1
|
Batool I, Bajcinca N. Stability analysis of a multiscale model of cell cycle dynamics coupled with quiescent and proliferating cell populations. PLoS One 2023; 18:e0280621. [PMID: 36662844 PMCID: PMC9858875 DOI: 10.1371/journal.pone.0280621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/04/2023] [Indexed: 01/22/2023] Open
Abstract
In this paper, we perform a mathematical analysis of our proposed nonlinear, multiscale mathematical model of physiologically structured quiescent and proliferating cell populations at the macroscale and cell-cycle proteins at the microscale. Cell cycle dynamics (microscale) are driven by growth factors derived from the total cell population of quiescent and proliferating cells. Cell-cycle protein concentrations, on the other hand, determine the rates of transition between the two subpopulations. Our model demonstrates the underlying impact of cell cycle dynamics on the evolution of cell population in a tissue. We study the model's well-posedness, derive steady-state solutions, and find sufficient conditions for the stability of steady-state solutions using semigroup and spectral theory. Finally, we performed numerical simulations to see how the parameters affect the model's nonlinear dynamics.
Collapse
Affiliation(s)
- Iqra Batool
- Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Mechanical and Process Engineering, Kaiserslautern, Germany
| | - Naim Bajcinca
- Rheinland-Pfälzische Technische Universität Kaiserslautern-Landau, Mechanical and Process Engineering, Kaiserslautern, Germany
| |
Collapse
|
2
|
Lundberg A, Yi JJJ, Lindström LS, Tobin NP. Reclassifying tumour cell cycle activity in terms of its tissue of origin. NPJ Precis Oncol 2022; 6:59. [PMID: 35987928 PMCID: PMC9392789 DOI: 10.1038/s41698-022-00302-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 07/13/2022] [Indexed: 01/02/2023] Open
Abstract
Genomic alterations resulting in loss of control over the cell cycle is a fundamental hallmark of human malignancies. Whilst pan-cancer studies have broadly assessed tumour genomics and their impact on oncogenic pathways, analyses taking the baseline signalling levels in normal tissue into account are lacking. To this end, we aimed to reclassify the cell cycle activity of tumours in terms of their tissue of origin and determine if any common DNA mutations, chromosome arm-level changes or signalling pathways contribute to an increase in baseline corrected cell cycle activity. Combining normal tissue and pan-cancer data from over 13,000 samples we demonstrate that tumours of gynaecological origin show the highest levels of corrected cell cycle activity, partially owing to hormonal signalling and gene expression changes. We also show that normal and tumour tissues can be separated into groups (quadrants) of low/high cell cycle activity and propose the hypothesis of an upper limit on these activity levels in tumours.
Collapse
Affiliation(s)
- Arian Lundberg
- Department of Radiation Oncology, University of California at San Francisco, San Francisco, CA, USA
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Department of Radiation Oncology, Stanford School of Medicine, Stanford, CA, USA
- Helen Diller Family Comperhensive Cancer Center, University of California at San Francisco, San Francisco, CA, USA
| | - Joan Jong Jing Yi
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Singapore
| | - Linda S Lindström
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Nicholas P Tobin
- Department of Oncology and Pathology, Karolinska Institutet and University Hospital, Stockholm, Sweden.
| |
Collapse
|
3
|
Regulation of Cell Cycle Progression by Growth Factor-Induced Cell Signaling. Cells 2021; 10:cells10123327. [PMID: 34943835 PMCID: PMC8699227 DOI: 10.3390/cells10123327] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2021] [Revised: 11/12/2021] [Accepted: 11/24/2021] [Indexed: 12/12/2022] Open
Abstract
The cell cycle is the series of events that take place in a cell, which drives it to divide and produce two new daughter cells. The typical cell cycle in eukaryotes is composed of the following phases: G1, S, G2, and M phase. Cell cycle progression is mediated by cyclin-dependent kinases (Cdks) and their regulatory cyclin subunits. However, the driving force of cell cycle progression is growth factor-initiated signaling pathways that control the activity of various Cdk–cyclin complexes. While the mechanism underlying the role of growth factor signaling in G1 phase of cell cycle progression has been largely revealed due to early extensive research, little is known regarding the function and mechanism of growth factor signaling in regulating other phases of the cell cycle, including S, G2, and M phase. In this review, we briefly discuss the process of cell cycle progression through various phases, and we focus on the role of signaling pathways activated by growth factors and their receptor (mostly receptor tyrosine kinases) in regulating cell cycle progression through various phases.
