staining into existing routine panels will improve the quality control analyses for HSC transplants.
Collapse
Affiliation(s)
- Stefan Radtke
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Markus Schmidt
- Department of Gynaecology and Obstetrics, Hospital Duisburg, Duisburg, Germany
| | - Klaus R Kimmig
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Angela Köninger
- Department of Gynecology and Obstetrics, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany
| |
Collapse
|
13
|
Görgens A, Ludwig AK, Möllmann M, Krawczyk A, Dürig J, Hanenberg H, Horn PA, Giebel B. Multipotent hematopoietic progenitors divide asymmetrically to create progenitors of the lymphomyeloid and erythromyeloid lineages. Stem Cell Reports 2014; 3:1058-72. [PMID: 25448068 PMCID: PMC4263999 DOI: 10.1016/j.stemcr.2014.09.016] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 09/23/2014] [Accepted: 09/24/2014] [Indexed: 01/21/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) can self-renew and create committed progenitors, a process supposed to involve asymmetric cell divisions (ACDs). Previously, we had linked the kinetics of CD133 expression with ACDs but failed to detect asymmetric segregation of classical CD133 epitopes on fixed, mitotic HSPCs. Now, by using a novel anti-CD133 antibody (HC7), we confirmed the occurrence of asymmetric CD133 segregation on paraformaldehyde-fixed and living HSPCs. After showing that HC7 binding does not recognizably affect biological features of human HSPCs, we studied ACDs in different HSPC subtypes and determined the developmental potential of arising daughter cells at the single-cell level. Approximately 70% of the HSPCs of the multipotent progenitor (MPP) fraction studied performed ACDs, and about 25% generated lymphoid-primed multipotent progenitor (LMPP) as wells as erythromyeloid progenitor (EMP) daughter cells. Since MPPs hardly created daughter cells maintaining MPP characteristics, our data suggest that under conventional culture conditions, ACDs are lineage instructive rather than self-renewing. The HC7 anti-CD133 antibody allows analyses of ACDs on fixed human HSPCs HC7 and AC133 anti-CD133 antibodies allow tracking of CD133 in living HSPCs Cells of the MPP fraction divide asymmetrically to create LMPP- and EMP-like cells ACDs of MPPs are lineage instructive rather than self-renewing
Collapse
Affiliation(s)
- André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; German Cancer Consortium (DKTK).
| | - Anna-Kristin Ludwig
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany
| | - Michael Möllmann
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany
| | - Adalbert Krawczyk
- Institute of Virology, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany
| | - Jan Dürig
- Department of Hematology, University Hospital Essen, University of Duisburg-Essen, Hufelandstraße 55, 45122 Essen, Germany
| | - Helmut Hanenberg
- Riley Hospital for Children, Indiana University School of Medicine, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA
| | - Peter A Horn
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; German Cancer Consortium (DKTK)
| | - Bernd Giebel
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstraße 179, 45147 Essen, Germany; German Cancer Consortium (DKTK).
| |
Collapse
|
14
|
Chaurasia P, Gajzer DC, Schaniel C, D'Souza S, Hoffman R. Epigenetic reprogramming induces the expansion of cord blood stem cells. J Clin Invest 2014; 124:2378-95. [PMID: 24762436 DOI: 10.1172/jci70313] [Citation(s) in RCA: 124] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Cord blood (CB) cells that express CD34 have extensive hematopoietic capacity and rapidly divide ex vivo in the presence of cytokine combinations; however, many of these CB CD34+ cells lose their marrow-repopulating potential. To overcome this decline in function, we treated dividing CB CD34+ cells ex vivo with several histone deacetylase inhibitors (HDACIs). Treatment of CB CD34+ cells with the most active HDACI, valproic acid (VPA), following an initial 16-hour cytokine priming, increased the number of multipotent cells (CD34+CD90+) generated; however, the degree of expansion was substantially greater in the presence of both VPA and cytokines for a full 7 days. Treated CD34+ cells were characterized based on the upregulation of pluripotency genes, increased aldehyde dehydrogenase activity, and enhanced expression of CD90, c-Kit (CD117), integrin α6 (CD49f), and CXCR4 (CD184). Furthermore, siRNA-mediated inhibition of pluripotency gene expression reduced the generation of CD34+CD90+ cells by 89%. Compared with CB CD34+ cells, VPA-treated CD34+ cells produced a greater number of SCID-repopulating cells and established multilineage hematopoiesis in primary and secondary immune-deficient recipient mice. These data indicate that dividing CB CD34+ cells can be epigenetically reprogrammed by treatment with VPA so as to generate greater numbers of functional CB stem cells for use as transplantation grafts.
Collapse
|
15
|
Görgens A, Radtke S, Horn PA, Giebel B. New relationships of human hematopoietic lineages facilitate detection of multipotent hematopoietic stem and progenitor cells. Cell Cycle 2013; 12:3478-82. [PMID: 24189527 DOI: 10.4161/cc.26900] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Three important goals of hematopoietic stem cell research are to understand of how hematopoietic stem cells (HSCs) self-renew, how lineage commitment takes place, and how HSCs can be expanded ex vivo. Research in this area requires a reliable model of hematopoiesis. Performing detailed functional analyses of human hematopoietic progenitor subsets, we recently gained evidence for new hematopoietic lineage relationships. (1) According to our data, neutrophils belong to the same branch of the hematopoietic tree as lymphocytes. In contrast, eosinophils and basophils derive from another branch, the erythro-myeloid branch. Here, after introducing the newly proposed hematopoietic model, we discuss its consequences for the identification and expansion of human multipotent progenitors and suggest a fast and reliable method to screen for multipotent hematopoietic cells in vitro.
Collapse
Affiliation(s)
- André Görgens
- Institute for Transfusion Medicine; University Hospital Essen; University of Duisburg-Essen; Essen, Germany
| | | | | | | |
Collapse
|
16
|
Shin JW, Swift J, Ivanovska I, Spinler KR, Buxboim A, Discher DE. Mechanobiology of bone marrow stem cells: from myosin-II forces to compliance of matrix and nucleus in cell forms and fates. Differentiation 2013; 86:77-86. [PMID: 23790394 PMCID: PMC3964600 DOI: 10.1016/j.diff.2013.05.001] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 05/01/2013] [Accepted: 05/02/2013] [Indexed: 12/22/2022]
Abstract
Adult stem cells and progenitors are of great interest for their clinical application as well as their potential to reveal deep sensitivities to microenvironmental factors. The bone marrow is a niche for at least two types of stem cells, and the prototype is the hematopoietic stem cell/progenitors (HSC/Ps), which have saved many thousands of patients for several decades now. In bone marrow, HSC/Ps interact functionally with marrow stromal cells that are often referred to as mesenchymal stem cells (MSCs) or derivatives thereof. Myosin and matrix elasticity greatly affect MSC function, and these mechanobiological factors are now being explored with HSC/Ps both in vitro and in vivo. Also emerging is a role for the nucleus as a mechanically sensitive organelle that is semi-permeable to transcription factors which are modified for nuclear entry by cytoplasmic mechanobiological pathways. Since therapies envisioned with induced pluripotent stem cells and embryonic stem cells generally involve in vitro commitment to an adult stem cell or progenitor, a very deep understanding of stem cell mechanobiology is essential to progress with these multi-potent cells.
