1
|
Robert C, Prista von Bonhorst F, Dupont G, Gonze D, De Decker Y. Role of tristability in the robustness of the differentiation mechanism. PLoS One 2025; 20:e0316666. [PMID: 40106426 PMCID: PMC11922266 DOI: 10.1371/journal.pone.0316666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 12/14/2024] [Indexed: 03/22/2025] Open
Abstract
During cell differentiation, identical pluripotent cells undergo a specification process marked by changes in the expression of key genes, regulated by transcription factors that can inhibit the transcription of a competing gene or activate their own transcription. This specification is orchestrated by gene regulatory networks (GRNs), encompassing transcription factors, biochemical reactions, and signalling cascades. Mathematical models for these GRNs have been proposed in various contexts, to replicate observed robustness in differentiation properties. This includes reproducible proportions of differentiated cells with respect to parametric or stochastic noise and the avoidance of transitions between differentiated states. Understanding the GRN components controlling these features is crucial. Our study thoroughly explored an extended version of the Toggle Switch model with auto-activation loops. This model represents cells evolving from common progenitors in one out of two fates (A or B, bistable regime) or, additionally, remaining in their progenitor state (C, tristable regime). Such a differentiation into populations with three distinct cell fates is observed during blastocyst formation in mammals, where inner cell mass cells can remain in that state or differentiate into epiblast cells or primitive endoderm. Systematic analysis revealed that the existence of a stable non-differentiated state significantly impacts the GRN's robustness against parametric variations and stochastic noise. This state reduces the sensitivity of cell populations to parameters controlling key gene expression asymmetry and prevents cells from making transitions after acquiring a new identity. Stochastic noise enhances robustness by decreasing sensitivity to initial expression levels and helping the system escape from the non-differentiated state to differentiated cell fates, making the differentiation more efficient.
Collapse
Affiliation(s)
- Corentin Robert
- Nonlinear Physical Chemistry Unit, Université Libre de Bruxelles (ULB), Brussels, Belgium
- Unit of Theoretical Chronobiology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Didier Gonze
- Unit of Theoretical Chronobiology, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Yannick De Decker
- Nonlinear Physical Chemistry Unit, Université Libre de Bruxelles (ULB), Brussels, Belgium
| |
Collapse
|
2
|
Warsi S, Dahl M, Smith EMK, Rydström A, Mansell E, Sigurdsson V, Sjöberg J, Soneji S, Rörby E, Siva K, Grahn THM, Liu Y, Blank U, Karlsson G, Karlsson S. Schlafen2 is a regulator of quiescence in adult murine hematopoietic stem cells. Haematologica 2022; 107:2884-2896. [PMID: 35615926 PMCID: PMC9713563 DOI: 10.3324/haematol.2021.279799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Indexed: 12/14/2022] Open
Abstract
Even though hematopoietic stem cells (HSC) are characterized by their ability to self-renew and differentiate, they primarily reside in quiescence. Despite the immense importance of this quiescent state, its maintenance and regulation is still incompletely understood. Schlafen2 (Slfn2) is a cytoplasmic protein known to be involved in cell proliferation, differentiation, quiescence, interferon response, and regulation of the immune system. Interestingly, Slfn2 is highly expressed in primitive hematopoietic cells. In order to investigate the role of Slfn2 in the regulation of HSC we have studied HSC function in the elektra mouse model, where the elektra allele of the Slfn2 gene contains a point mutation causing loss of function of the Slfn2 protein. We found that homozygosity for the elektra allele caused a decrease of primitive hematopoietic compartments in murine bone marrow. We further found that transplantation of elektra bone marrow and purified HSC resulted in a significantly reduced regenerative capacity of HSC in competitive transplantation settings. Importantly, we found that a significantly higher fraction of elektra HSC (as compared to wild-type HSC) were actively cycling, suggesting that the mutation in Slfn2 increases HSC proliferation. This additionally caused an increased amount of apoptotic stem and progenitor cells. Taken together, our findings demonstrate that dysregulation of Slfn2 results in a functional deficiency of primitive hematopoietic cells, which is particularly reflected by a drastically impaired ability to reconstitute the hematopoietic system following transplantation and an increase in HSC proliferation. This study thus identifies Slfn2 as a novel and critical regulator of adult HSC and HSC quiescence.
Collapse
Affiliation(s)
- Sarah Warsi
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University,Skåne University Hospital, Region Skåne,S. Warsi
| | - Maria Dahl
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Emma M. K. Smith
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Anna Rydström
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Els Mansell
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Valgardur Sigurdsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Julia Sjöberg
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Shamit Soneji
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University,Lund University Bioinformatics Core, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Emma Rörby
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Kavitha Siva
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Tan H. M. Grahn
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Yang Liu
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Ulrika Blank
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| | - Göran Karlsson
- Division of Molecular Hematology, Lund Stem Cell Center, Lund University
| | - Stefan Karlsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University
| |
Collapse
|
3
|
Warsi S, Blank U, Dahl M, Hooi Min Grahn T, Schmiderer L, Andradottir S, Karlsson S. BMP signaling is required for postnatal murine hematopoietic stem cell self-renewal. Haematologica 2021; 106:2203-2214. [PMID: 32675226 PMCID: PMC8327730 DOI: 10.3324/haematol.2019.236125] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Indexed: 12/12/2022] Open
Abstract
Life-long production of blood from hematopoietic stem cells (HSC) is a process of strict modulation. Intrinsic and extrinsic signals govern fate options like self-renewal – a cardinal feature of HSC. Bone morphogenetic proteins (BMP) have an established role in embryonic hematopoiesis, but less is known about its functions in adulthood. Previously, SMAD-mediated BMP signaling has been proven dispensable for HSC. However, the BMP type-II receptor (BMPR-II) is highly expressed in HSC, leaving the possibility that BMP function via alternative pathways. Here, we establish that BMP signaling is required for selfrenewal of adult HSC. Through conditional knockout we show that BMPR-II deficient HSC have impaired self-renewal and regenerative capacity. BMPR-II deficient cells have reduced p38 activation, implying that non-SMAD pathways operate downstream of BMP in HSC. Indeed, a majority of primitive hematopoietic cells do not engage in SMADmediated responses downstream of BMP in vivo. Furthermore, deficiency of BMPR-II results in increased expression of TJP1, a known regulator of self-renewal in other stem cells, and knockdown of TJP1 in primitive hematopoietic cells partly rescues the BMPR-II null phenotype. This suggests TJP1 may be a universal stem cell regulator. In conclusion, BMP signaling, in part mediated through TJP1, is required endogenously by adult HSC to maintain self-renewal capacity and proper resilience of the hematopoietic system during regeneration.
Collapse
Affiliation(s)
- Sarah Warsi
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Ulrika Blank
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Maria Dahl
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Tan Hooi Min Grahn
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | - Ludwig Schmiderer
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| | | | - Stefan Karlsson
- Division of Molecular Medicine and Gene Therapy, Lund Stem Cell Center, Lund University, Lund, Sweden
| |
Collapse
|
4
|
Search for Novel Plasma Membrane Proteins as Potential Biomarkers in Human Mesenchymal Stem Cells Derived from Dental Pulp, Adipose Tissue, Bone Marrow, and Hair Follicle. J Membr Biol 2021; 254:409-422. [PMID: 34230997 DOI: 10.1007/s00232-021-00190-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 06/07/2021] [Indexed: 10/20/2022]
Abstract
One of the drawbacks preventing the use of mesenchymal stem cells (MSCs) in clinical practice is the heterogeneous nature of their cultures. MSC cultures are not homogeneously formed by the MSCs and may contain non-mesenchymal cell types. Therefore, prior to use in clinics or research, complete characterization of MSCs should be performed to demonstrate the existence or absence of proper stem cell markers, many of which are happened to be cell-surface proteins. Unfortunately, the success of MSC characterization studies is limited due to the low specificity of the currently available cell-surface markers. Therefore, in this study, we aimed to investigate the plasma membrane (PM) proteins of MSCs isolated from human dental pulp (DP), adipose tissue (AT), bone marrow (BM), and hair follicle (HF) with the hope of proposing novel putative specific MSC markers. Differential-velocity centrifugation was used to enrich PM proteins. The isolated proteins were then identified by nLC-MS/MS and subjected to bioinformatics analysis. Proteins that were unique to each MSC type (CD9, CD10, CD63 for DP-MSCs; CD26, CD81, CD201, CD364 for AT-MSCs; Cd49a, CD49d for HF-MSCs; CD49e, CD56, CD92, CD97, CD156b, CD156c, CD220, CD221, CD298, CD315 for BM-MSCs) and common to all four MSC types (CD13, CD29, CD44, CD51, CD59, CD73, CD90) were identified. Uncharacterized proteins that have transmembrane (TM) domains were also detected. Some of the proteins identified in this study were the putative cell-surface markers that might be used for characterization of MSCs.
Collapse
|
5
|
Leguit RJ, Raymakers RAP, Hebeda KM, Goldschmeding R. CCN2 (Cellular Communication Network factor 2) in the bone marrow microenvironment, normal and malignant hematopoiesis. J Cell Commun Signal 2021; 15:25-56. [PMID: 33428075 PMCID: PMC7798015 DOI: 10.1007/s12079-020-00602-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 12/20/2020] [Indexed: 02/06/2023] Open
Abstract
CCN2, formerly termed Connective Tissue Growth Factor, is a protein belonging to the Cellular Communication Network (CCN)-family of secreted extracellular matrix-associated proteins. As a matricellular protein it is mainly considered to be active as a modifier of signaling activity of several different signaling pathways and as an orchestrator of their cross-talk. Furthermore, CCN2 and its fragments have been implicated in the regulation of a multitude of biological processes, including cell proliferation, differentiation, adhesion, migration, cell survival, apoptosis and the production of extracellular matrix products, as well as in more complex processes such as embryonic development, angiogenesis, chondrogenesis, osteogenesis, fibrosis, mechanotransduction and inflammation. Its function is complex and context dependent, depending on cell type, state of differentiation and microenvironmental context. CCN2 plays a role in many diseases, especially those associated with fibrosis, but has also been implicated in many different forms of cancer. In the bone marrow (BM), CCN2 is highly expressed in mesenchymal stem/stromal cells (MSCs). CCN2 is important for MSC function, supporting its proliferation, migration and differentiation. In addition, stromal CCN2 supports the maintenance and longtime survival of hematopoietic stem cells, and in the presence of interleukin 7, stimulates the differentiation of pro-B lymphocytes into pre-B lymphocytes. Overexpression of CCN2 is seen in the majority of B-acute lymphoblastic leukemias, especially in certain cytogenetic subgroups associated with poor outcome. In acute myeloid leukemia, CCN2 expression is increased in MSCs, which has been associated with leukemic engraftment in vivo. In this review, the complex function of CCN2 in the BM microenvironment and in normal as well as malignant hematopoiesis is discussed. In addition, an overview is given of data on the remaining CCN family members regarding normal and malignant hematopoiesis, having many similarities and some differences in their function.