Collapse
|
4
|
Molecular control of cell density-mediated exit to quiescence. Cell Rep 2021; 36:109436. [PMID: 34320337 PMCID: PMC8924979 DOI: 10.1016/j.celrep.2021.109436] [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: 08/22/2020] [Revised: 05/04/2021] [Accepted: 07/01/2021] [Indexed: 12/22/2022] Open
Abstract
Contact inhibition of cell proliferation regulates tissue size and prevents uncontrolled cell expansion. When cell density increases, contact inhibition can force proliferating cells into quiescence. Here we show that the variable memory of local cell density experienced by a mother cell controls the levels of the cyclin-dependent kinase (CDK) activator cyclin D1 and inhibitor p27 in newborn daughters, which direct cells to proliferation or quiescence. Much of this regulation can be explained by rapid suppression of ERK activity by high cell density in mothers, which leads to lower cyclin D1 and higher p27 levels in daughters. Strikingly, cell density and mitogen signals compete by shifting the ratio of cyclin D1/p27 levels below or above a single sharp threshold that controls the proliferation decision. Thus, the history of competing cell density and mitogen signals experienced by mothers is funneled into a precise activator-inhibitor balance that decides the fate of daughter cells. Using live single-cell microscopy, Fan and Meyer show that the decision of newborn daughter cells to proliferate or become quiescent is controlled by the memory of local cell density inherited from mother cells. This memory is mediated by an ultrasensitive activator-inhibitor balance between cyclin D1 and p27.
Collapse
|
5
|
Pennycook BR, Barr AR. Restriction point regulation at the crossroads between quiescence and cell proliferation. FEBS Lett 2020; 594:2046-2060. [PMID: 32564372 DOI: 10.1002/1873-3468.13867] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/08/2020] [Accepted: 06/10/2020] [Indexed: 02/11/2024]
Abstract
The coordination of cell proliferation with reversible cell cycle exit into quiescence is crucial for the development of multicellular organisms and for tissue homeostasis in the adult. The decision between quiescence and proliferation occurs at the restriction point, which is widely thought to be located in the G1 phase of the cell cycle, when cells integrate accumulated extracellular and intracellular signals to drive this binary cellular decision. On the molecular level, decision-making is exerted through the activation of cyclin-dependent kinases (CDKs). CDKs phosphorylate the retinoblastoma (Rb) transcriptional repressor to regulate the expression of cell cycle genes. Recently, the classical view of restriction point regulation has been challenged. Here, we review the latest findings on the activation of CDKs, Rb phosphorylation and the nature and position of the restriction point within the cell cycle.
Collapse
Affiliation(s)
- Betheney R Pennycook
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| | - Alexis R Barr
- Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK
- MRC London Institute of Medical Sciences, Imperial College London, London, UK
| |
Collapse
|
6
|
Min M, Rong Y, Tian C, Spencer SL. Temporal integration of mitogen history in mother cells controls proliferation of daughter cells. Science 2020; 368:1261-1265. [PMID: 32241885 PMCID: PMC8363187 DOI: 10.1126/science.aay8241] [Citation(s) in RCA: 69] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 12/19/2019] [Accepted: 03/17/2020] [Indexed: 01/17/2023]
Abstract
Multicellular organisms use mitogens to regulate cell proliferation, but how fluctuating mitogenic signals are converted into proliferation-quiescence decisions is poorly understood. In this work, we combined live-cell imaging with temporally controlled perturbations to determine the time scale and mechanisms underlying this system in human cells. Contrary to the textbook model that cells sense mitogen availability only in the G1 cell cycle phase, we find that mitogenic signaling is temporally integrated throughout the entire mother cell cycle and that even a 1-hour lapse in mitogen signaling can influence cell proliferation more than 12 hours later. Protein translation rates serve as the integrator that proportionally converts mitogen history into corresponding levels of cyclin D in the G2 phase of the mother cell, which controls the proliferation-quiescence decision in daughter cells and thereby couples protein production with cell proliferation.
