1
|
Heritable changes in division speed accompany the diversification of single T cell fate. Proc Natl Acad Sci U S A 2022; 119:2116260119. [PMID: 35217611 PMCID: PMC8892279 DOI: 10.1073/pnas.2116260119] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/18/2022] [Indexed: 11/18/2022] Open
Abstract
Rapid clonal expansion of antigen-specific T cells is a fundamental feature of adaptive immune responses. Here, we utilize continuous live-cell imaging in vitro to track the division speed and genealogical connections of all descendants derived from a single naive CD8+ T cell throughout up to ten divisions of activation-induced proliferation. Bayesian inference of tree-structured data reveals that clonal expansion is divided into a homogenously fast burst phase encompassing two to three divisions and a subsequent diversification phase during which T cells segregate into quickly dividing effector T cells and more slowly cycling memory precursors. Our work highlights cell cycle speed as a major heritable property that is regulated in parallel to key lineage decisions of activated T cells. Rapid clonal expansion of antigen-specific T cells is a fundamental feature of adaptive immune responses. It enables the outgrowth of an individual T cell into thousands of clonal descendants that diversify into short-lived effectors and long-lived memory cells. Clonal expansion is thought to be programmed upon priming of a single naive T cell and then executed by homogenously fast divisions of all of its descendants. However, the actual speed of cell divisions in such an emerging “T cell family” has never been measured with single-cell resolution. Here, we utilize continuous live-cell imaging in vitro to track the division speed and genealogical connections of all descendants derived from a single naive CD8+ T cell throughout up to ten divisions of activation-induced proliferation. This comprehensive mapping of T cell family trees identifies a short burst phase, in which division speed is homogenously fast and maintained independent of external cytokine availability or continued T cell receptor stimulation. Thereafter, however, division speed diversifies, and model-based computational analysis using a Bayesian inference framework for tree-structured data reveals a segregation into heritably fast- and slow-dividing branches. This diversification of division speed is preceded already during the burst phase by variable expression of the interleukin-2 receptor alpha chain. Later it is accompanied by selective expression of memory marker CD62L in slower dividing branches. Taken together, these data demonstrate that T cell clonal expansion is structured into subsequent burst and diversification phases, the latter of which coincides with specification of memory versus effector fate.
Collapse
|
2
|
Asada K, Takasawa K, Machino H, Takahashi S, Shinkai N, Bolatkan A, Kobayashi K, Komatsu M, Kaneko S, Okamoto K, Hamamoto R. Single-Cell Analysis Using Machine Learning Techniques and Its Application to Medical Research. Biomedicines 2021; 9:biomedicines9111513. [PMID: 34829742 PMCID: PMC8614827 DOI: 10.3390/biomedicines9111513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 10/06/2021] [Accepted: 10/19/2021] [Indexed: 01/14/2023] Open
Abstract
In recent years, the diversity of cancer cells in tumor tissues as a result of intratumor heterogeneity has attracted attention. In particular, the development of single-cell analysis technology has made a significant contribution to the field; technologies that are centered on single-cell RNA sequencing (scRNA-seq) have been reported to analyze cancer constituent cells, identify cell groups responsible for therapeutic resistance, and analyze gene signatures of resistant cell groups. However, although single-cell analysis is a powerful tool, various issues have been reported, including batch effects and transcriptional noise due to gene expression variation and mRNA degradation. To overcome these issues, machine learning techniques are currently being introduced for single-cell analysis, and promising results are being reported. In addition, machine learning has also been used in various ways for single-cell analysis, such as single-cell assay of transposase accessible chromatin sequencing (ATAC-seq), chromatin immunoprecipitation sequencing (ChIP-seq) analysis, and multi-omics analysis; thus, it contributes to a deeper understanding of the characteristics of human diseases, especially cancer, and supports clinical applications. In this review, we present a comprehensive introduction to the implementation of machine learning techniques in medical research for single-cell analysis, and discuss their usefulness and future potential.
Collapse
Affiliation(s)
- Ken Asada
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
- Correspondence: (K.A.); (R.H.); Tel.: +81-3-3547-5271 (R.H.)
| | - Ken Takasawa
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Hidenori Machino
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Satoshi Takahashi
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Norio Shinkai
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Amina Bolatkan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
| | - Kazuma Kobayashi
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
| | - Masaaki Komatsu
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo 103-0027, Japan; (K.T.); (H.M.); (S.T.); (N.S.); (A.B.); (M.K.)
| | - Syuzo Kaneko
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
| | - Koji Okamoto
- Division of Cancer Differentiation, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan;
| | - Ryuji Hamamoto
- Department of NCC Cancer Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan; (K.K.); (S.K.)