Collapse
Affiliation(s)
- Roos J. Leguit
- Department of Pathology, University Medical Center Utrecht, H04-312, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| | - Reinier A. P. Raymakers
- Department of Hematology, UMCU Cancer Center, Heidelberglaan 100 B02.226, 3584 CX Utrecht, The Netherlands
| | - Konnie M. Hebeda
- Department of Pathology, Radboud University Medical Centre, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
| | - Roel Goldschmeding
- Department of Pathology, University Medical Centre Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
| |
Collapse
|
6
|
Balise VD, Saito-Reis CA, Gillette JM. Tetraspanin Scaffold Proteins Function as Key Regulators of Hematopoietic Stem Cells. Front Cell Dev Biol 2020; 8:598. [PMID: 32754593 PMCID: PMC7381308 DOI: 10.3389/fcell.2020.00598] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 06/19/2020] [Indexed: 02/06/2023] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) are responsible for the development, maintenance, and regeneration of all the blood forming cells in the body, and as such, are critical for a number of patient therapies. For successful HSPC transplantation, stem cells must traffic through the blood and home to the bone marrow (BM) microenvironment or “niche,” which is composed of soluble factors, matrix proteins, and supportive cells. HSPC adhesion to, and signaling with, cellular and extracellular components of the niche provide instructional cues to balance stem cell self-renewal and differentiation. In this review, we will explore the regulation of these stem cell properties with a focus on the tetraspanin family of membrane proteins. Tetraspanins are molecular scaffolds that uniquely function to distribute proteins into highly organized microdomains comprising adhesion, signaling, and adaptor proteins. As such, tetraspanins contribute to many aspects of cell physiology as mediators of cell adhesion, trafficking, and signaling. We will summarize the many reports that identify tetraspanins as markers of specific HSPC populations. Moreover, we will discuss the various studies establishing the functional importance of tetraspanins in the regulation of essential HSPC processes including quiescence, migration, and niche adhesion. When taken together, studies outlined in this review suggest that several tetraspanins may serve as potential targets to modulate HSPC interactions with the BM niche, ultimately impacting future HSPC therapies.
Collapse
Affiliation(s)
- Victoria D Balise
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Chelsea A Saito-Reis
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States
| | - Jennifer M Gillette
- Department of Pathology, University of New Mexico Health Sciences Center, Albuquerque, NM, United States.,Comprehensive Cancer Center, The University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
7
|
Florian MC, Klose M, Sacma M, Jablanovic J, Knudson L, Nattamai KJ, Marka G, Vollmer A, Soller K, Sakk V, Cabezas-Wallscheid N, Zheng Y, Mulaw MA, Glauche I, Geiger H. Aging alters the epigenetic asymmetry of HSC division. PLoS Biol 2018; 16:e2003389. [PMID: 30235201 PMCID: PMC6168157 DOI: 10.1371/journal.pbio.2003389] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 10/02/2018] [Accepted: 08/23/2018] [Indexed: 01/01/2023] Open
Abstract
Hematopoietic stem cells (HSCs) balance self-renewal and differentiation to maintain homeostasis. With aging, the frequency of polar HSCs decreases. Cell polarity in HSCs is controlled by the activity of the small RhoGTPase cell division control protein 42 (Cdc42). Here we demonstrate—using a comprehensive set of paired daughter cell analyses that include single-cell 3D confocal imaging, single-cell transplants, single-cell RNA-seq, and single-cell transposase-accessible chromatin sequencing (ATAC-seq)—that the outcome of HSC divisions is strongly linked to the polarity status before mitosis, which is in turn determined by the level of the activity Cdc42 in stem cells. Aged apolar HSCs undergo preferentially self-renewing symmetric divisions, resulting in daughter stem cells with reduced regenerative capacity and lymphoid potential, while young polar HSCs undergo preferentially asymmetric divisions. Mathematical modeling in combination with experimental data implies a mechanistic role of the asymmetric sorting of Cdc42 in determining the potential of daughter cells via epigenetic mechanisms. Therefore, molecules that control HSC polarity might serve as modulators of the mode of stem cell division regulating the potential of daughter cells. Stem cells are unique cells that can differentiate to produce more stem cells or other types of cells and can divide both symmetrically (to produce daughter cells with the same fate) and asymmetrically (to produce one daughter cell that retains stem cell potential and one that differentiates). The mechanisms that control the outcome of stem cell divisions have been the focus of many studies; however, they remain mainly unknown. Here, we have analyzed these mechanisms in murine hematopoietic stem cells (HSCs) by directly comparing the epigenetic signature, the transcriptome, and the function of the two daughter cells stemming from the first division of either a young or an aged HSC. We observe that, while young HSCs divide mainly asymmetrically, aged HSCs divide primarily symmetrically. We find that the mode of division is tightly linked to stem cell polarity and is regulated by the activity level of the small RhoGTPase cell division control protein 42 (Cdc42). In addition, we show that the potential of daughter cells is further linked to the amount of the epigenetic mark H4K16ac and also to the amount of open chromatin allocated to a daughter cell, but it is not linked to its transcriptome. In summary, our study suggests that HSC polarity linked to Cdc42 activity drives the mode of division, while epigenetic mechanisms determine the functional outcome of the stem cell division.
Collapse
Affiliation(s)
- M. Carolina Florian
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
- * E-mail: (MCF); (HG)
| | - Markus Klose
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Mehmet Sacma
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
| | - Jelena Jablanovic
- Max Planck Institute (MPI) of Immunobiology and Epigenetics, Freiburg, Germany
| | - Luke Knudson
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
| | - Kalpana J. Nattamai
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Gina Marka
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
| | - Angelika Vollmer
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
| | - Karin Soller
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
| | - Vadim Sakk
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
| | | | - Yi Zheng
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
| | - Medhanie A. Mulaw
- Institute of Experimental Cancer Research, Medical Faculty, University of Ulm, Ulm, Germany
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Carl Gustav Carus Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Hartmut Geiger
- Institute of Molecular Medicine and Stem Cell Aging, University of Ulm, Ulm, Germany
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, United States of America
- * E-mail: (MCF); (HG)
| |
Collapse
|
8
|
The stem cell regulator PEDF is dispensable for maintenance and function of hematopoietic stem cells. Sci Rep 2017; 7:10134. [PMID: 28860613 PMCID: PMC5579195 DOI: 10.1038/s41598-017-09452-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/25/2017] [Indexed: 01/31/2023] Open
Abstract
Pigment epithelium derived factor (PEDF), a ubiquitously expressed 50 kDa secreted glycoprotein, was recently discovered to regulate self-renewal of neural stem cells and have a supportive effect on human embryonic stem cell growth. Here, we analyzed expression of PEDF in the murine hematopoietic stem cell (HSC) compartments and found that PEDF is highly expressed in primary long-term HSCs. Therefore, we characterized the hematopoietic system in a knockout mouse model for PEDF and using this model we surprisingly found that PEDF is dispensable for HSC regulation. PEDF knockout mice exhibit normal hematopoiesis in steady state conditions and the absence of PEDF lead to normal regeneration capacity in a serial competitive transplantation setting. Additionally, PEDF-deficient cells exhibit unaltered lineage distribution upon serial transplantations. When human cord blood stem and progenitor cells were cultured in media supplemented with recombinant PEDF they did not show changes in growth potential. Taken together, we report that PEDF is not a critical regulatory factor for HSC function during regeneration in vivo or growth of human stem/progenitor cells in vitro.
Collapse
|
9
|
Tyagi A, Vishnoi K, Kaur H, Srivastava Y, Roy BG, Das BC, Bharti AC. Cervical cancer stem cells manifest radioresistance: Association with upregulated AP-1 activity. Sci Rep 2017; 7:4781. [PMID: 28684765 PMCID: PMC5500478 DOI: 10.1038/s41598-017-05162-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 05/25/2017] [Indexed: 12/16/2022] Open
Abstract
Transcription factor AP-1 plays a central role in HPV-mediated cervical carcinogenesis. AP-1 has also been implicated in chemo-radio-resistance but the mechanism(s) remained unexplored. In the present study, cervical cancer stem-like cells (CaCxSLCs) isolated and enriched from cervical cancer cell lines SiHa and C33a demonstrated an elevated AP-1 DNA-binding activity in comparison to non-stem cervical cancer cells. Upon UV-irradiation, CaCxSLCs showed a UV exposure duration-dependent higher proliferation and highly increased AP-1 activity whereas it was completely abolished in non-stem cancer cells. CaCxSLCs also showed differential overexpression of c-Fos and c-Jun at transcript as well as in protein level. The loss of AP-1 activity and expression was accompanied by decrease in cell viability and proliferation in UV-irradiated non-stem cancer cells. Interestingly, CaCxSLCs treated with curcumin prior to UV-irradiation abolished AP-1 activity and a concomitant reduction in SP cells leading to abrogation of sphere forming ability, loss of proliferation, induction of apoptosis and the cells were poorly tumorigenic. The curcumin pre-treatment abolished the expression of c-Fos and c-Jun but upregulated Fra-1 expression in UV-irradiated CaCxSLCs. Thus, the study suggests a critical role of AP-1 protein in the manifestation of radioresistance but targeting with curcumin helps in radiosensitizing CaCxSLCs through upregulation of Fra-1.