Collapse
Affiliation(s)
- Mingwei Min
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA.
| | - Yao Rong
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
- Department of Molecular, Cellular and Developmental Biology, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Chengzhe Tian
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA
| | - Sabrina L Spencer
- Department of Biochemistry and BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80303, USA.
| |
Collapse
|
7
|
Le T, Winham CL, Andromidas F, Silver AC, Jellison ER, Levesque AA, Koob AO. Chimera RNA interference knockdown of γ-synuclein in human cortical astrocytes results in mitotic catastrophe. Neural Regen Res 2020; 15:1894-1902. [PMID: 32246638 PMCID: PMC7513975 DOI: 10.4103/1673-5374.280329] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Elevated levels of γ-synuclein (γ-syn) expression have been noted in the progression of glioblastomas, and also in the cerebrospinal fluid of patients diagnosed with neurodegenerative diseases. γ-Syn can be either internalized from the extracellular milieu or expressed endogenously by human cortical astrocytes. Internalized γ-syn results in increased cellular proliferation, brain derived neurotrophic factor release and astroprotection. However, the function of endogenous γ-syn in primary astrocytes, and the relationship to these two opposing disease states are unknown. γ-Syn is expressed by astrocytes in the human cortex, and to gain a better understanding of the role of endogenous γ-syn, primary human cortical astrocytes were treated with chimera RNA interference (RNAi) targeting γ-syn after release from cell synchronization. Quantitative polymerase chain reaction analysis demonstrated an increase in endogenous γ-syn expression 48 hours after release from cell synchronization, while RNAi reduced γ-syn expression to control levels. Immunocytochemistry of Ki67 and 5-bromodeoxyuridine showed chimera RNAi γ-syn knockdown reduced cellular proliferation at 24 and 48 hours after release from cell synchronization. To further investigate the consequence of γ-syn knockdown on the astrocytic cell cycle, phosphorylated histone H3 pSer10 (pHH3) and phosphorylated cyclin dependent kinase-2 pTyr15 (pCDK2) levels were observed via western blot analysis. The results revealed an elevated expression of pHH3, but not pCDK2, indicating γ-syn knockdown leads to disruption of the cell cycle and chromosomal compaction after 48 hours. Subsequently, flow cytometry with propidium iodide determined that increases in apoptosis coincided with γ-syn knockdown. Therefore, γ-syn exerts its effect to allow normal astrocytic progression through the cell cycle, as evidenced by decreased proliferation marker expression, increased pHH3, and mitotic catastrophe after knockdown. In this study, we demonstrated that the knockdown of γ-syn within primary human cortical astrocytes using chimera RNAi leads to cell cycle disruption and apoptosis, indicating an essential role for γ-syn in regulating normal cell division in astrocytes. Therefore, disruption to γ-syn function would influence astrocytic proliferation, and could be an important contributor to neurological diseases.
Collapse
Affiliation(s)
- Timmy Le
- Graduate Program in Neuroscience, Biology Department, University of Hartford, West Hartford, CT, USA
| | - Cynthia L Winham
- Graduate Program in Neuroscience, Biology Department, University of Hartford, West Hartford, CT, USA
| | - Fotis Andromidas
- Graduate Program in Neuroscience, Biology Department, University of Hartford, West Hartford, CT, USA
| | - Adam C Silver
- Graduate Program in Neuroscience, Biology Department, University of Hartford, West Hartford, CT, USA
| | - Evan R Jellison
- Department of Immunology, UCONN Health Center, Farmington, CT, USA
| | - Aime A Levesque
- Graduate Program in Neuroscience, Biology Department, University of Hartford, West Hartford, CT, USA
| | - Andrew O Koob
- Graduate Program in Neuroscience, Biology Department, University of Hartford, West Hartford, CT, USA
| |
Collapse
|
8
|
Ghaleb A, Yallowitz A, Marchenko N. Irradiation induces p53 loss of heterozygosity in breast cancer expressing mutant p53. Commun Biol 2019; 2:436. [PMID: 31799437 PMCID: PMC6881331 DOI: 10.1038/s42003-019-0669-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Accepted: 10/16/2019] [Indexed: 01/10/2023] Open
Abstract
Mutations in one allele of the TP53 gene in cancer early stages are frequently followed by the loss of the remaining wild-type allele (LOH) during tumor progression. However, the clinical impact of TP53 mutations and p53LOH, especially in the context of genotoxic modalities, remains unclear. Using MMTV;ErbB2 model carrying a heterozygous R172H p53 mutation, we report a previously unidentified oncogenic activity of mutant p53 (mutp53): the exacerbation of p53LOH after irradiation. We show that wild-type p53 allele is partially transcriptionally competent and enables the maintenance of the genomic integrity under normal conditions in mutp53 heterozygous cells. In heterozygous cells γ-irradiation promotes mutp53 stabilization, which suppresses DNA repair and the cell cycle checkpoint allowing cell cycle progression in the presence of inefficiently repaired DNA, consequently increases genomic instability leading to p53LOH. Hence, in mutp53 heterozygous cells, irradiation facilitates the selective pressure for p53LOH that enhances cancer cell fitness and provides the genetic plasticity for acquiring metastatic properties.