Collapse
Affiliation(s)
- Jae-Won Shin
- Molecular and Cell Biophysics Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
17
|
Görgens A, Radtke S, Möllmann M, Cross M, Dürig J, Horn PA, Giebel B. Revision of the human hematopoietic tree: granulocyte subtypes derive from distinct hematopoietic lineages. Cell Rep 2013; 3:1539-52. [PMID: 23707063 DOI: 10.1016/j.celrep.2013.04.025] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2012] [Revised: 03/18/2013] [Accepted: 04/25/2013] [Indexed: 12/11/2022] Open
Abstract
The classical model of hematopoiesis predicts a dichotomous lineage restriction of multipotent hematopoietic progenitors (MPPs) into common lymphoid progenitors (CLPs) and common myeloid progenitors (CMPs). However, this idea has been challenged by the identification of lymphoid progenitors retaining partial myeloid potential (e.g., LMPPs), implying that granulocytes can arise within both the classical lymphoid and the myeloid branches. Here, we resolve this issue by using cell-surface CD133 expression to discriminate functional progenitor populations. We show that eosinophilic and basophilic granulocytes as well as erythrocytes and megakaryocytes derive from a common erythro-myeloid progenitor (EMP), whereas neutrophilic granulocytes arise independently within a lympho-myeloid branch with long-term progenitor function. These findings challenge the concept of a CMP and restore dichotomy to the classical hematopoietic model.
Collapse
Affiliation(s)
- André Görgens
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, Virchowstr. 179, 45147 Essen, Germany
| | | | | | | | | | | | | |
Collapse
|
18
|
Marciniak-Czochra A, Stiehl T. Mathematical Models of Hematopoietic Reconstitution After Stem Cell Transplantation. CONTRIBUTIONS IN MATHEMATICAL AND COMPUTATIONAL SCIENCES 2013. [DOI: 10.1007/978-3-642-30367-8_9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
19
|
|
20
|
Nteliopoulos G, Gordon MY. Protein segregation between dividing hematopoietic progenitor cells in the determination of the symmetry/asymmetry of cell division. Stem Cells Dev 2012; 21:2565-80. [PMID: 22455336 DOI: 10.1089/scd.2011.0467] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
In the present study, we investigated how the symmetry/asymmetry of cell division in mitotic CD34(+) cells can be evaluated by determining the plane of cell division and the potential distribution of proteins between daughter cells. The orientation of the mitotic spindle is dependent upon the positioning of the centrosomes, which determine the plane of cell division and the sharing of proteins. If the functions of unequally shared proteins are relevant to the kinetics of cell division, they could determine whether the daughter cells undergo self-renewal or differentiation. The kinetic function of the proteins of interest was investigated using a colony-replating assay and carboxyfluorescein succinimidyl ester (CFSE) staining. We used Notch/Numb as a model system, since they have a role in balancing symmetric/asymmetric divisions. Mitotic cells were examined microscopically and centrosomal markers γ-tubulin/pericentrin were used with activated Notch-1 and Numb. We monitored the first crucial divisions by CFSE staining and found an inverse relationship between activated Notch and Numb expression, suggesting a reciprocal regulation. We suggest that the subpopulations expressing activated Notch or Numb have different cell fates. To determine the influence of Notch signaling on progenitor cell self-renewal, we used the γ-secretase inhibitor N-[N-(3,5-Difluorophenacetyl-L-alanyl)]-S-phenylglycine t-Butyl ester (DAPT). DAPT influences self-renewal/differentiation outcome by affecting the frequency of symmetric renewal divisions without affecting the rate of divisions. Overall, the purpose of this study was to establish a cellular system for predicting the symmetry/asymmetry of hematopoietic progenitor divisions at the level of centrosomes and protein distribution and to investigate the influence of these proteins on progenitor cell kinetics.
Collapse
Affiliation(s)
- Georgios Nteliopoulos
- Department of Haematology, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, London, United Kingdom.
| | | |
Collapse
|
21
|
Asymmetric segregation and self-renewal of hematopoietic stem and progenitor cells with endocytic Ap2a2. Blood 2012; 119:2510-22. [DOI: 10.1182/blood-2011-11-393272] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Abstract
The stem cell–intrinsic model of self-renewal via asymmetric cell division (ACD) posits that fate determinants be partitioned unequally between daughter cells to either activate or suppress the stemness state. ACD is a purported mechanism by which hematopoietic stem cells (HSCs) self-renew, but definitive evidence for this cellular process remains open to conjecture. To address this issue, we chose 73 candidate genes that function within the cell polarity network to identify potential determinants that may concomitantly alter HSC fate while also exhibiting asymmetric segregation at cell division. Initial gene-expression profiles of polarity candidates showed high and differential expression in both HSCs and leukemia stem cells. Altered HSC fate was assessed by our established in vitro to in vivo screen on a subcohort of candidate polarity genes, which revealed 6 novel positive regulators of HSC function: Ap2a2, Gpsm2, Tmod1, Kif3a, Racgap1, and Ccnb1. Interestingly, live-cell videomicroscopy of the endocytic protein AP2A2 shows instances of asymmetric segregation during HSC/progenitor cell cytokinesis. These results contribute further evidence that ACD is functional in HSC self-renewal, suggest a role for Ap2a2 in HSC activity, and provide a unique opportunity to prospectively analyze progeny from HSC asymmetric divisions.