Collapse
Affiliation(s)
- Abhishek Tyagi
- Molecular Oncology Laboratory, B.R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, 110007, India.,Division of Molecular Oncology, National Institute of Cancer Prevention and Research (NICPR), Noida, 201301, Uttar Pradesh, India.,Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India
| | - Kanchan Vishnoi
- Division of Molecular Oncology, National Institute of Cancer Prevention and Research (NICPR), Noida, 201301, Uttar Pradesh, India
| | - Harsimrut Kaur
- Molecular Oncology Laboratory, B.R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, 110007, India
| | - Yogesh Srivastava
- Division of Molecular Oncology, National Institute of Cancer Prevention and Research (NICPR), Noida, 201301, Uttar Pradesh, India
| | - Bal Gangadhar Roy
- Institute of Nuclear Medicine and Allied Sciences, Defence Research Development Organization, Delhi, 110 054, India
| | - Bhudev C Das
- Molecular Oncology Laboratory, B.R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, 110007, India. .,Stem Cell and Cancer Research Lab, Amity Institute of Molecular Medicine and Stem Cell Research (AIMMSCR), Amity University, Noida, Uttar Pradesh, 201313, India.
| | - Alok C Bharti
- Division of Molecular Oncology, National Institute of Cancer Prevention and Research (NICPR), Noida, 201301, Uttar Pradesh, India. .,Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, 110007, India.
| |
Collapse
|
10
|
Jia D, Jolly MK, Harrison W, Boareto M, Ben-Jacob E, Levine H. Operating principles of tristable circuits regulating cellular differentiation. Phys Biol 2017; 14:035007. [PMID: 28443829 DOI: 10.1088/1478-3975/aa6f90] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Many cell-fate decisions during embryonic development are governed by a motif comprised of two transcription factors (TFs) A and B that mutually inhibit each other and may self-activate. This motif, called as a self-activating toggle switch (SATS), can typically have three stable states (phenotypes)-two corresponding to differentiated cell fates, each of which has a much higher level of one TF than the other-[Formula: see text] or [Formula: see text]-and the third state corresponding to an 'undecided' stem-like state with similar levels of both A and B-[Formula: see text]. Furthermore, two or more SATSes can be coupled together in various topologies in different contexts, thereby affecting the coordination between multiple cellular decisions. However, two questions remain largely unanswered: (a) what governs the co-existence and relative stability of these three stable states? (b) What orchestrates the decision-making of coupled SATSes? Here, we first demonstrate that the co-existence and relative stability of the three stable states in an individual SATS can be governed by the relative strength of self-activation, external signals activating and/or inhibiting A and B, and mutual degradation between A and B. Simultaneously, we investigate the effects of these factors on the decision-making of two coupled SATSes. Our results offer novel understanding into the operating principles of individual and coupled tristable self-activating toggle switches (SATSes) regulating cellular differentiation and can yield insights into synthesizing three-way genetic circuits and understanding of cellular reprogramming.
Collapse
Affiliation(s)
- Dongya Jia
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005-1827, United States of America. Program in Systems, Synthetic and Physical Biology, Rice University, Houston, TX 77005-1827, United States of America
| | | | | | | | | | | |
Collapse
|
11
|
Tyagi A, Vishnoi K, Mahata S, Verma G, Srivastava Y, Masaldan S, Roy BG, Bharti AC, Das BC. Cervical Cancer Stem Cells Selectively Overexpress HPV Oncoprotein E6 that Controls Stemness and Self-Renewal through Upregulation of HES1. Clin Cancer Res 2016; 22:4170-84. [PMID: 26988248 DOI: 10.1158/1078-0432.ccr-15-2574] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Accepted: 02/19/2016] [Indexed: 11/16/2022]
Abstract
PURPOSE Perturbation of keratinocyte differentiation by E6/E7 oncoproteins of high-risk human papillomaviruses that drive oncogenic transformation of cells in squamocolumnar junction of the uterine cervix may confer "stem-cell like" characteristics. However, the crosstalk between E6/E7 and stem cell signaling during cervical carcinogenesis is not well understood. We therefore examined the role of viral oncoproteins in stem cell signaling and maintenance of stemness in cervical cancer. EXPERIMENTAL DESIGN Isolation and enrichment of cervical cancer stem-like cells (CaCxSLCs) was done from cervical primary tumors and cancer cell lines by novel sequential gating using a set of functional and phenotypic markers (ABCG2, CD49f, CD71, CD133) in defined conditioned media for assessing sphere formation and expression of self-renewal and stemness markers by FACS, confocal microscopy, and qRT-PCR. Differential expression level and DNA-binding activity of Notch1 and its downstream targets in CaCxSLCs as well as silencing of HPVE6/Hes1 by siRNA was evaluated by gel retardation assay, FACS, immunoblotting, and qRT-PCR followed by in silico and in vivo xenograft analysis. RESULTS CaCxSLCs showed spheroid-forming ability, expressed self-renewal and stemness markers Oct4, Sox2, Nanog, Lrig1, and CD133, and selectively overexpressed E6 and HES1 transcripts in both cervical primary tumors and cancer cell lines. The enriched CaCxSLCs were highly tumorigenic and did recapitulate primary tumor histology in nude mice. siRNA silencing of HPVE6 or Hes1 abolished sphere formation, downregulated AP-1-STAT3 signaling, and induced redifferentiation. CONCLUSIONS Our findings suggest the possible mechanism by which HPVE6 potentially regulate and maintain stem-like cancer cells through Hes1. Clin Cancer Res; 22(16); 4170-84. ©2016 AACR.
Collapse
Affiliation(s)
- Abhishek Tyagi
- Stem Cell and Cancer Research Laboratory, Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Campus, Noida, Uttar Pradesh, India. Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India. Molecular Oncology Laboratory, B.R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India
| | - Kanchan Vishnoi
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Sutapa Mahata
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Gaurav Verma
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Yogesh Srivastava
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Shashank Masaldan
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India
| | - Bal Gangadhar Roy
- Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi, India
| | - Alok C Bharti
- Division of Molecular Oncology, Institute of Cytology & Preventive Oncology (ICMR), Noida, Uttar Pradesh, India. Molecular Oncology Laboratory, Department of Zoology, University of Delhi, Delhi, India.
| | - Bhudev C Das
- Stem Cell and Cancer Research Laboratory, Amity Institute of Molecular Medicine and Stem Cell Research, Amity University Campus, Noida, Uttar Pradesh, India. Molecular Oncology Laboratory, B.R. Ambedkar Centre for Biomedical Research (ACBR), University of Delhi, Delhi, India.
| |
Collapse
|
12
|
Klamer S, Voermans C. The role of novel and known extracellular matrix and adhesion molecules in the homeostatic and regenerative bone marrow microenvironment. Cell Adh Migr 2015; 8:563-77. [PMID: 25482635 PMCID: PMC4594522 DOI: 10.4161/19336918.2014.968501] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Maintenance of haematopoietic stem cells and differentiation of committed progenitors occurs in highly specialized niches. The interactions of haematopoietic stem and progenitor cells (HSPCs) with cells, growth factors and extracellular matrix (ECM) components of the bone marrow (BM) microenvironment control homeostasis of HSPCs. We only start to understand the complexity of the haematopoietic niche(s) that comprises endosteal, arterial, sinusoidal, mesenchymal and neuronal components. These distinct niches produce a broad range of soluble factors and adhesion molecules that modulate HSPC fate during normal hematopoiesis and BM regeneration. Adhesive interactions between HSPCs and the microenvironment will influence their localization and differentiation potential. In this review we highlight the current understanding of the functional role of ECM- and adhesion (regulating) molecules in the haematopoietic niche during homeostatic and regenerative hematopoiesis. This knowledge may lead to the improvement of current cellular therapies and more efficient development of future cellular products.
Collapse
Affiliation(s)
- Sofieke Klamer
- a Department of Hematopoiesis; Sanquin Research; Landsteiner Laboratory; Academic Medical Centre ; University of Amsterdam ; Amsterdam , The Netherlands
| | | |
Collapse
|
13
|
Pina C, Teles J, Fugazza C, May G, Wang D, Guo Y, Soneji S, Brown J, Edén P, Ohlsson M, Peterson C, Enver T. Single-Cell Network Analysis Identifies DDIT3 as a Nodal Lineage Regulator in Hematopoiesis. Cell Rep 2015; 11:1503-10. [PMID: 26051941 PMCID: PMC4528262 DOI: 10.1016/j.celrep.2015.05.016] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 04/02/2015] [Accepted: 05/10/2015] [Indexed: 10/29/2022] Open
Abstract
We explore cell heterogeneity during spontaneous and transcription-factor-driven commitment for network inference in hematopoiesis. Since individual genes display discrete OFF states or a distribution of ON levels, we compute and combine pairwise gene associations from binary and continuous components of gene expression in single cells. Ddit3 emerges as a regulatory node with positive linkage to erythroid regulators and negative association with myeloid determinants. Ddit3 loss impairs erythroid colony output from multipotent cells, while forcing Ddit3 in granulo-monocytic progenitors (GMPs) enhances self-renewal and impedes differentiation. Network analysis of Ddit3-transduced GMPs reveals uncoupling of myeloid networks and strengthening of erythroid linkages. RNA sequencing suggests that Ddit3 acts through development or stabilization of a precursor upstream of GMPs with inherent Meg-E potential. The enrichment of Gata2 target genes in Ddit3-dependent transcriptional responses suggests that Ddit3 functions in an erythroid transcriptional network nucleated by Gata2.
Collapse
Affiliation(s)
- Cristina Pina
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK
| | - José Teles
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK; Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, 223 62 Lund, Sweden
| | - Cristina Fugazza
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK
| | - Gillian May
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK
| | - Dapeng Wang
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK
| | - Yanping Guo
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK
| | - Shamit Soneji
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK
| | - John Brown
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK
| | - Patrik Edén
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, 223 62 Lund, Sweden
| | - Mattias Ohlsson
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, 223 62 Lund, Sweden
| | - Carsten Peterson
- Computational Biology and Biological Physics, Department of Astronomy and Theoretical Physics, Lund University, 223 62 Lund, Sweden
| | - Tariq Enver
- Stem Cell Laboratory, UCL Cancer Institute, University College London, London W1CE 6BT, UK.
| |
Collapse
|
14
|
Modeling and visualizing cell type switching. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2014; 2014:293980. [PMID: 24834107 PMCID: PMC4009162 DOI: 10.1155/2014/293980] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 12/20/2013] [Accepted: 01/10/2014] [Indexed: 01/13/2023]
Abstract
Understanding cellular differentiation is critical in explaining development and for taming diseases such as cancer. Differentiation is conventionally represented using bifurcating lineage trees. However, these lineage trees cannot readily capture or quantify all the types of transitions now known to occur between cell types, including transdifferentiation or differentiation off standard paths. This work introduces a new analysis and visualization technique that is capable of representing all possible transitions between cell states compactly, quantitatively, and intuitively. This method considers the regulatory network of transcription factors that control cell type determination and then performs an analysis of network dynamics to identify stable expression profiles and the potential cell types that they represent. A visualization tool called CellDiff3D creates an intuitive three-dimensional graph that shows the overall direction and probability of transitions between all pairs of cell types within a lineage. In this study, the influence of gene expression noise and mutational changes during myeloid cell differentiation are presented as a demonstration of the CellDiff3D technique, a new approach to quantify and envision all possible cell state transitions in any lineage network.