- Correspondence: (K.A.); (R.H.); Tel.: +81-3-3547-5271 (R.H.)
| |
Collapse
|
3
|
Weiskittel TM, Correia C, Yu GT, Ung CY, Kaufmann SH, Billadeau DD, Li H. The Trifecta of Single-Cell, Systems-Biology, and Machine-Learning Approaches. Genes (Basel) 2021; 12:1098. [PMID: 34356114 PMCID: PMC8306972 DOI: 10.3390/genes12071098] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/13/2021] [Accepted: 07/18/2021] [Indexed: 12/18/2022] Open
Abstract
Together, single-cell technologies and systems biology have been used to investigate previously unanswerable questions in biomedicine with unparalleled detail. Despite these advances, gaps in analytical capacity remain. Machine learning, which has revolutionized biomedical imaging analysis, drug discovery, and systems biology, is an ideal strategy to fill these gaps in single-cell studies. Machine learning additionally has proven to be remarkably synergistic with single-cell data because it remedies unique challenges while capitalizing on the positive aspects of single-cell data. In this review, we describe how systems-biology algorithms have layered machine learning with biological components to provide systems level analyses of single-cell omics data, thus elucidating complex biological mechanisms. Accordingly, we highlight the trifecta of single-cell, systems-biology, and machine-learning approaches and illustrate how this trifecta can significantly contribute to five key areas of scientific research: cell trajectory and identity, individualized medicine, pharmacology, spatial omics, and multi-omics. Given its success to date, the systems-biology, single-cell omics, and machine-learning trifecta has proven to be a potent combination that will further advance biomedical research.
Collapse
Affiliation(s)
- Taylor M. Weiskittel
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Mayo Clinic, 200 First, Street SW, Rochester, MN 55905, USA; (T.M.W.); (C.C.); (G.T.Y.); (C.Y.U.); (S.H.K.)
| | - Cristina Correia
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Mayo Clinic, 200 First, Street SW, Rochester, MN 55905, USA; (T.M.W.); (C.C.); (G.T.Y.); (C.Y.U.); (S.H.K.)
| | - Grace T. Yu
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Mayo Clinic, 200 First, Street SW, Rochester, MN 55905, USA; (T.M.W.); (C.C.); (G.T.Y.); (C.Y.U.); (S.H.K.)
| | - Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Mayo Clinic, 200 First, Street SW, Rochester, MN 55905, USA; (T.M.W.); (C.C.); (G.T.Y.); (C.Y.U.); (S.H.K.)
| | - Scott H. Kaufmann
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Mayo Clinic, 200 First, Street SW, Rochester, MN 55905, USA; (T.M.W.); (C.C.); (G.T.Y.); (C.Y.U.); (S.H.K.)
| | - Daniel D. Billadeau
- Department of Immunology, Mayo Clinic College of Medicine and Science, Mayo Clinic, 200 First, Street SW, Rochester, MN 55905, USA;
| | - Hu Li
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine and Science, Mayo Clinic, 200 First, Street SW, Rochester, MN 55905, USA; (T.M.W.); (C.C.); (G.T.Y.); (C.Y.U.); (S.H.K.)