Collapse
|
22
|
Tomasetti C. On the probability of random genetic mutations for various types of tumor growth. Bull Math Biol 2012; 74:1379-95. [PMID: 22311065 DOI: 10.1007/s11538-012-9717-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2011] [Accepted: 01/13/2012] [Indexed: 11/24/2022]
Abstract
In this work, we consider the problem of estimating the probability for a specific random genetic mutation to be present in a tumor of a given size. Previous mathematical models have been based on stochastic methods where the tumor was assumed to be homogeneous and, on average, growing exponentially. In contrast, we are able to obtain analytical results for cases where the exponential growth of cancer has been replaced by other, arguably more realistic types of growth of a heterogeneous tumor cell population. Our main result is that the probability that a given random mutation will be present by the time a tumor reaches a certain size, is independent of the type of curve assumed for the average growth of the tumor, at least for a general class of growth curves. The same is true for the related estimate of the expected number of mutants present in a tumor of a given size, if mutants are indeed present.
Collapse
Affiliation(s)
- Cristian Tomasetti
- Department of Biostatistics, Harvard University, and Dana-Farber Cancer Institute, CLS11007, 450 Brookline Ave, Boston, MA 02215, USA.
| |
Collapse
|
23
|
Abstract
Hematopoiesis is the process that generates all the cell types of the blood, which are responsible for oxygen transport and immune defense. It has been now more than 50 years from the demonstration that blood cells derive from a common ancestor called Hematopoietic Stem Cell (HSC) McCulloch and Till (1960). Thus, the hematopoietic process relies on the unlimited and distinctive self-renewal ability of HSC, which in the adult mammalian organisms reside in the bone marrow, but their generation occurs during embryonic life. Questions still remain about how HSCs acquire and maintain the features of self-renewal and pluripotency that define stem-cell populations. Notch is a crucial signaling pathway involved in the generation of cell diversity and stem-cell maintenance in different systems. In some cases, Notch prevents differentiation, while in other contexts Notch directly participates in promoting cell differentiation. In the following sections, we will review what is known about the role of Notch in HSC establishment and hematopoietic cell lineage specification.
Collapse
|
24
|
Characterization and Classification of Stem Cells. Regen Med 2011. [DOI: 10.1007/978-90-481-9075-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
25
|
Role of symmetric and asymmetric division of stem cells in developing drug resistance. Proc Natl Acad Sci U S A 2010; 107:16766-71. [PMID: 20826440 DOI: 10.1073/pnas.1007726107] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Often, resistance to drugs is an obstacle to a successful treatment of cancer. In spite of the importance of the problem, the actual mechanisms that control the evolution of drug resistance are not fully understood. Many attempts to study drug resistance have been made in the mathematical modeling literature. Clearly, in order to understand drug resistance, it is imperative to have a good model of the underlying dynamics of cancer cells. One of the main ingredients that has been recently introduced into the rapidly growing pool of mathematical cancer models is stem cells. Surprisingly, this all-so-important subset of cells has not been fully integrated into existing mathematical models of drug resistance. In this work we incorporate the various possible ways in which a stem cell may divide into the study of drug resistance. We derive a previously undescribed estimate of the probability of developing drug resistance by the time a tumor is detected and calculate the expected number of resistant cancer stem cells at the time of tumor detection. To demonstrate the significance of this approach, we combine our previously undescribed mathematical estimates with clinical data that are taken from a recent six-year follow-up of patients receiving imatinib for the first-line treatment of chronic myelogenous leukemia. Based on our analysis we conclude that leukemia stem cells must tend to renew symmetrically as opposed to their healthy counterparts that predominantly divide asymmetrically.
Collapse
|
26
|
Wein F, Pietsch L, Saffrich R, Wuchter P, Walenda T, Bork S, Horn P, Diehlmann A, Eckstein V, Ho AD, Wagner W. N-cadherin is expressed on human hematopoietic progenitor cells and mediates interaction with human mesenchymal stromal cells. Stem Cell Res 2010; 4:129-39. [PMID: 20116358 DOI: 10.1016/j.scr.2009.12.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 12/11/2009] [Accepted: 12/22/2009] [Indexed: 10/20/2022] Open
Abstract
Specific cell-cell junctions between hematopoietic stem cells (HSC) and their niche have been shown to regulate stem cell function. N-cadherin was suggested to play a central role in this process, whereas other studies indicated that it did not play an essential role in the murine model. We have analyzed the role of N-cadherin for interaction between hematopoietic progenitor cells (HPC) and supportive mesenchymal stromal cells (MSC) in a human-human setting. Expression of N-cadherin and of cadherin-11 (osteoblast cadherin) was analyzed in HPC by quantitative RT-PCR, Western blot, and flow cytometry. N-cadherin and cadherin-11 were expressed in HPC at a moderate level, whereas they were not detectable in differentiated cells. Confocal laser scanning microscopy revealed that N-cadherin and beta-catenin are colocalized at the junction of HPC and MSC. siRNA knockdown of N-cadherin or cadherin-11 as well as treatment with the blocking function antibody decreased adhesive interaction of HPC to MSC. Furthermore, knockdown of N-cadherin or blocking function antibody impaired maintenance of long-term culture-initiating cells (LTC-IC) on coculture of HPC and MSC. These results indicate that N-cadherin is involved in the bidirectional interaction of human HPC with their cellular determinants in the niche.
Collapse
Affiliation(s)
- Frederik Wein
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Marciniak-Czochra A, Stiehl T, Wagner W. Modeling of replicative senescence in hematopoietic development. Aging (Albany NY) 2009; 1:723-32. [PMID: 20195386 PMCID: PMC2830082 DOI: 10.18632/aging.100072] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2009] [Accepted: 07/21/2009] [Indexed: 12/19/2022]
Abstract
Hematopoietic stem cells (HSC) give rise to an enormous number of blood cells throughout our life. In contrast their number of cell divisions preceding senescence is limited underin vitro culture conditions. Here we consider the question whether HSC can rejuvenate indefinitely or if the number of cell divisions is restricted. We have developed a multi-compartmental model for hematopoietic differentiation based on ordinary differential equations. The model is based on the hypothesis that in each step of maturation, the percentage of self-renewal versus differentiation is regulated by a single external feedback mechanism. We simulate the model under the assumption that hematopoietic differentiation precedes the six steps of maturation and the cells ultimately cease to proliferate after 50 divisions. Our results demonstrate that it is conceivable to maintain hematopoiesis over a life-time if HSC have a slow division rate and a high self-renewal rate. With age, the feedback signal increases and this enhances self-renewal, which results in the increase of the number of stem and progenitor cells. This study demonstrates that replicative senescence is compatible with life-long hematopoiesis and that model predictions are in line with experimental observations. Thus, HSC might not divide indefinitely with potentially important clinical implications.