Collapse
|
15
|
Ishihara J, Umemoto T, Yamato M, Shiratsuchi Y, Takaki S, Petrich BG, Nakauchi H, Eto K, Kitamura T, Okano T. Nov/CCN3 regulates long-term repopulating activity of murine hematopoietic stem cells via integrin αvβ3. Int J Hematol 2014; 99:393-406. [PMID: 24563081 DOI: 10.1007/s12185-014-1534-x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2014] [Revised: 01/29/2014] [Accepted: 01/30/2014] [Indexed: 01/01/2023]
Abstract
Throughout life, hematopoietic stem cells (HSCs) sustain the blood cell supply through their capacities for self-renewal and multilineage differentiation. These processes are regulated within a specialized microenvironment termed the 'niche'. Here, we show a novel mechanism for regulating HSC function that is mediated by nephroblastoma overexpressed (Nov/CCN3), a matricellular protein member of the CCN family. We found that Nov contributes to the maintenance of long-term repopulating (LTR) activity through association with integrin αvβ3 on HSCs. The resultant β3 integrin outside-in signaling is dependent on thrombopoietin (TPO), a crucial cytokine involved in HSC maintenance. TPO was required for Nov binding to integrin αvβ3, and stimulated Nov expression in HSCs. However, in the presence of IFNγ, a cytokine known to impair HSC function, not only was TPO-induced expression of Nov suppressed, but the LTR activity was conversely impaired by TPO-mediated ligation of integrin αvβ3 with exogenous ligands, including Nov, as well. Thus, Nov/integrin αvβ3-mediated maintenance of HSCs appears to be modulated by simultaneous stimulation by other cytokines. Our finding suggests that this system contributes to the regulation of HSCs within the bone marrow niche.
Collapse
Affiliation(s)
- Jun Ishihara
- Institute of Advanced Biomedical Engineering and Science, Tokyo Women's Medical University, 8-1 Kawadacho, Shinjuku-ku, Tokyo, 162-8666, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Abstract
Polarization of cell phenotypes, a common strategy to achieve cell type diversity in metazoa, results from binary cell-fate decisions in the branching pedigree of development. Such "either-or" fate decisions are controlled by two opposing cell fate-determining transcription factors. Each of the two distinct "master regulators" promotes differentiation of its respective sister lineage. But they also suppress one other, leading to their mutually exclusive expression in the two ensuing lineages. Thus, promiscuous coexistence of the antagonist regulators in the same cell, the hallmark of the common "undecided" progenitor of two sister lineages, is considered unstable. This antagonism ensures robust polarization into two discretely distinct cell types. But now the immune system's T-helper (Th) cells and their two canonical subtypes, Th1 and Th2 cells, tell a different story, as revealed in three papers recently published in PLOS Biology. The intermediate state that co-expresses the two opposing master regulators of the Th1 and Th2 subtypes, T-bet and Gata3, is highly stable and is not necessarily an undecided precursor. Instead, the Th1/Th2 hybrid cell is a robust new type with properties of both Th1 and Th2 cells. These hybrid cells are functionally active and possess the benefit of moderation: self-limitation of effector T cell function to prevent excessive inflammation, a permanent risk in host defense that can cause tissue damage or autoimmunity. Gene regulatory network analysis suggests that stabilization of the intermediate center in a polarizing system can be achieved by minor tweaking of the architecture of the mutual suppression gene circuit, and thus is a design option readily available to evolution.
Collapse
Affiliation(s)
- Sui Huang
- Institute for Systems Biology, Seattle, Washington, USA.
| |
Collapse
|
17
|
The Tetraspanin CD9 Affords High-Purity Capture of All Murine Hematopoietic Stem Cells. Cell Rep 2013; 4:642-8. [DOI: 10.1016/j.celrep.2013.07.020] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 06/05/2013] [Accepted: 07/17/2013] [Indexed: 11/17/2022] Open
|
18
|
Canals-Hamann AZ, das Neves RP, Reittie JE, Iñiguez C, Soneji S, Enver T, Buckle VJ, Iborra FJ. A biophysical model for transcription factories. BMC BIOPHYSICS 2013; 6:2. [PMID: 23394119 PMCID: PMC3740778 DOI: 10.1186/2046-1682-6-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 12/05/2012] [Indexed: 02/07/2023]
Abstract
Summary Transcription factories are nuclear domains where gene transcription takes place
although the molecular basis for their formation and maintenance are unknown. In this
study, we explored how the properties of chromatin as a polymer may contribute to the
structure of transcription factories. We found that transcriptional active chromatin
contains modifications like histone H4 acetylated at Lysine 16 (H4K16ac). Single
fibre analysis showed that this modification spans the entire body of the gene.
Furthermore, H4K16ac genes cluster in regions up to 500 Kb alternating active and
inactive chromatin. The introduction of H4K16ac in chromatin induces stiffness in the
chromatin fibre. The result of this change in flexibility is that chromatin could
behave like a multi-block copolymer with repetitions of stiff-flexible
(active-inactive chromatin) components. Copolymers with such structure self-organize
through spontaneous phase separation into microdomains. Consistent with such model
H4K16ac chromatin form foci that associates with nascent transcripts. We propose that
transcription factories are the result of the spontaneous concentration of H4K16ac
chromatin that are in proximity, mainly in cis.
Collapse
Affiliation(s)
- Ana Z Canals-Hamann
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford, OX3 9DS, UK.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Oh IH, Humphries RK. Concise review: Multidimensional regulation of the hematopoietic stem cell state. Stem Cells 2012; 30:82-8. [PMID: 22083966 DOI: 10.1002/stem.776] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hematopoietic stem cells (HSCs) are characterized by their unique function to produce all lineages of blood cells throughout life. Such tissue-specific function of HSC is attributed to their ability to execute self-renewal and multilineage differentiation. Accumulating evidence indicates that the undifferentiated state of HSC is characterized by dynamic maintenance of chromatin structures and epigenetic plasticity. Conversely, quiescence, self-renewal, and differentiation of HSCs are dictated by complex regulatory mechanisms involving specific transcription factors and microenvironmental crosstalk between stem cells and multiple compartments of niches in bone marrows. Thus, multidimensional regulatory inputs are integrated into two opposing characters of HSCs-maintenance of undifferentiated state analogous to pluripotent stem cells but execution of tissue-specific hematopoietic functions. Further studies on the interplay of such regulatory forces as "cell fate determinant" will likely shed the light on diverse spectrums of tissue-specific stem cells.
Collapse
Affiliation(s)
- Il-Hoan Oh
- Catholic High Performance Cell Therapy Center and Department of Medical Lifescience, The Catholic University of Korea, Seoul, Korea.
| | | |
Collapse
|
20
|
SPARC is dispensable for murine hematopoiesis, despite its suspected pathophysiological role in 5q-myelodysplastic syndrome. Leukemia 2012; 26:2416-9. [DOI: 10.1038/leu.2012.97] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
21
|
Transient noise amplification and gene expression synchronization in a bistable mammalian cell-fate switch. Cell Rep 2012; 1:215-24. [PMID: 22832195 DOI: 10.1016/j.celrep.2012.01.007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 01/17/2012] [Accepted: 01/31/2012] [Indexed: 11/21/2022] Open
Abstract
Progenitor cells within a clonal population show variable proclivity toward lineage commitment and differentiation. This cell-to-cell variability has been attributed to transcriptome-wide gene expression noise generated by fluctuations in the amount of cellular machinery and stochasticity in the biochemical reactions involved in protein synthesis. It therefore remains unclear how a signaling network, in the presence of such noise, can execute unequivocal cell-fate decisions from external cues. Here, we use mathematical modeling and model-guided experiments to reveal functional interplay between instructive signaling and noise in erythropoiesis. We present evidence that positive transcriptional feedback loops in a lineage-specific receptor signaling pathway can generate ligand-induced memory to engender robust, switch-like responses. These same feedback loops can also transiently amplify gene expression noise in the signaling network, suggesting that external cues can actually bias seemingly stochastic decisions during cell-fate specification. Gene expression levels among key effectors in the signaling pathway are uncorrelated in the initial population of progenitor cells but become synchronized after addition of ligand, which activates the transcriptional feedback loops. Finally, we show that this transient noise amplification and gene expression synchronization induced by ligand can directly influence cell survival and differentiation kinetics within the population.
Collapse
|
22
|
Fijneman RJA, Anderson RA, Richards E, Liu J, Tijssen M, Meijer GA, Anderson J, Rod A, O'Sullivan MG, Scott PM, Cormier RT. Runx1 is a tumor suppressor gene in the mouse gastrointestinal tract. Cancer Sci 2012; 103:593-599. [PMID: 22171576 PMCID: PMC5439111 DOI: 10.1111/j.1349-7006.2011.02189.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The Runx1 transcription factor plays an important role in tissue homeostasis through its effects on stem/progenitor cell populations and differentiation. The effect of Runx1 on epithelial differentiation of the secretory cell lineage of the colon was recently demonstrated. This study aimed to examine the role of Runx1 in tumor development in epithelial cells of the gastrointestinal tract. Conditional knockout mice that lacked Runx1 expression in epithelial cells of the GI tract were generated. These mice were crossed onto the Apc(Min) background, killed and their intestinal tumor phenotypes were compared with Apc(Min) Runx1 wild-type control mice. Apc-wild-type Runx1-mutant mice were also examined for tumor development. Colons from Runx1 knockout and wild-type mice were used for genome-wide mRNA expression analyses followed by gene-specific quantitative RT-PCR of whole colon and colon epithelium to identify Runx1 target genes. Runx1 deficiency in intestinal epithelial cells significantly enhanced tumorigenesis in Apc(Min) mice. Notably, epithelial Runx1 deficiency in Apc-wild-type mice was sufficient to cause tumor development. Absence of Runx1 was associated with global changes in the expression of genes involved in inflammation and intestinal metabolism, and with gene sets indicative of a metastatic phenotype and poor prognosis. Gene-specific analysis of Runx1-deficient colon epithelium revealed increased expression of genes linked to an expansion of the stem/progenitor cell population. These results identify Runx1 as a novel tumor suppressor gene for gastrointestinal tumors and support a role for Runx1 in maintaining the balance between the intestinal stem/progenitor cell population and epithelial differentiation of the GI tract.
Collapse
Affiliation(s)
- Remond J A Fijneman
- Department of Pathology, VU University Medical Center, Amsterdam, Netherlands
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Although the nonrandom nature of interphase chromosome arrangement is widely accepted, how nuclear organization relates to genomic function remains unclear. Nuclear subcompartments may play a role by offering rich microenvironments that regulate chromatin state and ensure optimal transcriptional efficiency. Technological advances now provide genome-wide and four-dimensional analyses, permitting global characterizations of nuclear order. These approaches will help uncover how seemingly separate nuclear processes may be coupled and aid in the effort to understand the role of nuclear organization in development and disease.