| |
Collapse
|
4
|
Bast L, Buck MC, Hecker JS, Oostendorp RAJ, Götze KS, Marr C. Computational modeling of stem and progenitor cell kinetics identifies plausible hematopoietic lineage hierarchies. iScience 2021; 24:102120. [PMID: 33665548 PMCID: PMC7897991 DOI: 10.1016/j.isci.2021.102120] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/08/2021] [Accepted: 01/22/2021] [Indexed: 12/11/2022] Open
Abstract
Classically, hematopoietic stem cell (HSC) differentiation is assumed to occur via progenitor compartments of decreasing plasticity and increasing maturity in a specific, hierarchical manner. The classical hierarchy has been challenged in the past by alternative differentiation pathways. We abstracted experimental evidence into 10 differentiation hierarchies, each comprising 7 cell type compartments. By fitting ordinary differential equation models with realistic waiting time distributions to time-resolved data of differentiating HSCs from 10 healthy human donors, we identified plausible lineage hierarchies and rejected others. We found that, for most donors, the classical model of hematopoiesis is preferred. Surprisingly, multipotent lymphoid progenitor differentiation into granulocyte-monocyte progenitors is plausible in 90% of samples. An in silico analysis confirmed that, even for strong noise, the classical model can be identified robustly. Our computational approach infers differentiation hierarchies in a personalized fashion and can be used to gain insights into kinetic alterations of diseased hematopoiesis. We assembled 10 lineage hierarchy models of human hematopoiesis Multiparameter immunophenotyping determines HSC differentiation for 10 healthy donors ODE fitting and model selection allows to identify plausible hierarchies A simulation study confirms robustness of model selection for different noise levels
Collapse
Affiliation(s)
- Lisa Bast
- Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.,Technical University of Munich, Department of Mathematics, Chair of Mathematical Modeling of Biological Systems, Garching, Germany
| | - Michèle C Buck
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III, Munich, Germany
| | - Judith S Hecker
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III, Munich, Germany
| | - Robert A J Oostendorp
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III, Munich, Germany
| | - Katharina S Götze
- Technical University of Munich, School of Medicine, Klinikum rechts der Isar, Department of Internal Medicine III, Munich, Germany.,German Cancer Consortium (DKTK), Heidelberg, Partner Site Munich, Germany
| | - Carsten Marr
- Helmholtz Zentrum München-German Research Center for Environmental Health, Institute of Computational Biology, Neuherberg, Germany.,Technical University of Munich, Department of Mathematics, Chair of Mathematical Modeling of Biological Systems, Garching, Germany
| |
Collapse
|
5
|
Clément F, Robin F, Yvinec R. Stochastic nonlinear model for somatic cell population dynamics during ovarian follicle activation. J Math Biol 2021; 82:12. [PMID: 33528641 DOI: 10.1007/s00285-021-01561-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 11/17/2020] [Accepted: 12/21/2020] [Indexed: 10/22/2022]
Abstract
In mammals, female germ cells are sheltered within somatic structures called ovarian follicles, which remain in a quiescent state until they get activated, all along reproductive life. We investigate the sequence of somatic cell events occurring just after follicle activation, starting by the awakening of precursor somatic cells, and their transformation into proliferative cells. We introduce a nonlinear stochastic model accounting for the joint dynamics of the two cell types, and allowing us to investigate the potential impact of a feedback from proliferative cells onto precursor cells. To tackle the key issue of whether cell proliferation is concomitant or posterior to cell awakening, we assess both the time needed for all precursor cells to awake, and the corresponding increase in the total cell number with respect to the initial cell number. Using the probabilistic theory of first passage times, we design a numerical scheme based on a rigorous finite state projection and coupling techniques to compute the mean extinction time and the cell number at extinction time. We find that the feedback term clearly lowers the number of proliferative cells at the extinction time. We calibrate the model parameters using an exact likelihood approach. We carry out a comprehensive comparison between the initial model and a series of submodels, which helps to select the critical cell events taking place during activation, and suggests that awakening is prominent over proliferation.
Collapse
Affiliation(s)
- Frédérique Clément
- Inria, Centre de recherche Inria Saclay-Île-de-France, 91120, Palaiseau, France
| | - Frédérique Robin
- Inria, Centre de recherche Inria Saclay-Île-de-France, 91120, Palaiseau, France
| | - Romain Yvinec
- Inria, Centre de recherche Inria Saclay-Île-de-France, 91120, Palaiseau, France. .,PRC, INRAE, CNRS, Université de Tours UMR PRC, Centre INRAE Val de Loire, 37380, Nouzilly, France.
| |
Collapse
|
6
|
Abstract
Statistical and mathematical modeling are crucial to describe, interpret, compare, and predict the behavior of complex biological systems including the organization of hematopoietic stem and progenitor cells in the bone marrow environment. The current prominence of high-resolution and live-cell imaging data provides an unprecedented opportunity to study the spatiotemporal dynamics of these cells within their stem cell niche and learn more about aberrant, but also unperturbed, normal hematopoiesis. However, this requires careful quantitative statistical analysis of the spatial and temporal behavior of cells and the interaction with their microenvironment. Moreover, such quantification is a prerequisite for the construction of hypothesis-driven mathematical models that can provide mechanistic explanations by generating spatiotemporal dynamics that can be directly compared to experimental observations. Here, we provide a brief overview of statistical methods in analyzing spatial distribution of cells, cell motility, cell shapes, and cellular genealogies. We also describe cell-based modeling formalisms that allow researchers to simulate emergent behavior in a multicellular system based on a set of hypothesized mechanisms. Together, these methods provide a quantitative workflow for the analytic and synthetic study of the spatiotemporal behavior of hematopoietic stem and progenitor cells.