Collapse
Affiliation(s)
- Amr Ghaleb
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794-8691 USA
| | - Alisha Yallowitz
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794-8691 USA
- Weill Cornell Medicine, 1300 York Avenue, LC-902, New York, NY 10065 USA
| | - Natalia Marchenko
- Department of Pathology, Stony Brook University, Stony Brook, NY 11794-8691 USA
| |
Collapse
|
9
|
Vasjari L, Bresan S, Biskup C, Pai G, Rubio I. Ras signals principally via Erk in G1 but cooperates with PI3K/Akt for Cyclin D induction and S-phase entry. Cell Cycle 2019; 18:204-225. [PMID: 30560710 DOI: 10.1080/15384101.2018.1560205] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Numerous studies exploring oncogenic Ras or manipulating physiological Ras signalling have established an irrefutable role for Ras as driver of cell cycle progression. Despite this wealth of information the precise signalling timeline and effectors engaged by Ras, particularly during G1, remain obscure as approaches for Ras inhibition are slow-acting and ill-suited for charting discrete Ras signalling episodes along the cell cycle. We have developed an approach based on the inducible recruitment of a Ras-GAP that enforces endogenous Ras inhibition within minutes. Applying this strategy to inhibit Ras stepwise in synchronous cell populations revealed that Ras signaling was required well into G1 for Cyclin D induction, pocket protein phosphorylation and S-phase entry, irrespective of whether cells emerged from quiescence or G2/M. Unexpectedly, Erk, and not PI3K/Akt or Ral was activated by Ras at mid-G1, albeit PI3K/Akt signalling was a necessary companion of Ras/Erk for sustaining cyclin-D levels and G1/S transition. Our findings chart mitogenic signaling by endogenous Ras during G1 and identify limited effector engagement restricted to Raf/MEK/Erk as a cogent distinction from oncogenic Ras signalling.
Collapse
Affiliation(s)
- Ledia Vasjari
- a Institute of Molecular Cell Biology, Center for Molecular Biomedicine , Jena University Hospital , Jena , Germany
| | - Stephanie Bresan
- a Institute of Molecular Cell Biology, Center for Molecular Biomedicine , Jena University Hospital , Jena , Germany
| | - Christoph Biskup
- b Biomolecular Photonics Group , Jena University Hospital , Jena , Germany
| | - Govind Pai
- a Institute of Molecular Cell Biology, Center for Molecular Biomedicine , Jena University Hospital , Jena , Germany
| | - Ignacio Rubio
- a Institute of Molecular Cell Biology, Center for Molecular Biomedicine , Jena University Hospital , Jena , Germany
| |
Collapse
|
10
|
Abstract
The Restriction Point was originally defined as the moment that cells commit to the cell cycle and was later suggested to coincide with hyperphosphorylation of the retinoblastoma protein (Rb). Current cell cycle models posit that cells exit mitosis into a pre-Restriction Point state, where they have low cyclin-dependent kinase (CDK) activity and hypophosphorylated Rb; passage through the Restriction Point then occurs in late G1. Recent single-cell studies have challenged the current paradigm, raising questions about the location of the Restriction Point and the notion that cells exit mitosis into a pre-Restriction Point state. Here, we use a variety of single-cell techniques to show that both noncancer and cancer cells bifurcate into two subpopulations after anaphase, marked by increasing vs. low CDK2 activity and hyper- vs. hypophosphorylation of Rb. Notably, subpopulations with hyper- and hypophosphorylated Rb are present within minutes after anaphase, delineating one subpopulation that never "uncrosses" the Restriction Point and continues cycling and another subpopulation that exits mitosis into an uncommitted pre-Restriction Point state. We further show that the CDK inhibitor p21 begins rising in G2 in mother cells whose daughters exit mitosis into the pre-Restriction Point, CDK2low state. Furthermore, degradation of p21 coincides with escape from the CDK2low state and passage through the Restriction Point. Together, these data support a model in which only a subset of cells returns to a pre-Restriction Point state after mitosis and where the Restriction Point is sensitive to not only mitogens, but also inherited DNA replication stress via p21.