Collapse
Affiliation(s)
- Indika Rajapakse
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | |
Collapse
|
24
|
de la Casa-Esperón E. From mammals to viruses: the Schlafen genes in developmental, proliferative and immune processes. Biomol Concepts 2011; 2:159-69. [DOI: 10.1515/bmc.2011.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Accepted: 04/29/2011] [Indexed: 11/15/2022] Open
Abstract
AbstractThe Schlafen genes have been associated with proliferation control and with several differentiation processes, as well as with disparate phenotypes such as immune response, embryonic lethality and meiotic drive. They constitute a gene family with widespread distribution in mammals, where they are expressed in several tissues, predominantly those of the immune system. Moreover, horizontal transfer of these genes to orthopoxviruses suggests a role of the viral Schlafens in evasion to the host immune response. The expression and functional studies of this gene family will be reviewed under the prism of their evolution and diversification, the challenges they pose and the future avenues of research.
Collapse
Affiliation(s)
- Elena de la Casa-Esperón
- 1Albacete Science and Technology Park, Regional Center for Biomedical Research (C.R.I.B.) at the University of Castilla-La Mancha, C/Almansa 14, 02006 Albacete, Spain
| |
Collapse
|
25
|
Abstract
Understanding the pattern of gene expression during erythropoiesis is crucial for a synthesis of erythroid developmental biology. Here, we isolated 4 distinct populations at successive erythropoietin-dependent stages of erythropoiesis, including the terminal, pyknotic stage. The transcriptome was determined using Affymetrix arrays. First, we demonstrated the importance of using defined cell populations to identify lineage and temporally specific patterns of gene expression. Cells sorted by surface expression profile not only express significantly fewer genes than unsorted cells but also demonstrate significantly greater differences in the expression levels of particular genes between stages than unsorted cells. Second, using standard software, we identified more than 1000 transcripts not previously observed to be differentially expressed during erythroid maturation, 13 of which are highly significantly terminally regulated, including RFXAP and SMARCA4. Third, using matched filtering, we identified 12 transcripts not previously reported to be continuously up-regulated in maturing human primary erythroblasts. Finally, using transcription factor binding site analysis, we identified potential transcription factors that may regulate gene expression during terminal erythropoiesis. Our stringent lists of differentially regulated and continuously expressed transcripts containing many genes with undiscovered functions in erythroblasts are a resource for future functional studies of erythropoiesis. Our Human Erythroid Maturation database is available at https://cellline.molbiol.ox.ac.uk/eryth/index.html. [corrected].
Collapse
|
26
|
Macrophage activation and differentiation signals regulate schlafen-4 gene expression: evidence for Schlafen-4 as a modulator of myelopoiesis. PLoS One 2011; 6:e15723. [PMID: 21249125 PMCID: PMC3017543 DOI: 10.1371/journal.pone.0015723] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2010] [Accepted: 11/28/2010] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The ten mouse and six human members of the Schlafen (Slfn) gene family all contain an AAA domain. Little is known of their function, but previous studies suggest roles in immune cell development. In this report, we assessed Slfn regulation and function in macrophages, which are key cellular regulators of innate immunity. METHODOLOGY/PRINCIPAL FINDINGS Multiple members of the Slfn family were up-regulated in mouse bone marrow-derived macrophages (BMM) by the Toll-like Receptor (TLR)4 agonist lipopolysaccharide (LPS), the TLR3 agonist Poly(I∶C), and in disease-affected joints in the collagen-induced model of rheumatoid arthritis. Of these, the most inducible was Slfn4. TLR agonists that signal exclusively through the MyD88 adaptor protein had more modest effects on Slfn4 mRNA levels, thus implicating MyD88-independent signalling and autocrine interferon (IFN)-β in inducible expression. This was supported by the substantial reduction in basal and LPS-induced Slfn4 mRNA expression in IFNAR-1⁻/⁻ BMM. LPS causes growth arrest in macrophages, and other Slfn family genes have been implicated in growth control. Slfn4 mRNA levels were repressed during macrophage colony-stimulating factor (CSF-1)-mediated differentiation of bone marrow progenitors into BMM. To determine the role of Slfn4 in vivo, we over-expressed the gene specifically in macrophages in mice using a csf1r promoter-driven binary expression system. Transgenic over-expression of Slfn4 in myeloid cells did not alter macrophage colony formation or proliferation in vitro. Monocyte numbers, as well as inflammatory macrophages recruited to the peritoneal cavity, were reduced in transgenic mice that specifically over-expressed Slfn4, while macrophage numbers and hematopoietic activity were increased in the livers and spleens. CONCLUSIONS Slfn4 mRNA levels were up-regulated during macrophage activation but down-regulated during differentiation. Constitutive Slfn4 expression in the myeloid lineage in vivo perturbs myelopoiesis. We hypothesise that the down-regulation of Slfn4 gene expression during macrophage differentiation is a necessary step in development of this lineage.
Collapse
|
27
|
Rajapakse I, Scalzo D, Tapscott SJ, Kosak ST, Groudine M. Networking the nucleus. Mol Syst Biol 2010; 6:395. [PMID: 20664641 PMCID: PMC2925527 DOI: 10.1038/msb.2010.48] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Accepted: 05/31/2010] [Indexed: 12/13/2022] Open
Abstract
The nuclei of differentiating cells exhibit several fundamental principles of self-organization. They are composed of many dynamical units connected physically and functionally to each other—a complex network—and the different parts of the system are mutually adapted and produce a characteristic end state. A unique cell-specific signature emerges over time from complex interactions among constituent elements that delineate coordinate gene expression and chromosome topology. Each element itself consists of many interacting components, all dynamical in nature. Self-organizing systems can be simplified while retaining complex information using approaches that examine the relationship between elements, such as spatial relationships and transcriptional information. These relationships can be represented using well-defined networks. We hypothesize that during the process of differentiation, networks within the cell nucleus rewire according to simple rules, from which a higher level of order emerges. Studying the interaction within and among networks provides a useful framework for investigating the complex organization and dynamic function of the nucleus.
Collapse
Affiliation(s)
- Indika Rajapakse
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | |
Collapse
|
28
|
Wang J, Xu L, Wang E, Huang S. The potential landscape of genetic circuits imposes the arrow of time in stem cell differentiation. Biophys J 2010; 99:29-39. [PMID: 20655830 DOI: 10.1016/j.bpj.2010.03.058] [Citation(s) in RCA: 145] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2009] [Revised: 03/30/2010] [Accepted: 03/19/2010] [Indexed: 01/18/2023] Open
Abstract
Differentiation from a multipotent stem or progenitor state to a mature cell is an essentially irreversible process. The associated changes in gene expression patterns exhibit time-directionality. This "arrow of time" in the collective change of gene expression across multiple stable gene expression patterns (attractors) is not explained by the regulated activation, the suppression of individual genes which are bidirectional molecular processes, or by the standard dynamical models of the underlying gene circuit which only account for local stability of attractors. To capture the global dynamics of this nonequilibrium system and gain insight in the time-asymmetry of state transitions, we computed the quasipotential landscape of the stochastic dynamics of a canonical gene circuit that governs branching cell fate commitment. The potential landscape reveals the global dynamics and permits the calculation of potential barriers between cell phenotypes imposed by the circuit architecture. The generic asymmetry of barrier heights indicates that the transition from the uncommitted multipotent state to differentiated states is inherently unidirectional. The model agrees with observations and predicts the extreme conditions for reprogramming cells back to the undifferentiated state.
Collapse
Affiliation(s)
- Jin Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, China.
| | | | | | | |
Collapse
|
29
|
Kimura 木村丹香子 A, Martin C, Robinson GW, Simone JM, Chen W, Wickre MC, O'Shea JJ, Hennighausen L. The gene encoding the hematopoietic stem cell regulator CCN3/NOV is under direct cytokine control through the transcription factors STAT5A/B. J Biol Chem 2010; 285:32704-32709. [PMID: 20720003 DOI: 10.1074/jbc.m110.141804] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cytokines control the biology of hematopoietic stem cells (HSCs) and progenitor cells in part through the transcription factors STAT5A/B. To investigate the target genes of STAT5A/B activated by cytokines in HSCs and progenitors, we performed microarray analyses using Lineage(-) Sca-1(+) c-Kit(+) (KSL) cells in the presence and absence of STAT5A/B. Stimulation with a mixture containing IL-3, IL-6, stem cell factor, thrombopoietin, and Flt3 ligand induced Ccn3/Nov mRNA over 100-fold in WT (control) but not Stat5a/b-null KSL cells. CCN3/NOV is a positive regulator of human HSC self-renewal and development of committed blood cells. Without stimulation, the Ccn3/Nov signal level was low in control KSL cells similar to Stat5a/b-null KSL cells. To determine which cytokine activates the Ccn3/Nov gene, we analyzed Lineage(-) c-Kit(+) (KL) and 32D cells using quantitative PCR and ChIP assays. Although stimulation with a mixture lacking IL-3 prevented the induction of Ccn3/Nov in control KL cells, IL-3 alone could induce Ccn3/Nov mRNA in control KL and 32D cells. ChIP assays using 32D cells revealed IL-3-induced binding of STAT5A/B to a γ-interferon-activated sequences site in the Ccn3/Nov gene promoter. This is the first report that Ccn3/Nov is directly induced by cytokines through STAT5A/B.