Collapse
|
7
|
Lineage marker synchrony in hematopoietic genealogies refutes the PU.1/GATA1 toggle switch paradigm. Nat Commun 2018; 9:2697. [PMID: 30002371 PMCID: PMC6043612 DOI: 10.1038/s41467-018-05037-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 05/25/2018] [Indexed: 01/21/2023] Open
Abstract
Molecular regulation of cell fate decisions underlies health and disease. To identify molecules that are active or regulated during a decision, and not before or after, the decision time point is crucial. However, cell fate markers are usually delayed and the time of decision therefore unknown. Fortunately, dividing cells induce temporal correlations in their progeny, which allow for retrospective inference of the decision time point. We present a computational method to infer decision time points from correlated marker signals in genealogies and apply it to differentiating hematopoietic stem cells. We find that myeloid lineage decisions happen generations before lineage marker onsets. Inferred decision time points are in agreement with data from colony assay experiments. The levels of the myeloid transcription factor PU.1 do not change during, but long after the predicted lineage decision event, indicating that the PU.1/GATA1 toggle switch paradigm cannot explain the initiation of early myeloid lineage choice.
Collapse
|
8
|
MacLean AL, Hong T, Nie Q. Exploring intermediate cell states through the lens of single cells. CURRENT OPINION IN SYSTEMS BIOLOGY 2018; 9:32-41. [PMID: 30450444 PMCID: PMC6238957 DOI: 10.1016/j.coisb.2018.02.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
As our catalog of cell states expands, appropriate characterization of these states and the transitions between them is crucial. Here we discuss the roles of intermediate cell states (ICSs) in this growing collection. We begin with definitions and discuss evidence for the existence of ICSs and their relevance in various tissues. We then provide a list of possible functions for ICSs with examples. Finally, we describe means by which ICSs and their functional roles can be identified from single-cell data or predicted from models.
Collapse
Affiliation(s)
- Adam L. MacLean
- Department of Mathematics and Center for Complex Biological Systems, University of California, Irvine, CA 92697, United States
| | - Tian Hong
- Department of Biochemistry & Cellular and Molecular Biology, University of Tennessee, Knoxville, TN 37966, United States
| | - Qing Nie
- Department of Mathematics and Center for Complex Biological Systems, University of California, Irvine, CA 92697, United States,Department of Developmental and Cell Biology, University of California, Irvine, CA 92697, United States
| |
Collapse
|
9
|
Olariu V, Peterson C. Kinetic models of hematopoietic differentiation. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2018; 11:e1424. [PMID: 29660842 PMCID: PMC6191385 DOI: 10.1002/wsbm.1424] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Revised: 02/13/2018] [Accepted: 03/16/2018] [Indexed: 01/02/2023]
Abstract
As cell and molecular biology is becoming increasingly quantitative, there is an upsurge of interest in mechanistic modeling at different levels of resolution. Such models mostly concern kinetics and include gene and protein interactions as well as cell population dynamics. The final goal of these models is to provide experimental predictions, which is now taking on. However, even without matured predictions, kinetic models serve the purpose of compressing a plurality of experimental results into something that can empower the data interpretation, and importantly, suggesting new experiments by turning "knobs" in silico. Once formulated, kinetic models can be executed in terms of molecular rate equations for concentrations or by stochastic simulations when only a limited number of copies are involved. Developmental processes, in particular those of stem and progenitor cell commitments, are not only topical but also particularly suitable for kinetic modeling due to the finite number of key genes involved in cellular decisions. Stem and progenitor cell commitment processes have been subject to intense experimental studies over the last decade with some emphasis on embryonic and hematopoietic stem cells. Gene and protein interactions governing these processes can be modeled by binary Boolean rules or by continuous-valued models with interactions set by binding strengths. Conceptual insights along with tested predictions have emerged from such kinetic models. Here we review kinetic modeling efforts applied to stem cell developmental systems with focus on hematopoiesis. We highlight the future challenges including multi-scale models integrating cell dynamical and transcriptional models. This article is categorized under: Models of Systems Properties and Processes > Mechanistic Models Developmental Biology > Stem Cell Biology and Regeneration.