Collapse
Affiliation(s)
- Anna Marciniak-Czochra
- Interdisciplinary Center of Scientific Computing (IWR), Institute of Applied Mathematics, University of Heidelberg, Germany
| | | | | |
Collapse
|
28
|
Marciniak-Czochra A, Stiehl T, Ho AD, Jäger W, Wagner W. Modeling of asymmetric cell division in hematopoietic stem cells--regulation of self-renewal is essential for efficient repopulation. Stem Cells Dev 2009; 18:377-85. [PMID: 18752377 DOI: 10.1089/scd.2008.0143] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Hematopoietic stem cells (HSCs) are characterized by their ability of self-renewal to replenish the stem cell pool and differentiation to more mature cells. The subsequent stages of progenitor cells also share some of this dual ability. It is yet unknown whether external signals modulate proliferation rate or rather the fraction of self-renewal. We propose three multicompartment models, which rely on a single external feedback mechanism. In Model 1 the signal enhances proliferation, whereas the self-renewal rates in all compartments are fixed. In Model 2 the signal regulates the rate of self-renewal, whereas the proliferation rate is unchanged. In Model 3, the signal regulates both proliferation and self-renewal rates. This study demonstrates that a unique strictly positive stable steady state can only be achieved by regulation of the rate of self-renewal. Furthermore, it requires a lower number of effective cell doublings. In order to maintain the stem cell pool, the self-renewal ratio of the HSC has to be > or =50% and it has to be higher than the self-renewal ratios of all downstream compartments. Interestingly, the equilibrium level of mature cells depends only on the parameters of self-renewal of HSC and it is independent of the parameters of dynamics of all upstream compartments. The model is compatible with the increase of leukocyte numbers following HSC transplantation. This study demonstrates that extrinsic regulation of the self-renewal rate of HSC is most essential in the process of hematopoiesis.
Collapse
Affiliation(s)
- Anna Marciniak-Czochra
- Interdisciplinary Center of Scientific Computing (IWR), Institute of Applied Mathematics, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | |
Collapse
|
29
|
Walenda T, Bork S, Horn P, Wein F, Saffrich R, Diehlmann A, Eckstein V, Ho AD, Wagner W. Co-culture with mesenchymal stromal cells increases proliferation and maintenance of haematopoietic progenitor cells. J Cell Mol Med 2009; 14:337-50. [PMID: 19432817 PMCID: PMC3837622 DOI: 10.1111/j.1582-4934.2009.00776.x] [Citation(s) in RCA: 126] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mesenchymal stromal cells (MSC) have been suggested to provide a suitable cellular environment for in vitro expansion of haematopoietic stem and progenitor cells (HPC) from umbilical cord blood. In this study, we have simultaneously analysed the cell division history and immunophenotypic differentiation of HPC by using cell division tracking with carboxyfluorescein diacetate N-succinimidyl ester (CFSE). Co-culture with MSC greatly enhanced proliferation of human HPC, especially of the more primitive CD34(+)CD38(-) fraction. Without co-culture CD34 and CD133 expressions decreased after several cell divisions, whereas CD38 expression was up-regulated after some cell divisions and then diminished in fast proliferating cells. Co-culture with MSC maintained a primitive immunophenotype (CD34(+), CD133(+) and CD38(-)) for more population doublings, whereas up-regulation of differentiation markers (CD13, CD45 and CD56) in HPC was delayed to higher numbers of cell divisions. Especially MSC of early cell passages maintained CD34 expression in HPC over more cell divisions, whereas MSC of higher passages further enhanced their proliferation rate. Inhibition of mitogen-activated protein kinase 1 (MAPK1) impaired proliferation and differentiation of HPC, but not maintenance of long-term culture initiating cells. siRNA knockdown of N-cadherin and VCAM1 in feeder layer cells increased the fraction of slow dividing HPC, whereas knockdown of integrin beta 1 (ITGB1) and CD44 impaired their differentiation. In conclusion, MSC support proliferation as well as self-renewal of HPC with primitive immunophenotype. The use of early passages of MSC and genetic manipulation of proteins involved in HPC-MSC interaction might further enhance cord blood expansion on MSC.
Collapse
Affiliation(s)
- Thomas Walenda
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Hawkins ED, Russell SM. Upsides and downsides to polarity and asymmetric cell division in leukemia. Oncogene 2009; 27:7003-17. [PMID: 19029941 DOI: 10.1038/onc.2008.350] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The notion that polarity regulators can act as tumor suppressors in epithelial cells is now well accepted. The function of these proteins in lymphocytes is less well explored, and their possible function as suppressors of leukemia has had little attention so far. We review the literature on lymphocyte polarity and the growing recognition that polarity proteins have an important function in lymphocyte function. We then describe molecular relationships between the polarity network and signaling pathways that have been implicated in leukemogenesis, which suggest mechanisms by which the polarity network might impact on leukemogenesis. We particularly focus on the possibility that disruption of polarity might alter asymmetric cell division (ACD), and that this might be a leukemia-initiating event. We also explore the converse possibility that leukemic stem cells might be produced or maintained by ACD, and therefore that Dlg, Scribble and Lgl might be important regulators of this process.
Collapse
Affiliation(s)
- E D Hawkins
- Immune Signalling Laboratory, Cancer Immunology, Research Division, Peter MacCallum Cancer Centre, East Melbourne, Victoria, Australia
| | | |
Collapse
|
31
|
Bosio A, Huppert V, Donath S, Hennemann P, Malchow M, Heinlein UAO. Isolation and enrichment of stem cells. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2009; 114:23-72. [PMID: 19347268 DOI: 10.1007/10_2008_38] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
Stem cells have the potential to revolutionize tissue regeneration and engineering. Both general types of stem cells, those with pluripotent differentiation potential as well as those with multipotent differentiation potential, are of equal interest. They are important tools to further understanding of general cellular processes, to refine industrial applications for drug target discovery and predictive toxicology, and to gain more insights into their potential for tissue regeneration. This chapter provides an overview of existing sorting technologies and protocols, outlines the phenotypic characteristics of a number of different stem cells, and summarizes their potential clinical applications.