Collapse
Affiliation(s)
- Akiko Kimura 木村丹香子
- From the Laboratory of Genetics and Physiology, NIDDK, Bethesda, Maryland 20892.
| | - Cyril Martin
- From the Laboratory of Genetics and Physiology, NIDDK, Bethesda, Maryland 20892
| | - Gertraud W Robinson
- From the Laboratory of Genetics and Physiology, NIDDK, Bethesda, Maryland 20892
| | - James M Simone
- Flow Cytometry Section, Office of Science and Technology, NIAMS, Bethesda, Maryland 20892
| | - Weiping Chen
- Microarray Core Facility with the Genomic Core Laboratory, NIDDK, Bethesda, Maryland 20892
| | - Mark C Wickre
- From the Laboratory of Genetics and Physiology, NIDDK, Bethesda, Maryland 20892
| | - John J O'Shea
- Molecular Immunology and Inflammation Branch, NIAMS, National Institutes of Health, Bethesda, Maryland 20892
| | - Lothar Hennighausen
- From the Laboratory of Genetics and Physiology, NIDDK, Bethesda, Maryland 20892
| |
Collapse
|
30
|
Pasini D, Malatesta M, Jung HR, Walfridsson J, Willer A, Olsson L, Skotte J, Wutz A, Porse B, Jensen ON, Helin K. Characterization of an antagonistic switch between histone H3 lysine 27 methylation and acetylation in the transcriptional regulation of Polycomb group target genes. Nucleic Acids Res 2010; 38:4958-69. [PMID: 20385584 PMCID: PMC2926606 DOI: 10.1093/nar/gkq244] [Citation(s) in RCA: 280] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Polycomb group (PcG) proteins are transcriptional repressors, which regulate proliferation and cell fate decisions during development, and their deregulated expression is a frequent event in human tumours. The Polycomb repressive complex 2 (PRC2) catalyzes trimethylation (me3) of histone H3 lysine 27 (K27), and it is believed that this activity mediates transcriptional repression. Despite the recent progress in understanding PcG function, the molecular mechanisms by which the PcG proteins repress transcription, as well as the mechanisms that lead to the activation of PcG target genes are poorly understood. To gain insight into these mechanisms, we have determined the global changes in histone modifications in embryonic stem (ES) cells lacking the PcG protein Suz12 that is essential for PRC2 activity. We show that loss of PRC2 activity results in a global increase in H3K27 acetylation. The methylation to acetylation switch correlates with the transcriptional activation of PcG target genes, both during ES cell differentiation and in MLL-AF9-transduced hematopoietic stem cells. Moreover, we provide evidence that the acetylation of H3K27 is catalyzed by the acetyltransferases p300 and CBP. Based on these data, we propose that the PcG proteins in part repress transcription by preventing the binding of acetyltransferases to PcG target genes.
Collapse
Affiliation(s)
- Diego Pasini
- Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Nakanishi T, Chumsri S, Khakpour N, Brodie AH, Leyland-Jones B, Hamburger AW, Ross DD, Burger AM. Side-population cells in luminal-type breast cancer have tumour-initiating cell properties, and are regulated by HER2 expression and signalling. Br J Cancer 2010; 102:815-26. [PMID: 20145614 PMCID: PMC2833247 DOI: 10.1038/sj.bjc.6605553] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2009] [Revised: 12/21/2009] [Accepted: 12/22/2009] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND The expression of side-population (SP) cells and their relation to tumour-initiating cells (T-ICs) have been insufficiently studied in breast cancer (BC). We therefore evaluated primary cell cultures derived from patients and a panel of human BC cell lines with luminal- or basal-molecular signatures for the presence of SP and BC stem cell markers. METHODS The SPs from luminal-type BC were analysed for BC T-IC characteristics, including human epidermal growth factor receptor 2 (HER2), ERalpha, IGFBP7 expression and their ability to initiate tumours in non-obese diabetic severe combined immunodeficiency (NOD/SCID) mice. Pharmacological modulators were used to assess the effects of HER2 signalling and breast cancer-resistance protein (BCRP) expression on SPs. RESULTS The SP was more prevalent in the luminal subtype of BC compared with the basal subtype. HER2 expression was significantly correlated with the occurrence of an SP (r(2)=0.75, P=0.0003). Disappearance of SP in the presence of Ko143, a specific inhibitor of the ATP-binding cassette transporter BCRP, suggests that BCRP is the predominant transporter expressed in this population. The SP also decreased in the presence of HER2 signalling inhibitors AG825 or trastuzumab, strengthening the notion that HER2 contributed to the SP phenotype, likely through downstream AKT signalling. The SP cells from luminal-type MCF-7 cells with enforced expression of HER2, and primary cells with luminal-like properties from a BC patient, displayed enrichment in cells capable of repopulating tumours in NOD/SCID mice. Engraftment of SP cells was inhibited by pretreatment with AG825 or by in vivo treatment with trastuzumab. INTERPRETATION Our findings indicate an important role of HER2 in regulating SP and hence T-ICs in BC, which may account for the poor responsiveness of HER2-positive BCs to chemotherapy, as well as their aggressiveness.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily G, Member 2
- ATP-Binding Cassette Transporters/antagonists & inhibitors
- ATP-Binding Cassette Transporters/metabolism
- Animals
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal, Humanized
- Blotting, Western
- Breast Neoplasms/metabolism
- Breast Neoplasms/pathology
- Drug Resistance, Neoplasm
- Female
- Flow Cytometry
- Humans
- Insulin-Like Growth Factor Binding Proteins/antagonists & inhibitors
- Insulin-Like Growth Factor Binding Proteins/metabolism
- Mice
- Mice, Inbred NOD
- Mice, Nude
- Mice, SCID
- Neoplasm Proteins/antagonists & inhibitors
- Neoplasm Proteins/metabolism
- Neoplastic Stem Cells/pathology
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, ErbB-2/antagonists & inhibitors
- Receptor, ErbB-2/metabolism
- Signal Transduction
- Trastuzumab
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- T Nakanishi
- Departments of Medicine, Pathology, Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, Marlene and Stewart Greenebaum Cancer Center (UMGCC), Baltimore, MD, USA
| | - S Chumsri
- Departments of Medicine, Pathology, Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, Marlene and Stewart Greenebaum Cancer Center (UMGCC), Baltimore, MD, USA
| | - N Khakpour
- Departments of Medicine, Pathology, Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, Marlene and Stewart Greenebaum Cancer Center (UMGCC), Baltimore, MD, USA
| | - A H Brodie
- Departments of Medicine, Pathology, Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, Marlene and Stewart Greenebaum Cancer Center (UMGCC), Baltimore, MD, USA
| | - B Leyland-Jones
- Department of Hematology and Medical Oncology, Winship Cancer Center, Emory University, Atlanta, GA, USA
| | - A W Hamburger
- Departments of Medicine, Pathology, Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, Marlene and Stewart Greenebaum Cancer Center (UMGCC), Baltimore, MD, USA
| | - D D Ross
- Departments of Medicine, Pathology, Pharmacology and Experimental Therapeutics, University of Maryland, School of Medicine, Marlene and Stewart Greenebaum Cancer Center (UMGCC), Baltimore, MD, USA
- Baltimore VA Medical Center, Baltimore, MD, USA
| | - A M Burger
- Barbara Ann Karmanos Cancer Institute and Department of Pharmacology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
32
|
The aryl hydrocarbon receptor: regulation of hematopoiesis and involvement in the progression of blood diseases. Blood Cells Mol Dis 2010; 44:199-206. [PMID: 20171126 DOI: 10.1016/j.bcmd.2010.01.005] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Accepted: 01/08/2010] [Indexed: 12/17/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is a basic helix-loop-helix protein that belongs to the superfamily of environment-sensing PAS (Per-ARNT-Sim) proteins. A large number of ligands have been described to bind AhR and promote its nuclear translocation. In the nucleus, the AhR and its dimerization partner the AhR nuclear translocator (ARNT) form a DNA-binding complex that acts as a transcriptional regulator. Animal and human data suggest that, beyond its mediating responses to xenobiotic and/or unknown endogenous ligands, the AhR has a role, although as yet undefined, in the regulation of cell cycle and inflammation. The AhR also appears to regulate the hematopoietic and immune systems during development and adult life in a cell-specific manner. While accidental exposure to xenobiotic AhR ligands has been associated with leukemia in humans, the specific mechanisms of AhR involvement are still not completely understood. However, recent data are consistent with a functional role of the AhR in the maintenance of hematopoietic stem and/or progenitor cells (HSCs/HPCs). Studies highlighting AhR regulation of HSCs/HPCs provide a rational framework to understand their biology, a role of the AhR in hematopoietic diseases, and a means to develop interventions for these diseases.
Collapse
|
33
|
Gasiewicz TA, Singh KP, Casado FL. The aryl hydrocarbon receptor has an important role in the regulation of hematopoiesis: implications for benzene-induced hematopoietic toxicity. Chem Biol Interact 2009; 184:246-51. [PMID: 19896476 DOI: 10.1016/j.cbi.2009.10.019] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 10/22/2009] [Accepted: 10/24/2009] [Indexed: 10/20/2022]
Abstract
The aryl hydrocarbon receptor (AhR) belongs to the basic helix-loop-helix (bHLH) Per-Arnt-Sim (PAS) family of transcription factors. Many of these proteins are involved in regulating responses to signals in the tissue environment such as hypoxia, oxidation-reduction status, and circadian rhythms. Although the AhR is well studied as a mediator of the toxicity of certain xenobiotics, the normal physiological function remains unknown. However, accumulating data support a hypothesis that the AhR has an important function in the regulation of hematopoietic stem cells (HSCs). Persistent AhR activation by dioxin, a potent xenobiotic AhR agonist, results in altered numbers and function of HSCs in mouse bone marrow. Analysis of HSCs from AhR null-allele mice also indicates that lack of AhR expression results in altered characteristics and function of these cells. HSCs from these animals are hyperproliferative and have altered cell cycle. In addition, aging AhR-KO mice show characteristics consistent with premature bone marrow senescence and are prone to hematopoietic disease. Finally, some data suggest that the expression of the Ahr gene is regulated under conditions that control HSC proliferation. The presence of a normal functioning AhR may provide an important advantage to organisms by regulating the balance between quiescence and proliferation and preventing the premature exhaustion of HSCs and sensitivity to genetic alterations. This function assists in the preservation of HSC function and long-term multi-lineage generation over the lifespan of the organism. This also implicates a role for the AhR in the aging process. Furthermore, these functions may affect the sensitivity of HSCs to certain xenobiotics, including benzene. Defining the molecular mechanisms by which these events occur may lead to the identification of previously undefined roles of this transcription factor in human diseases, particularly those caused or affected by xenobiotics.
Collapse
Affiliation(s)
- Thomas A Gasiewicz
- Department of Environmental Medicine, University of Rochester Medical Center, 601 Elmwood Avenue, Rochester, NY 14642, USA.
| | | | | |
Collapse
|
34
|
Abstract
Abstract
The granulocyte-macrophage colony-stimulating factor (GM-CSF)/interleukin (IL)–3/IL-5 receptor family regulates the production and function of myeloid cells. These cytokines signal through receptor complexes that consist of unique ligand-binding α-chains and common signaling β-chains. IL-5 is distinct from IL-3 and GM-CSF in its capacity to induce eosinophil development, however, the molecular mechanisms that generate functional diversity within this receptor family are mostly unknown. Here, we characterized the selective IL-5Rα–binding adapter protein syntenin in IL-5R function. Syntenin and IL-5Rα colocalize at the plasma membrane and in early endosomal compartments. Manipulation of syntenin expression by ectopic expression or knockdown selectively modulated IL-5R but not GM-CSF receptor signaling, and severely affected IL-5–induced eosinophil differentiation from primary human CD34+ hematopoietic progenitor cells. We found syntenin up-regulated during eosinophilopoiesis but down-regulated during neutropoiesis. Syntenin forms complexes with multiple IL-5Rα chains, suggesting that syntenin-enhanced IL-5R output may result from stabilization of an IL-5–induced oligomeric receptor complex. These data demonstrate that cytokine-specific functions can be transduced by unique receptor α-chain–associating adapter proteins.