Collapse
Affiliation(s)
- Justin Moser
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80309
| | - Iain Miller
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80309
| | - Dylan Carter
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303
- BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80309
| | - Sabrina L Spencer
- Department of Biochemistry, University of Colorado-Boulder, Boulder, CO 80303;
- BioFrontiers Institute, University of Colorado-Boulder, Boulder, CO 80309
| |
Collapse
|
11
|
So WK, Cheung TH. Molecular Regulation of Cellular Quiescence: A Perspective from Adult Stem Cells and Its Niches. Methods Mol Biol 2018; 1686:1-25. [PMID: 29030809 DOI: 10.1007/978-1-4939-7371-2_1] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cellular quiescence is a reversible growth arrest state. In response to extracellular environment, quiescent cells are capable of resuming proliferation for tissue homeostasis and tissue regeneration. Subpopulations of adult stem cells remain quiescent and reside in their specialized stem cell niches. Within the niche, they interact with a repertoire of niche components. Niche integrates signals to maintain quiescence or gear stem cells toward regeneration. Recent studies provide insights into the regulatory components of stem cell niche and their influence on residing stem cells. Aberrant niche activities perturb stem cell quiescence and activation, compromise stem cell functions, and contribute to tissue aging and disease pathogenesis. This review covers current knowledge regarding cellular quiescence with a focus on original and emerging concepts of how niches influence stem cell quiescence.
Collapse
Affiliation(s)
- Wai-Kin So
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China
| | - Tom H Cheung
- Division of Life Science, Center for Stem Cell Research, State Key Laboratory of Molecular Neuroscience, Center of Systems Biology and Human Health, Institute for Advanced Study, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong, China.
| |
Collapse
|
12
|
Pandey N, Vinod PK. Mathematical modelling of reversible transition between quiescence and proliferation. PLoS One 2018; 13:e0198420. [PMID: 29856829 PMCID: PMC5983510 DOI: 10.1371/journal.pone.0198420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/19/2018] [Indexed: 11/18/2022] Open
Abstract
Cells switch between quiescence and proliferation states for maintaining tissue homeostasis and regeneration. At the restriction point (R-point), cells become irreversibly committed to the completion of the cell cycle independent of mitogen. The mechanism involving hyper-phosphorylation of retinoblastoma (Rb) and activation of transcription factor E2F is linked to the R-point passage. However, stress stimuli trigger exit from the cell cycle back to the mitogen-sensitive quiescent state after Rb hyper-phosphorylation but only until APC/CCdh1 inactivation. In this study, we developed a mathematical model to investigate the reversible transition between quiescence and proliferation in mammalian cells with respect to mitogen and stress signals. The model integrates the current mechanistic knowledge and accounts for the recent experimental observations with cells exiting quiescence and proliferating cells. We show that Cyclin E:Cdk2 couples Rb-E2F and APC/CCdh1 bistable switches and temporally segregates the R-point and the G1/S transition. A redox-dependent mutual antagonism between APC/CCdh1 and its inhibitor Emi1 makes the inactivation of APC/CCdh1 bistable. We show that the levels of Cdk inhibitor (CKI) and mitogen control the reversible transition between quiescence and proliferation. Further, we propose that shifting of the mitogen-induced transcriptional program to G2-phase in proliferating cells might result in an intermediate Cdk2 activity at the mitotic exit and in the immediate inactivation of APC/CCdh1. Our study builds a coherent framework and generates hypotheses that can be further explored by experiments.