Collapse
Affiliation(s)
- Andreas Bosio
- Miltenyi Biotec GmbH, Friedrich-Ebert-Strasse 68, 51429, Bergisch Gladbach, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Ashkenazi R, Gentry SN, Jackson TL. Pathways to tumorigenesis--modeling mutation acquisition in stem cells and their progeny. Neoplasia 2008; 10:1170-82. [PMID: 18953426 PMCID: PMC2570593 DOI: 10.1593/neo.08572] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 08/13/2008] [Accepted: 08/14/2008] [Indexed: 01/06/2023]
Abstract
Most adult tissues consist of stem cells, progenitors, and mature cells, and this hierarchical architecture may play an important role in the multistep process of carcinogenesis. Here, we develop and discuss the important predictions of a simple mathematical model of cancer initiation and early progression within a hierarchically structured tissue. This work presents a model that incorporates both the sequential acquisition of phenotype altering mutations and tissue hierarchy. The model simulates the progressive effect of accumulating mutations that lead to an increase in fitness or the induction of genetic instability. A novel aspect of the model is that symmetric self-renewal, asymmetric division, and differentiation are all incorporated, and this enables the quantitative study of the effect of mutations that deregulate the normal, homeostatic stem cell division pattern. The model is also capable of predicting changes in both tissue composition and in the progression of cells along their lineage at any given time and for various sequences of mutations. Simulations predict that the specific order in which mutations are acquired is crucial for determining the pace of cancer development. Interestingly, we find that the importance of genetic stability differs significantly depending on the physiological expression of mutations related to symmetric self-renewal and differentiation of stem and progenitor cells. In particular, mutations that lead to the alteration of the stem cell division pattern or the acquisition of some degree of immortality in committed progenitors lead to an early onset of cancer and diminish the impact of genetic instability.
Collapse
Affiliation(s)
- Rina Ashkenazi
- Department of Mathematics, University of Michigan, Ann Arbor, MI 48109, USA.
| | | | | |
Collapse
|
33
|
Wagner W, Horn P, Bork S, Ho AD. Aging of hematopoietic stem cells is regulated by the stem cell niche. Exp Gerontol 2008; 43:974-80. [DOI: 10.1016/j.exger.2008.04.007] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2008] [Revised: 04/04/2008] [Accepted: 04/10/2008] [Indexed: 12/16/2022]
|
34
|
Abstract
Hematopoietic stem cells have the potential to develop into multipotent and different lineage-restricted progenitor cells that subsequently generate all mature blood cell types. The classical model of hematopoietic lineage commitment proposes a first restriction point at which all multipotent hematopoietic progenitor cells become committed either to the lymphoid or to the myeloid development, respectively. Recently, this model has been challenged by the identification of murine as well as human hematopoietic progenitor cells with lymphoid differentiation capabilities that give rise to a restricted subset of the myeloid lineages. As the classical model does not include cells with such capacities, these findings suggest the existence of alternative developmental pathways that demand the existence of additional branches in the classical hematopoietic tree. Together with some phenotypic criteria that characterize different subsets of multipotent and lineage-restricted progenitor cells, we summarize these recent findings here.
Collapse
Affiliation(s)
- Bernd Giebel
- Institute for Transplantation Diagnostics and Cellular Therapeutics, Heinrich Heine University, Düsseldorf, Germany.
| | | |
Collapse
|
35
|
Congdon KL, Reya T. Divide and conquer: how asymmetric division shapes cell fate in the hematopoietic system. Curr Opin Immunol 2008; 20:302-7. [PMID: 18554882 DOI: 10.1016/j.coi.2008.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2008] [Revised: 05/08/2008] [Accepted: 05/11/2008] [Indexed: 10/22/2022]
Abstract
A fundamental mechanism by which cells can give rise to daughters with different fates is via asymmetric division. During asymmetric division, a mother cell generates daughter cells that go on to adopt different fates because of differential segregation of cell fate determinants. Although originally characterized in invertebrates, asymmetric division has recently been shown to regulate cell fate decisions in the mammalian hematopoietic system, playing crucial roles in stem cell renewal, lymphocyte activation, and leukemogenesis. These discoveries have opened new doors to understanding how regulation of division pattern contributes to the normal development and function of the immune system as well as how its dysregulation can lead to cancer.
Collapse
Affiliation(s)
- Kendra L Congdon
- Department of Pharmacology and Cancer Biology, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
36
|
Wagner W, Saffrich R, Ho AD. The Stromal Activity of Mesenchymal Stromal Cells. Transfus Med Hemother 2008; 35:185-193. [PMID: 21547116 PMCID: PMC3083286 DOI: 10.1159/000128956] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2007] [Accepted: 12/06/2007] [Indexed: 12/29/2022] Open
Abstract
SUMMARY: The mechanism that regulates self-renewal and differentiation of hematopoietic stem cells (HSC) is a central question in stem cell biology that might ultimately lead to reliable protocols for in vitro expansion of HSC. Cellular fate is governed by cell-cell interaction with the microenvironment in the bone marrow, the stem cell niche. Mesenchymal stromal cells (MSC) are precursors of the cellular components, and they secrete extracellular matrix proteins of the bone marrow stroma. Therefore, MSC feeder layer might provide a suitable in vitro model system for the stem cell niche. In vitro assays demonstrate that MSC maintain the stem cell function of HSC and that MSC from bone marrow have a higher hematopoiesis supportive activity than MSC from adipose tissue. Co-cultivation with MSC might pave the way for expansion of long-term repopulating HSC, and various clinical trials indicate that co-transplantation of HSC and MSC might enhance engraftment. Thus, MSC are promising tools to elucidate the underlying mechanism of the cellular microenvironment. The large variety of preparative protocols for isolation and cultivation of MSC affects their stromal activity. Standardized isolation methods and molecular characterization of MSC are of utmost importance for reproducible isolation of hematopoiesis supportive stromal cells and for their potential clinical application.
Collapse
Affiliation(s)
- Wolfgang Wagner
- Department of Medicine V, University of Heidelberg, Germany
- Department of Physiology and Pathophysiology, University of Heidelberg, Germany
| | | | - Anthony D. Ho
- Department of Medicine V, University of Heidelberg, Germany
| |
Collapse
|
37
|
Rieger MA, Schroeder T. Exploring hematopoiesis at single cell resolution. Cells Tissues Organs 2008; 188:139-49. [PMID: 18230950 DOI: 10.1159/000114540] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hematopoietic stem cell research has made tremendous progress over the last decades, and blood has become one of the best understood mammalian stem cell systems. The easy accessibility of hematopoietic cells, which are not tightly embedded in tissue, has supported this fast development. However, the hematopoietic system also exhibits disadvantages over other stem cell systems: the identity of individual cells is quickly lost when followed in cell culture and developmental stages cannot easily be distinguished by morphology. Therefore, difficulties to constantly analyze the fate of single cells are one reason for many open questions in hematopoiesis. So far, most findings are based on endpoint analyses of populations, consisting of heterogeneous cells in different stages of development or cell cycle. However, endpoint analyses merely reflect the result of a progressive sequence of fate decisions, whereas individual decisions, which would elucidate stem cell behavior, are not investigated. Thorough observation of the fate of individual cells and their progeny over many generations will add to a comprehensive understanding of the regulation of stem cell behavior. Here, we review current attempts of single cell analyses in hematopoiesis research and outline how time-lapse imaging and single cell tracking can contribute to approaching long-standing questions in hematopoiesis.