Collapse
Affiliation(s)
- Victor Olariu
- Department of Computational Biology, Lund University, Lund, Sweden
| | - Carsten Peterson
- Department of Computational Biology, Lund University, Lund, Sweden
| |
Collapse
|
10
|
Lunger I, Fawaz M, Rieger MA. Single-cell analyses to reveal hematopoietic stem cell fate decisions. FEBS Lett 2017; 591:2195-2212. [PMID: 28600837 DOI: 10.1002/1873-3468.12712] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 05/19/2017] [Accepted: 06/02/2017] [Indexed: 12/15/2022]
Abstract
Hematopoietic stem cells (HSCs) are the best studied adult stem cells with enormous clinical value. Most of our knowledge about their biology relies on assays at the single HSC level. However, only the recent advances in developing new single cell technologies allowed the elucidation of the complex regulation of HSC fate decision control. This Review will focus on current attempts to investigate individual HSCs at molecular and functional levels. The advantages of these technologies leading to groundbreaking insights into hematopoiesis will be highlighted, and the challenges facing these technologies will be discussed. The importance of combining molecular and functional assays to enlighten regulatory networks of HSC fate decision control, ideally at high temporal resolution, becomes apparent for future studies.
Collapse
Affiliation(s)
- Ilaria Lunger
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Malak Fawaz
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Michael A Rieger
- Department of Medicine, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany.,German Cancer Consortium (DKTK), Heidelberg, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
MacLean AL, Lo Celso C, Stumpf MP. Concise Review: Stem Cell Population Biology: Insights from Hematopoiesis. Stem Cells 2016; 35:80-88. [DOI: 10.1002/stem.2508] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/19/2016] [Accepted: 08/21/2016] [Indexed: 01/01/2023]
Affiliation(s)
- Adam L. MacLean
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| | - Cristina Lo Celso
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| | - Michael P.H. Stumpf
- Department of Life Sciences; Imperial College London; South Kensington Campus London United Kingdom
| |
Collapse
|
12
|
Abstract
Molecular noise in gene regulatory networks has two intrinsic components, one part being due to fluctuations caused by the birth and death of protein or mRNA molecules which are often present in small numbers and the other part arising from gene state switching, a single molecule event. Stochastic dynamics of gene regulatory circuits appears to be largely responsible for bifurcations into a set of multi-attractor states that encode different cell phenotypes. The interplay of dichotomous single molecule gene noise with the nonlinear architecture of genetic networks generates rich and complex phenomena. In this paper, we elaborate on an approximate framework that leads to simple hybrid multi-scale schemes well suited for the quantitative exploration of the steady state properties of large-scale cellular genetic circuits. Through a path sum based analysis of trajectory statistics, we elucidate the connection of these hybrid schemes to the underlying master equation and provide a rigorous justification for using dichotomous noise based models to study genetic networks. Numerical simulations of circuit models reveal that the contribution of the genetic noise of single molecule origin to the total noise is significant for a wide range of kinetic regimes.
Collapse
Affiliation(s)
- Davit A Potoyan
- Department of Chemistry and Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
| | - Peter G Wolynes
- Department of Chemistry and Center for Theoretical Biological Physics, Rice University, Houston, Texas 77005, USA
| |
Collapse
|
13
|
Bartels M, Murphy K, Rieter E, Bruin M. Understanding chronic neutropenia: life is short. Br J Haematol 2015; 172:157-69. [PMID: 26456767 DOI: 10.1111/bjh.13798] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The pathophysiological mechanisms underlying chronic neutropenia are extensive, varying from haematopoietic stem cell disorders resulting in defective neutrophil production, to accelerated apoptosis of neutrophil progenitors or circulating mature neutrophils. While the knowledge concerning genetic defects associated with congenital neutropenia or bone marrow failure is increasing rapidly, the functional role and consequences of these genetic alterations is often not well understood. In addition, there is a large group of diseases, including primary immunodeficiencies and metabolic diseases, in which chronic neutropenia is one of the symptoms, while there is no clear bone marrow pathology or haematopoietic stem cell dysfunction. Altogether, these disease entities illustrate the complexity of normal neutrophil development, the functional role of the (bone marrow) microenvironment and the increased propensity to undergo apoptosis, which is typical for neutrophils. The large variety of disorders associated with chronic neutropenia makes classification almost impossible and possibly not desirable, based on the clinical phenotypes. However, a better understanding of the regulation of normal myeloid differentiation and neutrophil development is of great importance in the diagnostic evaluation of unexplained chronic neutropenia. In this review we propose insights in the pathophysiology of chronic neutropenia in the context of the functional role of key players during normal neutrophil development, neutrophil release and neutrophil survival.