Collapse
|
35
|
Integrating extrinsic and intrinsic cues into a minimal model of lineage commitment for hematopoietic progenitors. PLoS Comput Biol 2009; 5:e1000518. [PMID: 19911036 PMCID: PMC2736398 DOI: 10.1371/journal.pcbi.1000518] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2009] [Accepted: 08/25/2009] [Indexed: 02/08/2023] Open
Abstract
Autoregulation of transcription factors and cross-antagonism between lineage-specific transcription factors are a recurrent theme in cell differentiation. An equally prevalent event that is frequently overlooked in lineage commitment models is the upregulation of lineage-specific receptors, often through lineage-specific transcription factors. Here, we use a minimal model that combines cell-extrinsic and cell-intrinsic elements of regulation in order to understand how both instructive and stochastic events can inform cell commitment decisions in hematopoiesis. Our results suggest that cytokine-mediated positive receptor feedback can induce a “switch-like” response to external stimuli during multilineage differentiation by providing robustness to both bipotent and committed states while protecting progenitors from noise-induced differentiation or decommitment. Our model provides support to both the instructive and stochastic theories of commitment: cell fates are ultimately driven by lineage-specific transcription factors, but cytokine signaling can strongly bias lineage commitment by regulating these inherently noisy cell-fate decisions with complex, pertinent behaviors such as ligand-mediated ultrasensitivity and robust multistability. The simulations further suggest that the kinetics of differentiation to a mature cell state can depend on the starting progenitor state as well as on the route of commitment that is chosen. Lastly, our model shows good agreement with lineage-specific receptor expression kinetics from microarray experiments and provides a computational framework that can integrate both classical and alternative commitment paths in hematopoiesis that have been observed experimentally. Complex biomolecular interaction pathways in signaling networks can lead to non-intuitive behaviors that can prove critical for the regulation and robustness of biological processes. In this work, we present a signaling topology that can generate dynamic responses that are particularly pertinent to cell commitment in hematopoiesis. Our minimal model explores fundamental questions of instructive signaling that have persisted in cell-fate decisions. We show that even when lineage commitment decisions are inherently noisy, external cytokine signals, amplified by receptor upregulation, can bias the lineage choices of a progenitor cell. The multipotent progenitor, based on its differentiation potential, can exhibit several layers of memory to provide stability to both intermediate and mature states and can potentially bypass canonical intermediate states in generating mature cell types. Thus, our model provides a computational framework that can accommodate both classical and non-classical commitment paths in hematopoiesis.
Collapse
|
36
|
Undifferentiated hematopoietic cells are characterized by a genome-wide undermethylation dip around the transcription start site and a hierarchical epigenetic plasticity. Blood 2009; 114:4968-78. [PMID: 19752395 DOI: 10.1182/blood-2009-01-197780] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Evidence for the epigenetic regulation of hematopoietic stem cells (HSCs) is growing, but the genome-wide epigenetic signature of HSCs and its functional significance remain unclear. In this study, from a genome-wide comparison of CpG methylation in human CD34(+) and CD34(-) cells, we identified a characteristic undermethylation dip around the transcription start site of promoters and an overmethylation of flanking regions in undifferentiated CD34(+) cells. This "bivalent-like" CpG methylation pattern around the transcription start site was more prominent in genes not associated with CpG islands (CGI(-)) than CGI(+) genes. Undifferentiated hematopoietic cells also exhibited dynamic chromatin associated with active transcription and a higher turnover of histone acetylation than terminally differentiated cells. Interestingly, inhibition of chromatin condensation by chemical treatment (5-azacytidine, trichostatin A) enhanced the self-renewal of "stimulated" HSCs in reconstituting bone marrows but not "steady-state" HSCs in stationary phase bone marrows. In contrast, similar treatments on more mature cells caused partial phenotypic dedifferentiation and apoptosis at levels correlated with their hematopoietic differentiation. Taken together, our study reveals that the undifferentiated state of hematopoietic cells is characterized by a unique epigenetic signature, which includes dynamic chromatin structures and an epigenetic plasticity that correlates to level of undifferentiation.
Collapse
|
37
|
Ribeiro AS, Dai X, Yli-Harja O. Variability of the distribution of differentiation pathway choices regulated by a multipotent delayed stochastic switch. J Theor Biol 2009; 260:66-76. [DOI: 10.1016/j.jtbi.2009.05.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Revised: 05/25/2009] [Accepted: 05/26/2009] [Indexed: 11/17/2022]
|
38
|
Abstract
The stunning possibility of "reprogramming" differentiated somatic cells to express a pluripotent stem cell phenotype (iPS, induced pluripotent stem cell) and the "ground state" character of pluripotency reveal fundamental features of cell fate regulation that lie beyond existing paradigms. The rarity of reprogramming events appears to contradict the robustness with which the unfathomably complex phenotype of stem cells can reliably be generated. This apparent paradox, however, is naturally explained by the rugged "epigenetic landscape" with valleys representing "preprogrammed" attractor states that emerge from the dynamical constraints of the gene regulatory network. This article provides a pedagogical primer to the fundamental principles of gene regulatory networks as integrated dynamic systems and reviews recent insights in gene expression noise and fate determination, thereby offering a formal framework that may help us to understand why cell fate reprogramming events are inherently rare and yet so robust.
Collapse
Affiliation(s)
- Sui Huang
- Institute for Biocomplexity and Informatics, University of Calgary, Calgary, AB, Canada.
| |
Collapse
|
39
|
Halley JD, Burden FR, Winkler DA. Stem cell decision making and critical-like exploratory networks. Stem Cell Res 2009; 2:165-77. [DOI: 10.1016/j.scr.2009.03.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 02/24/2009] [Accepted: 03/06/2009] [Indexed: 10/21/2022] Open
|
40
|
Chang SJ, Huang TS, Wang KL, Wang TY, Yang YC, Chang MDT, Wu YH, Wang HW. Genetic network analysis of human CD34+ hematopoietic stem/precursor cells. Taiwan J Obstet Gynecol 2009; 47:422-30. [PMID: 19126509 DOI: 10.1016/s1028-4559(09)60010-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE Somatic CD34+ hematopoietic stem/precursor cells (HSPCs) give rise to hematopoietic cells and endothelial cells and have been used in clinical applications. Understanding the genes responsible for stemness and how they interact with each other will help us to manipulate these cells more efficiently in the future. MATERIALS AND METHODS We performed microarray analysis on human CD34+ HSPCs and on two different progeny cell types, i.e. microvascular endothelial cells and peripheral blood mononuclear cells. Systems biology and advanced bioinformatics tools were used to help clarify the genetic networks associated with these stem cell genes. RESULTS We identified CD34+ HSPC genes and found that they were involved in critical biologic processes such as cell cycle regulation, chromosome organization, and DNA repair. We also identified a novel precursor gene cluster on chromosome 19p13.3. Analysis of HSPC-enriched genes using systems biology tools revealed a complex genetic network functioning in CD34+ cells, in which several genes acted as hubs to maintain the stability (such as GATA1) or connectivity (such as hepatic growth factor) of the whole network. CONCLUSION This study provides the foundation for a more detailed understanding of CD34+ HSPCs.
Collapse
Affiliation(s)
- Shing-Jyh Chang
- Department of Obstetrics and Gynecology, Mackay Memorial Hospital, and National Tsing Hua University, HsinChu, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
The emergence of lineage-specific chromosomal topologies from coordinate gene regulation. Proc Natl Acad Sci U S A 2009; 106:6679-84. [PMID: 19276122 DOI: 10.1073/pnas.0900986106] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Although the importance of chromosome organization during mitosis is clear, it remains to be determined whether the nucleus assumes other functionally relevant chromosomal topologies. We have previously shown that homologous chromosomes have a tendency to associate during hematopoiesis according to their distribution of coregulated genes, suggesting cell-specific nuclear organization. Here, using the mathematical approaches of distance matrices and coupled oscillators, we model the dynamic relationship between gene expression and chromosomal associations during the differentiation of a multipotential hematopoietic progenitor. Our analysis reveals dramatic changes in total genomic order: Commitment of the progenitor results in an initial increase in entropy at both the level of gene coregulation and chromosomal organization, which we suggest represents a phase transition, followed by a progressive decline in entropy during differentiation. The stabilization of a highly ordered state in the differentiated cell types results in lineage-specific chromosomal topologies and is related to the emergence of coherence-or self-organization-between chromosomal associations and coordinate gene regulation. We discuss how these observations may be generally relevant to cell fate decisions encountered by progenitor/stem cells.
Collapse
|
42
|
Hoxa6 potentiates short-term hemopoietic cell proliferation and extended self-renewal. Exp Hematol 2009; 37:322-33.e3. [DOI: 10.1016/j.exphem.2008.10.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2008] [Revised: 10/10/2008] [Accepted: 10/28/2008] [Indexed: 11/23/2022]
|
43
|
Chen J, Olsen J, Ford S, Mirza S, Walker A, Murphy JM, Young IG. A New Isoform of Interleukin-3 Receptor α with Novel Differentiation Activity and High Affinity Binding Mode. J Biol Chem 2009; 284:5763-73. [DOI: 10.1074/jbc.m808197200] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
44
|
Chickarmane V, Enver T, Peterson C. Computational modeling of the hematopoietic erythroid-myeloid switch reveals insights into cooperativity, priming, and irreversibility. PLoS Comput Biol 2009; 5:e1000268. [PMID: 19165316 PMCID: PMC2613533 DOI: 10.1371/journal.pcbi.1000268] [Citation(s) in RCA: 96] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2008] [Accepted: 12/05/2008] [Indexed: 11/24/2022] Open
Abstract
Hematopoietic stem cell lineage choices are decided by genetic networks that are turned ON/OFF in a switch-like manner. However, prior to lineage commitment, genes are primed at low expression levels. Understanding the underlying molecular circuitry in terms of how it governs both a primed state and, at the other extreme, a committed state is of relevance not only to hematopoiesis but also to developmental systems in general. We develop a computational model for the hematopoietic erythroid-myeloid lineage decision, which is determined by a genetic switch involving the genes PU.1 and GATA-1. Dynamical models based upon known interactions between these master genes, such as mutual antagonism and autoregulation, fail to make the system bistable, a desired feature for robust lineage determination. We therefore suggest a new mechanism involving a cofactor that is regulated as well as recruited by one of the master genes to bind to the antagonistic partner that is necessary for bistability and hence switch-like behavior. An interesting fallout from this architecture is that suppression of the cofactor through external means can lead to a loss of cooperativity, and hence to a primed state for PU.1 and GATA-1. The PU.1–GATA-1 switch also interacts with another mutually antagonistic pair, –FOG-1. The latter pair inherits the state of its upstream master genes and further reinforces the decision due to several feedback loops, thereby leading to irreversible commitment. The genetic switch, which handles the erythroid-myeloid lineage decision, is an example of a network that implements both a primed and a committed state by regulating cooperativity through recruitment of cofactors. Perturbing the feedback between the master regulators and downstream targets suggests potential reprogramming strategies. The approach points to a framework for lineage commitment studies in general and could aid the search for lineage-determining genes. An important question in developmental biology is how different lineage choices are regulated at the genetic level. Robust lineage decisions are implemented by genetic switches, whereby one set of master genes are ON and another set are OFF, leading to a specific expression pattern of genes for a particular lineage. We develop a computational model to illustrate these principles as applied to the hematopoietic erythroid-myeloid lineage choice, where two master regulator genes, PU.1 and GATA-1, function as a genetic switch. The model, which is based upon known interactions, suggests missing interactions between the master genes, which we hypothesize, so as to reproduce the desired dynamics. Furthermore, there exist feedback interactions between the master genes and their downstream targets. When these are included in the model, the dynamics imply that the feedback is responsible for irreversible commitment. Our results suggest the search for missing interactions between the master genes in terms of a coregulated cofactor. The second important result of the model is that reprogramming irreversible cell fate may be possible by perturbing feedback regulation between the master genes and their downstream targets. Hence, dynamical modeling provides prediction of novel mechanisms and also strategies for reprogramming the fates of cells.