Collapse
Affiliation(s)
- Michael A Rieger
- Institute of Stem Cell Research, Helmholtz Zentrum Munchen - German Research Center for Environmental Health, Neuherberg/Munich, Germany
| | | |
Collapse
|
38
|
Martinez-Agosto JA, Mikkola HKA, Hartenstein V, Banerjee U. The hematopoietic stem cell and its niche: a comparative view. Genes Dev 2008; 21:3044-60. [PMID: 18056420 DOI: 10.1101/gad.1602607] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Stem cells have been identified as a source of virtually all highly differentiated cells that are replenished during the lifetime of an animal. The critical balance between stem and differentiated cell populations is crucial for the long-term maintenance of functional tissue types. Stem cells maintain this balance by choosing one of several alternate fates: self-renewal, commitment to differentiate, and senescence or cell death. These characteristics comprise the core criteria by which these cells are usually defined. The self-renewal property is important, as it allows for extended production of the corresponding differentiated cells throughout the life span of the animal. A microenvironment that is supportive of stem cells is commonly referred to as a stem cell niche. In this review, we first present some general concepts regarding stem cells and their niches, comparing stem cells of many different kinds from diverse organisms, and in the second part, we compare specific aspects of hematopoiesis and the niches that support hematopoiesis in Drosophila, zebrafish and mouse.
Collapse
Affiliation(s)
- Julian A Martinez-Agosto
- Department of Human Genetics and Department of Pediatrics, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, California 90095, USA
| | | | | | | |
Collapse
|
39
|
Wagner W, Wein F, Roderburg C, Saffrich R, Diehlmann A, Eckstein V, Ho AD. Adhesion of human hematopoietic progenitor cells to mesenchymal stromal cells involves CD44. Cells Tissues Organs 2007; 188:160-9. [PMID: 18160820 DOI: 10.1159/000112821] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Direct cell-cell contact between hematopoietic progenitor cells (HPC) and their cellular microenvironment is essential for maintenance of 'stemness'. We have previously demonstrated that a feeder layer of human mesenchymal stromal cells (MSC) could provide a surrogate model as a niche for human HPC. Maintenance of long-term culture-initiating cells was significantly lower on fibroblasts. METHODS Adhesion of HPC to MSC was further analyzed using our recently described adhesion assay based on gravitational force upon inversion and in combination with specific antibodies against CD44. RESULTS Adhesion of KG1a and CD34+ cells was significantly reduced by administration of a monoclonal CD44 antibody and for KG1a to a greater extent than for CD34+ cells. Interaction of HPC and MSC was further analyzed by laser scanning confocal microscopy. CD44 was located on the uropod of CD34+ cells at the site of contact with MSC and both cell types were interwoven by a network of fibronectin. CONCLUSION Various adhesion proteins, including CD44, are involved in the contact of human HPC and human MSC and further analysis of the relative significance and interaction of these proteins will be crucial for the understanding of the mechanism of this specific cell-cell interaction.
Collapse
Affiliation(s)
- Wolfgang Wagner
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | | | |
Collapse
|
40
|
Giebel B. Cell polarity and asymmetric cell division within human hematopoietic stem and progenitor cells. Cells Tissues Organs 2007; 188:116-26. [PMID: 18160821 DOI: 10.1159/000112842] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Like other somatic stem cells, hematopoietic stem cells (HSC) contain the capacity to self-renew and to give rise to committed progenitor cells that are able to replenish all hematopoietic cell types. To keep a constant level of HSC, the decision whether their progeny maintain the stem cell fate or become committed to differentiation needs to be highly controlled. In this context it became evident that HSC niches fulfill important functions in keeping the level of HSC more or less constant. Before discovering such niches, it was widely assumed that HSC divide asymmetrically to give birth to a daughter cell maintaining the stem cell fate and to another one which is committed to differentiation. Here, I summarize some of the experimental data being compatible with the model of asymmetric cell division and review some of our latest findings, which demonstrate the occurrence of asymmetric cell divisions within the HSC and hematopoietic progenitor cell compartment. Since cell polarity is an essential prerequisite for asymmetrically dividing as well as for migrating cells, I will also discuss some aspects of cell polarity of primitive hematopoietic cells.
Collapse
Affiliation(s)
- Bernd Giebel
- Institute for Transplantation Diagnostics and Cellular Therapeutics, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
41
|
Boquest AC, Noer A, Collas P. Epigenetic programming of mesenchymal stem cells from human adipose tissue. ACTA ACUST UNITED AC 2007; 2:319-29. [PMID: 17848719 DOI: 10.1007/bf02698059] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 01/01/2023]
Abstract
Stromal stem cells identified in various adult mesenchymal tissues (commonly called mesenchymal stem cells [MSCs]) have in past years received more attention as a result of their potential interest as replacement cells in regenerative medicine. An abundant and easily accessible source of adult human MSCs are stem cells harvested from liposuction material. Similarly to bone marrow-derived MSCs, human adipose tissue-derived stem cells (ASCs) can give rise to a variety of cell types in vitro and in vivo; however, they have a propensity to differentiate into primarily mesodermal lineages. Even so, their capacity to differentiate into nonadipogenic mesodermal pathways seems to be restricted. Emerging DNA methylation profiles at adipogenic and nonadipogenic gene promoters in freshly isolated, cultured, or differentiated ASCs aim to provide an epigenetic explanation for this restrictive differentiation potential. A review of these studies indicates that human ASCs are epigenetically marked by mosaic hypomethylation of adipogenic promoters, whereas nonadipogenic lineage-specific promoters are hypermethylated. Surprisingly, in vitro differentiation toward various pathways maintains the overall methylation profiles of undifferentiated cells, raising the hypothesis that ASCs are at least epigenetically preprogrammed for adipogenesis. Novel attempts at reprogramming the epigenome of MSCs have been initiated to enhance the differentiation capacity of these cells.