Collapse
Affiliation(s)
- Marije Bartels
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Kate Murphy
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Ester Rieter
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Marrie Bruin
- Department of Paediatric Haematology and Stem Cell Transplantation, University Medical Centre Utrecht, Utrecht, the Netherlands
| |
Collapse
|
14
|
Strasser MK, Feigelman J, Theis FJ, Marr C. Inference of spatiotemporal effects on cellular state transitions from time-lapse microscopy. BMC SYSTEMS BIOLOGY 2015; 9:61. [PMID: 26391569 PMCID: PMC4578671 DOI: 10.1186/s12918-015-0208-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 09/08/2015] [Indexed: 11/10/2022]
Abstract
BACKGROUND Time-lapse microscopy allows to monitor cell state transitions in a spatiotemporal context. Combined with single cell tracking and appropriate cell state markers, transition events can be observed within the genealogical relationship of a proliferating population. However, to infer the correlations between the spatiotemporal context and cell state transitions, statistical analysis with an appropriately large number of samples is required. RESULTS Here, we present a method to infer spatiotemporal features predictive of the state transition events observed in time-lapse microscopy data. We first formulate a generative model, simulate different scenarios, such as time-dependent or local cell density-dependent transitions, and illustrate how to estimate univariate transition rates. Second, we formulate the problem in a machine-learning language using regularized linear models. This allows for a multivariate analysis and to disentangle indirect dependencies via feature selection. We find that our method can accurately recover the relevant features and reconstruct the underlying interaction kernels if a critical number of samples is available. Finally, we explicitly use the tree structure of the data to validate if the estimated model is sufficient to explain correlated transition events of sister cells. CONCLUSIONS Using synthetic cellular genealogies, we prove that our method is able to correctly identify features predictive of state transitions and we moreover validate the chosen model. Our approach allows to estimate the number of cellular genealogies required for the proposed spatiotemporal statistical analysis, and we thus provide an important tool for the experimental design of challenging single cell time-lapse microscopy assays.
Collapse
Affiliation(s)
- Michael K Strasser
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.
| | - Justin Feigelman
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.
- Department of Mathematics, Technische Universität München, Boltzmannstr. 3, Garching, 85747, Germany.
| | - Fabian J Theis
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.
- Department of Mathematics, Technische Universität München, Boltzmannstr. 3, Garching, 85747, Germany.
| | - Carsten Marr
- Institute of Computational Biology, Helmholtz Zentrum München, German Research Center for Environmental Health, Ingolstädter Landstr. 1, Neuherberg, 85764, Germany.
| |
Collapse
|
15
|
|
16
|
Wu J, Rostami MR, Tzanakakis ES. Stem cell modeling: From gene networks to cell populations. Curr Opin Chem Eng 2013; 2:17-25. [PMID: 23914346 DOI: 10.1016/j.coche.2013.01.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Despite rapid advances in the field of stem/progenitor cells through experimental studies, relevant modeling approaches have not progressed with a similar pace. Various models have focused on particular aspects of stem cell physiology including gene regulatory networks, gene expression noise and signaling cascades activated by exogenous factors. However, the self-renewal and differentiation of stem cells is driven by the coordinated regulation of events at the subcellular, intercellular and milieu levels. Such events also span multiple time domains from the fast molecular reactions governing gene expression to the slower cell cycle and division. Thus, the development of multiscale computational frameworks for stem cell populations is highly desirable. Multiscale models are expected to aid the design of efficient differentiation strategies and bioprocesses for the generation of therapeutically useful stem cell progeny. Yet, challenges in making these models tractable and pairing those to sufficient experimental data prevent their wide adoption by the stem cell community. Here, we review modeling approaches reported for stem cell populations and associated hurdles.
Collapse
Affiliation(s)
- Jincheng Wu
- Department of Chemical and Biological Engineering, State University of New York at Buffalo, Buffalo, NY 14260
| | | | | |
Collapse
|