Collapse
Affiliation(s)
- Nishtha Pandey
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, India
| | - P. K. Vinod
- Center for Computational Natural Sciences and Bioinformatics, International Institute of Information Technology, Hyderabad, India
- * E-mail:
| |
Collapse
|
13
|
Arora M, Moser J, Phadke H, Basha AA, Spencer SL. Endogenous Replication Stress in Mother Cells Leads to Quiescence of Daughter Cells. Cell Rep 2018; 19:1351-1364. [PMID: 28514656 DOI: 10.1016/j.celrep.2017.04.055] [Citation(s) in RCA: 124] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 03/23/2017] [Accepted: 04/19/2017] [Indexed: 12/17/2022] Open
Abstract
Mammalian cells have two fundamentally different states, proliferative and quiescent, but our understanding of how and why cells switch between these states is limited. We previously showed that actively proliferating populations contain a subpopulation that enters quiescence (G0) in an apparently stochastic manner. Using single-cell time-lapse imaging of CDK2 activity and DNA damage, we now show that unresolved endogenous replication stress in the previous (mother) cell cycle prompts p21-dependent entry of daughter cells into quiescence immediately after mitosis. Furthermore, the amount of time daughter cells spend in quiescence is correlated with the extent of inherited damage. Our study thus links replication errors in one cell cycle to the fate of daughter cells in the subsequent cell cycle. More broadly, this work reveals that entry into quiescence is not purely stochastic but has a strong deterministic component arising from a memory of events that occurred in the previous generation(s).
Collapse
Affiliation(s)
- Mansi Arora
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Justin Moser
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Harsha Phadke
- Department of Electrical, Computer and Energy Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Ashik Akbar Basha
- Department of Electrical, Computer and Energy Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Sabrina L Spencer
- Department of Chemistry and Biochemistry, University of Colorado Boulder, Boulder, CO 80309, USA.
| |
Collapse
|
14
|
Jones ZW, Leander R, Quaranta V, Harris LA, Tyson DR. A drift-diffusion checkpoint model predicts a highly variable and growth-factor-sensitive portion of the cell cycle G1 phase. PLoS One 2018; 13:e0192087. [PMID: 29432467 PMCID: PMC5809023 DOI: 10.1371/journal.pone.0192087] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Accepted: 01/17/2018] [Indexed: 11/28/2022] Open
Abstract
Even among isogenic cells, the time to progress through the cell cycle, or the intermitotic time (IMT), is highly variable. This variability has been a topic of research for several decades and numerous mathematical models have been proposed to explain it. Previously, we developed a top-down, stochastic drift-diffusion+threshold (DDT) model of a cell cycle checkpoint and showed that it can accurately describe experimentally-derived IMT distributions [Leander R, Allen EJ, Garbett SP, Tyson DR, Quaranta V. Derivation and experimental comparison of cell-division probability densities. J. Theor. Biol. 2014;358:129-135]. Here, we use the DDT modeling approach for both descriptive and predictive data analysis. We develop a custom numerical method for the reliable maximum likelihood estimation of model parameters in the absence of a priori knowledge about the number of detectable checkpoints. We employ this method to fit different variants of the DDT model (with one, two, and three checkpoints) to IMT data from multiple cell lines under different growth conditions and drug treatments. We find that a two-checkpoint model best describes the data, consistent with the notion that the cell cycle can be broadly separated into two steps: the commitment to divide and the process of cell division. The model predicts one part of the cell cycle to be highly variable and growth factor sensitive while the other is less variable and relatively refractory to growth factor signaling. Using experimental data that separates IMT into G1 vs. S, G2, and M phases, we show that the model-predicted growth-factor-sensitive part of the cell cycle corresponds to a portion of G1, consistent with previous studies suggesting that the commitment step is the primary source of IMT variability. These results demonstrate that a simple stochastic model, with just a handful of parameters, can provide fundamental insights into the biological underpinnings of cell cycle progression.