Collapse
Affiliation(s)
- Andrew C Boquest
- Institute of Basic Medical Sciences, Department of Biochemistry, Faculty of Medicine, University of Oslo, Blindern, 0317 Oslo, Norway
| | | | | |
Collapse
|
42
|
Gottschling S, Eckstein V, Saffrich R, Jonás A, Uhrig M, Krause U, Seckinger A, Miesala K, Horsch K, Straub BK, Ho AD. Primitive and committed human hematopoietic progenitor cells interact with primary murine neural cells and are induced to undergo self-renewing cell divisions. Exp Hematol 2007; 35:1858-71. [PMID: 17697743 DOI: 10.1016/j.exphem.2007.06.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2007] [Revised: 06/04/2007] [Accepted: 06/18/2007] [Indexed: 12/23/2022]
Abstract
OBJECTIVE Studies in animal models have indicated that hematopoietic progenitor cells (HPC) migrate and home to the central nervous system and might acquire neural features under specific circumstances. The interaction between HPC and the neural environment and the functional effect on hematopoiesis have not yet been defined. METHODS CD34(+)133(+) cells from mobilized peripheral blood were cocultured with primary murine neurons or astrocytes. Chemotaxis and adhesive interactions were studied by applying beta(1)- and beta(2)-integrin function-blocking anibodies. The impact of neural feeder layers on integrin expression of HPC and the presence of appropriate adhesion ligands on neural cells were determined by immunostaining and flow cytometry. The hematopoietic long-term fate was monitored by time-lapse microscopy of individual cell-division history followed by long-term culture-initiating cell (LTC-IC) and colony-forming cell (CFC) assays. Neural differentiation was assessed by immunostaining against specific neuronal and glial antigens. RESULTS The 23.0% +/- 4.9% of HPC showed stromal cell-derived factor-1-induced migration toward neural cells, and 20.2% +/- 1.6% displayed firm beta(1)-integrin-mediated adhesion to astrocytes. The latter expressed appropriate adhesion ligands, stabilized beta(1)-integrin expression, and increased beta(2)-integrin expression of HPC. Neural differentiation of HPC could not be identified but astrocytes were able to induce limited self-renewing cell divisions of HPC and thus maintain 25.8% +/- 3.4% of the initial LTC-IC and 80.7% +/- 1.9% of the initial CFC. CONCLUSION Human HPC are able to interact with neural cells and interaction maintains, albeit to a limited extent, the self-renewal capability of HPC.
Collapse
Affiliation(s)
- Sandra Gottschling
- Department of Medicine V, Ruprecht-Karls University, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Ho AD, Wagner W. The beauty of asymmetry: asymmetric divisions and self-renewal in the haematopoietic system. Curr Opin Hematol 2007; 14:330-6. [PMID: 17534157 DOI: 10.1097/moh.0b013e3281900f12] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
PURPOSE OF REVIEW The hallmark of stem cells is their dual abilities to self-renew and to differentiate into multiple lineages. To fulfill these functions they must undergo asymmetric division. A central question in developmental biology is how can a single cell divide to produce two progeny cells that adopt different fates? We provided evidence of the significance of asymmetric division in human haematopoietic stem cells. RECENT FINDINGS By monitoring the symmetry of divisions of haematopoietic stem cells and following their subsequent developmental potentials at the single cell level, we established a relationship between divisional kinetics and self-renewal capacity. Direct cell-cell contact with cellular determinants in the niche has been shown to play an essential role in maintaining stemness. The creation of in-vitro models for the niche, such as human mesenchymal stromal cells, has provided a controlled laboratory environment in which the relative significance of chemokines and adhesion molecules can be studied. SUMMARY Identification of the molecular interactions between stem cells and their niche has led to an understanding of the mechanisms that control the self-renewal of stem cells. Ultimately, molecular signals triggered by adhesion and junction complexes are responsible for the adoption of specific cell fate.
Collapse
Affiliation(s)
- Anthony D Ho
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany.
| | | |
Collapse
|
44
|
Beckmann J, Scheitza S, Wernet P, Fischer JC, Giebel B. Asymmetric cell division within the human hematopoietic stem and progenitor cell compartment: identification of asymmetrically segregating proteins. Blood 2007; 109:5494-501. [PMID: 17332245 DOI: 10.1182/blood-2006-11-055921] [Citation(s) in RCA: 98] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
The findings that many primitive human hematopoietic cells give rise to daughter cells that adopt different cell fates and/or show different proliferation kinetics suggest that hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs) can divide asymmetrically. However, definitive experimental demonstration is lacking due to the current absence of asymmetrically segregating marker molecules within the primitive hematopoietic cell compartment. Thus, it remains an open question as to whether HSCs/HPCs have the capability to divide asymmetrically, or whether the differences that have been observed are established by extrinsic mechanisms that act on postmitotic progenitors. Here, we have identified 4 proteins (CD53, CD62L/L-selectin, CD63/lamp-3, and CD71/transferrin receptor) that segregate differentially in about 20% of primitive human hematopoietic cells that divide in stroma-free cultures. Therefore, this indicates for the first time that HSCs/HPCs have the capability to divide asymmetrically. Remarkably, these proteins, in combination with the surrogate stem-cell marker CD133, help to discriminate the more primitive human cultivated HSCs/HPCs. Since 3 of these proteins, the transferrin receptor and the tetraspanins CD53 and CD63, are endosomal-associated proteins, they may provide a link between the endosomal compartment and the process of asymmetric cell division within the HSC/HPC compartment.
Collapse
Affiliation(s)
- Julia Beckmann
- Institute for Transplantation Diagnostics and Cellular Therapeutics, Heinrich-Heine-University Düsseldorf, Germany
| | | | | | | | | |
Collapse
|
45
|
Wagner W, Wein F, Roderburg C, Saffrich R, Faber A, Krause U, Schubert M, Benes V, Eckstein V, Maul H, Ho AD. Adhesion of hematopoietic progenitor cells to human mesenchymal stem cells as a model for cell−cell interaction. Exp Hematol 2007; 35:314-25. [PMID: 17258080 DOI: 10.1016/j.exphem.2006.10.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2006] [Revised: 08/30/2006] [Accepted: 10/05/2006] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The significant role of direct contact between hematopoietic progenitor cells (HPC) and the cellular microenvironment for maintaining "stemness" has been demonstrated. Human mesenchymal stem cell (MSC) feeder layers represent a surrogate model for this interaction. Specific adhesion molecules are responsible for this cell-cell contact. METHODS To define cell-cell contact between HPC and MSC, we have studied adhesive interaction of various fractions of HPC by using a novel assay based on gravitational force upon inversion. Adherent and nonadherent cells were separated and further analyzed with regard to gene expression and long-term hematopoietic culture initiating cell (LTC-IC) frequency. RESULTS HPC subsets with higher self-renewing capacity demonstrated significantly higher adherence to human MSC (CD34(+) vs CD34(-), CD34(+)/CD38(-) vs CD34(+)/CD38(+), slow dividing fraction vs fast dividing fraction). LTC-IC frequency was significantly higher in the adherent fraction than in the nonadherent fraction. Furthermore, genes coding for adhesion proteins and extracellular matrix were higher expressed in the adherent subsets of CD34(+) cells (fibronectin 1, cadherin 11, vascular cell adhesion molecule-1, connexin 43, integrin beta-like 1, and TGFBI). CONCLUSION In this study we have demonstrated that primitive subsets of HPC have higher affinity to human MSC. The essential role of specific junction proteins for stabilization of cell-cell contact is indicated by their significant higher expression.