Collapse
Affiliation(s)
- Vijay Chickarmane
- Division of Biology, California Institute of Technology, Pasadena, California, United States of America
| | - Tariq Enver
- MRC Molecular Biology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, United Kingdom
- Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden
| | - Carsten Peterson
- Lund Strategic Research Center for Stem Cell Biology and Cell Therapy, Lund University, Lund, Sweden
- Computational Biology and Biological Physics, Department of Theoretical Physics, Lund University, Lund, Sweden
- * E-mail:
| |
Collapse
|
45
|
Singh KP, Casado FL, Opanashuk LA, Gasiewicz TA. The aryl hydrocarbon receptor has a normal function in the regulation of hematopoietic and other stem/progenitor cell populations. Biochem Pharmacol 2008; 77:577-87. [PMID: 18983985 DOI: 10.1016/j.bcp.2008.10.001] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2008] [Revised: 10/01/2008] [Accepted: 10/01/2008] [Indexed: 12/17/2022]
Abstract
The aryl hydrocarbon receptor (AhR) is known mainly as the mediator for the toxicity of certain xenobiotics. However, there is also much information to indicate that this transcription factor has important biological functions. Here we review the evidence that the AhR has a significant role in the regulation of hematopoietic stem cells (HSCs). Data to support this come from studies with xenobiotic AhR ligands, phenotypic analyses of mice lacking AhR, examining the presence and regulation of the AhR within HSCs, knowledge of genes and signaling pathways regulated by the AhR, and investigations of hematopoietic disorders. Based on this information, we hypothesize that AhR expression is necessary for the proper maintenance of quiescence in HSCs, and that AhR down-regulation is essential for "escape" from quiescence and subsequent proliferation of these cells. This implicates the AhR as a negative regulator of hematopoiesis with a function of curbing excessive or unnecessary proliferation. This provides an important advantage by preventing the premature exhaustion of HSCs and sensitivity to genetic alterations, thus preserving HSC function and long-term multi-lineage generation over the lifespan of the organism. This also implicates a role of the AhR in aging processes. AhR dysregulation may result in the altered ability of HSCs to sense appropriate signals in the bone marrow microenvironment leading to hematopoietic disease. It is also reasonable to hypothesize that this protein has an important function in the regulation of other tissue stem cell populations. Suggestive evidence is consistent with a role in skin and neural stem cells.
Collapse
Affiliation(s)
- Kameshwar P Singh
- Department of Environmental Medicine, University of Rochester School of Medicine, Rochester, NY 14642, USA
| | | | | | | |
Collapse
|
46
|
Elkon R, Agami R. Removal of AU bias from microarray mRNA expression data enhances computational identification of active microRNAs. PLoS Comput Biol 2008; 4:e1000189. [PMID: 18833292 PMCID: PMC2533120 DOI: 10.1371/journal.pcbi.1000189] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2008] [Accepted: 08/20/2008] [Indexed: 01/25/2023] Open
Abstract
Elucidation of regulatory roles played by microRNAs (miRs) in various biological networks is one of the greatest challenges of present molecular and computational biology. The integrated analysis of gene expression data and 3′-UTR sequences holds great promise for being an effective means to systematically delineate active miRs in different biological processes. Applying such an integrated analysis, we uncovered a striking relationship between 3′-UTR AU content and gene response in numerous microarray datasets. We show that this relationship is secondary to a general bias that links gene response and probe AU content and reflects the fact that in the majority of current arrays probes are selected from target transcript 3′-UTRs. Therefore, removal of this bias, which is in order in any analysis of microarray datasets, is of crucial importance when integrating expression data and 3′-UTR sequences to identify regulatory elements embedded in this region. We developed visualization and normalization schemes for the detection and removal of such AU biases and demonstrate that their application to microarray data significantly enhances the computational identification of active miRs. Our results substantiate that, after removal of AU biases, mRNA expression profiles contain ample information which allows in silico detection of miRs that are active in physiological conditions. MicroRNAs are a novel class of genes that encodes for short RNA molecules recognized to play key roles in the regulation of many biological networks. MicroRNAs, predicted to collectively target more than 30% of all human protein-coding genes, suppress gene expression by binding to regulatory elements usually embedded in the 3′-UTRs of their target mRNAs. Despite intensive efforts in recent years, biological functions carried out by microRNAs have been characterized for only a small number of these genes, making elucidation of their roles one of the greatest challenges of biology today. Bioinformatics analyses can significantly help meet this challenge. In particular, the integrated analysis of microarray mRNA expression data and 3′-UTR sequences holds great promise for systematic dissection of regulatory networks controlled by microRNAs. Applying such integrated analysis to numerous microarray datasets, we disclosed a major technical bias that hampers the identification of active microRNAs from mRNA expression profiles. We developed visualization and normalization schemes for detection and removal of the bias and demonstrate that their application to microarray data significantly enhances the identification of active microRNAs. Given the broad use of microarrays and the ever-growing interest in microRNAs, we anticipate that the methods we introduced will be widely adopted.
Collapse
Affiliation(s)
- Ran Elkon
- Division of Gene Regulation, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Reuven Agami
- Division of Gene Regulation, The Netherlands Cancer Institute, Amsterdam, The Netherlands
- * E-mail:
| |
Collapse
|
47
|
Halley JD, Winkler DA, Burden FR. Toward a Rosetta stone for the stem cell genome: Stochastic gene expression, network architecture, and external influences. Stem Cell Res 2008; 1:157-68. [DOI: 10.1016/j.scr.2008.03.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2008] [Revised: 03/17/2008] [Accepted: 03/21/2008] [Indexed: 02/05/2023] Open
|
48
|
Steel factor coordinately regulates the molecular signature and biologic function of hematopoietic stem cells. Blood 2008; 112:560-7. [PMID: 18502833 DOI: 10.1182/blood-2007-10-117820] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Hematopoietic stem cells (HSCs) regenerated in vivo display sustained differences in their self-renewal and differentiation activities. Variations in Steel factor (SF) signaling are known to affect these functions in vitro, but the cellular and molecular mechanisms involved are not understood. To address these issues, we evaluated highly purified HSCs maintained in single-cell serum-free cultures containing 20 ng/mL IL-11 plus 1, 10, or 300 ng/mL SF. Under all conditions, more than 99% of the cells traversed a first cell cycle with similar kinetics. After 8 hours in the 10 or 300 ng/mL SF conditions, the frequency of HSCs remained unchanged. However, in the next 8 hours (ie, 6 hours before any cell divided), HSC integrity was sustained only in the 300 ng/mL SF cultures. The cells in these cultures also contained significantly higher levels of Bmi1, Lnk, and Ezh2 transcripts but not of several other regulators. Assessment of 21 first division progeny pairs further showed that only those generated in 300 ng/mL SF cultures contained HSCs and pairs of progeny with similar differentiation programs were not observed. Thus, SF signaling intensity can directly and coordinately alter the transcription factor profile and long-term repopulating ability of quiescent HSCs before their first division.
Collapse
|
49
|
Zhao L, Neumann B, Murphy K, Silke J, Gonda TJ. Lack of reproducible growth inhibition by Schlafen1 and Schlafen2 in vitro. Blood Cells Mol Dis 2008; 41:188-93. [PMID: 18479948 DOI: 10.1016/j.bcmd.2008.03.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2008] [Accepted: 03/24/2008] [Indexed: 11/17/2022]
Abstract
The Schlafen gene family has been implicated in lymphoid and myeloid maturation and differentiation as well as inflammation. However, little is known about the functions of this gene family except that anti-proliferative activities, particularly for Schlafen1, the prototype member of the family, have been reported. This was shown mainly by ectopic expression of Schlafen1 in murine fibroblasts resulting in growth inhibition and a G1 cell cycle arrest apparently via repression of Cyclin D1 expression. However, we have been unable to reproduce these findings. Schlafen1 and Schlafen2 failed to inhibit cell proliferation, cause G1 cell cycle arrest, or affect Cyclin D1 level in murine fibroblasts. This was regardless of whether overexpression was constitutive, induced or from transient transfections. Moreover, in our hands, Schlafen1 and -2 do not appear to regulate the activity of Cyclin D1 promoter. Importantly, we also showed that Schlafen1 and -2 do not play anti-proliferative roles in more physiologically-relevant myeloid cell lines. We therefore suggest that Schlafen1 and Schlafen2 might not have obligatory anti-proliferative activities, at least in vitro, and that efforts to explore their functions should be directed to other aspects, such as haemopoietic development and immune response.
Collapse
Affiliation(s)
- Liang Zhao
- University of Queensland Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Brisbane, Australia
| | | | | | | | | |
Collapse
|
50
|
Wang W, Horner DN, Chen WLK, Zandstra PW, Audet J. Synergy between erythropoietin and stem cell factor during erythropoiesis can be quantitatively described without co-signaling effects. Biotechnol Bioeng 2008; 99:1261-72. [DOI: 10.1002/bit.21677] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|