Collapse
Affiliation(s)
- Wolfgang Wagner
- Department of Medicine V, University of Heidelberg, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Gottschling S, Saffrich R, Seckinger A, Krause U, Horsch K, Miesala K, Ho AD. Human mesenchymal stromal cells regulate initial self-renewing divisions of hematopoietic progenitor cells by a beta1-integrin-dependent mechanism. Stem Cells 2006; 25:798-806. [PMID: 17110618 DOI: 10.1634/stemcells.2006-0513] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In previous reports, we have demonstrated that only direct cell-cell contact with stromal cells, such as the murine stromal cell line AFT024, was able to alter the cell division kinetics and self-renewing capacity of hematopoietic progenitor cells (HPC). Because beta(1)-integrins were shown to be crucial for the interaction of HPC with the bone marrow microenvironment, we have studied the role of beta(1)-integrins in the regulation of self-renewing cell divisions. For this purpose, we used primary human mesenchymal stromal (MS) cells as in vitro surrogate niche and monitored the division history and subsequent functional fate of individually plated CD34(+)133(+) cells in the absence or presence of an anti-beta(1)-integrin blocking antibody by time-lapse microscopy and subsequent long-term culture-initiating cell (LTC-IC) assays. beta(1)-Integrin-mediated contact with MS cells significantly increased the proportion of asymmetrically dividing cells and led to a substantial increase of LTC-IC. Provided that beta(1)-integrin-mediated contact was available within the first 72 hours, human MS cells were able to recruit HPC into cell cycle and accelerate their division kinetics without loss of stem cell function. Activation of beta(1)-integrins by ligands alone (e.g., fibronectin and vascular cell adhesion molecule-1) was not sufficient to alter the cell division symmetry and promote self-renewal of HPC, thus indicating an indirect effect. These results have provided evidence that primary human MS cells are able to induce self-renewing divisions of HPC by a beta(1)-integrin-dependent mechanism.
Collapse
Affiliation(s)
- Sandra Gottschling
- Department of Medicine V, Ruprecht-Karls University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany
| | | | | | | | | | | | | |
Collapse
|
47
|
Noer A, Sørensen AL, Boquest AC, Collas P. Stable CpG hypomethylation of adipogenic promoters in freshly isolated, cultured, and differentiated mesenchymal stem cells from adipose tissue. Mol Biol Cell 2006; 17:3543-56. [PMID: 16760426 PMCID: PMC1525236 DOI: 10.1091/mbc.e06-04-0322] [Citation(s) in RCA: 120] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stem cells from adipose tissue can differentiate into mesodermal lineages. Differentiation potential, however, varies between clones of adipose stem cells (ASCs), raising the hypothesis that epigenetic differences account for this variability. We report here a bisulfite sequencing analysis of CpG methylation of adipogenic (leptin [LEP], peroxisome proliferator-activated receptor gamma 2 [PPARG2], fatty acid-binding protein 4 [FABP4], and lipoprotein lipase [LPL]) promoters and of nonadipogenic (myogenin [MYOG], CD31, and GAPDH) loci in freshly isolated human ASCs and in cultured ASCs, in relation to gene expression and differentiation potential. Uncultured ASCs display hypomethylated adipogenic promoters, in contrast to myogenic and endothelial loci, which are methylated. Adipogenic promoters exhibit mosaic CpG methylation, on the basis of heterogeneous methylation between cells and of variation in the extent of methylation of a given CpG between donors, and both between and within clonal cell lines. DNA methylation reflects neither transcriptional status nor potential for gene expression upon differentiation. ASC culture preserves hypomethylation of adipogenic promoters; however, between- and within-clone mosaic methylation is detected. Adipogenic differentiation also maintains the overall CpG hypomethylation of LEP, PPARG2, FABP4, and LPL despite demethylation of specific CpGs and transcriptional induction. Furthermore, enhanced methylation at adipogenic loci in primary differentiated cells unrelated to adipogenesis argues for ASC specificity of the hypomethylated state of these loci. Therefore, mosaic hypomethylation of adipogenic promoters may constitute a molecular signature of ASCs, and DNA methylation does not seem to be a determinant of differentiation potential of these cells.
Collapse
Affiliation(s)
- Agate Noer
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Anita L. Sørensen
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Andrew C. Boquest
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| | - Philippe Collas
- Department of Biochemistry, Institute of Basic Medical Sciences, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
48
|
Ho AD, Wagner W. Bone marrow niche and leukemia. ERNST SCHERING FOUNDATION SYMPOSIUM PROCEEDINGS 2006:125-139. [PMID: 17939299 DOI: 10.1007/2789_2007_048] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2023]
Abstract
Mounting evidence indicates that human cancers may originate from malignant transformation of stem cells. The most convincing proof is found in acute myeloid leukemia, where only a small subset of slowly dividing cells was able to induce transplantable acute myeloid leukemia. Normal hematopoietic stem cells (HSC) are characterized by their unlimited ability to self-renew, give rise to a multitude of cells that exhibit more differentiated features, and show slow division kinetics. Using human HSC and mesenchymal stromal cells (MSC) as models, we and others have demonstrated the vital role of the cellular niche in maintaining the self-renewing capacity, that is, "stemness" of HSC. Without direct contact with the cellular niche, HSC tend to differentiate and lose their stemness. Similar to their normal counterparts, leukemia stem cells divide slowly and maintain their self-renewal capacity through interaction with the niche. As a consequence, they are resistant to conventional chemotherapy strategies that target rapidly dividing cells. Thus it is of utmost importance to understand the interaction between cellular niche and normal HSC as well as between leukemia stem cells and the niche to provide a basis for more efficient treatment strategies.
Collapse
Affiliation(s)
- A D Ho
- Department of Medicine V, University of Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg, Germany.
| | | |
Collapse
|