Collapse
Affiliation(s)
- Zack W. Jones
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, United States of America
| | - Rachel Leander
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro, TN 37132, United States of America
| | - Vito Quaranta
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States of America
| | - Leonard A. Harris
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States of America
| | - Darren R. Tyson
- Department of Biochemistry, Vanderbilt University School of Medicine, Nashville, TN 37232, United States of America
| |
Collapse
|
15
|
Schwarz C, Johnson A, Kõivomägi M, Zatulovskiy E, Kravitz CJ, Doncic A, Skotheim JM. A Precise Cdk Activity Threshold Determines Passage through the Restriction Point. Mol Cell 2018; 69:253-264.e5. [PMID: 29351845 PMCID: PMC5790185 DOI: 10.1016/j.molcel.2017.12.017] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 11/28/2017] [Accepted: 12/19/2017] [Indexed: 11/24/2022]
Abstract
At the restriction point (R), mammalian cells irreversibly commit to divide. R has been viewed as a point in G1 that is passed when growth factor signaling initiates a positive feedback loop of Cdk activity. However, recent studies have cast doubt on this model by claiming R occurs prior to positive feedback activation in G1 or even before completion of the previous cell cycle. Here we reconcile these results and show that whereas many commonly used cell lines do not exhibit a G1 R, primary fibroblasts have a G1 R that is defined by a precise Cdk activity threshold and the activation of cell-cycle-dependent transcription. A simple threshold model, based solely on Cdk activity, predicted with more than 95% accuracy whether individual cells had passed R. That a single measurement accurately predicted cell fate shows that the state of complex regulatory networks can be assessed using a few critical protein activities.
Collapse
Affiliation(s)
- Clayton Schwarz
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Amy Johnson
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | - Mardo Kõivomägi
- Department of Biology, Stanford University, Stanford, CA 94305, USA
| | | | | | - Andreas Doncic
- Department of Cell Biology & Green Center for Systems Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jan M Skotheim
- Department of Biology, Stanford University, Stanford, CA 94305, USA.
| |
Collapse
|
16
|
Matson JP, Cook JG. Cell cycle proliferation decisions: the impact of single cell analyses. FEBS J 2017; 284:362-375. [PMID: 27634578 PMCID: PMC5296213 DOI: 10.1111/febs.13898] [Citation(s) in RCA: 107] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 08/23/2016] [Accepted: 09/13/2016] [Indexed: 12/16/2022]
Abstract
Cell proliferation is a fundamental requirement for organismal development and homeostasis. The mammalian cell division cycle is tightly controlled to ensure complete and precise genome duplication and segregation of replicated chromosomes to daughter cells. The onset of DNA replication marks an irreversible commitment to cell division, and the accumulated efforts of many decades of molecular and cellular studies have probed this cellular decision, commonly called the restriction point. Despite a long-standing conceptual framework of the restriction point for progression through G1 phase into S phase or exit from G1 phase to quiescence (G0), recent technical advances in quantitative single cell analysis of mammalian cells have provided new insights. Significant intercellular heterogeneity revealed by single cell studies and the discovery of discrete subpopulations in proliferating cultures suggests the need for an even more nuanced understanding of cell proliferation decisions. In this review, we describe some of the recent developments in the cell cycle field made possible by quantitative single cell experimental approaches.
Collapse
Affiliation(s)
- Jacob P. Matson
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
| | - Jeanette G. Cook
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill. Chapel Hill, North Carolina 27599
| |
Collapse
|
17
|
Abstract
How and when eukaryotic cells make the irrevocable commitment to divide remain central questions in the cell-cycle field. Parallel studies in yeast and mammalian cells seemed to suggest analogous control mechanisms operating during the G1 phase—at Start or the restriction (R) point, respectively—to integrate nutritional and developmental signals and decide between distinct cell fates: cell-cycle arrest or exit versus irreversible commitment to a round of division. Recent work has revealed molecular mechanisms underlying this decision-making process in both yeast and mammalian cells but also cast doubt on the nature and timing of cell-cycle commitment in multicellular organisms. These studies suggest an expanded temporal window of mitogen sensing under certain growth conditions, illuminate unexpected obstacles and exit ramps on the path to full cell-cycle commitment, and raise new questions regarding the functions of cyclin-dependent kinases (CDKs) that drive G1 progression and S-phase entry.
Collapse
Affiliation(s)
- Robert P Fisher
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
18
|
Sun X, Zheng X, Zhang J, Zhou T, Yan G, Zhu W. Mathematical modeling reveals a critical role for cyclin D1 dynamics in phenotype switching during glioma differentiation. FEBS Lett 2015; 589:2304-11. [PMID: 26188547 DOI: 10.1016/j.febslet.2015.07.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2015] [Revised: 07/08/2015] [Accepted: 07/13/2015] [Indexed: 10/23/2022]
Abstract
Glioma differentiation therapy is a novel modality to increase anti-glioma effects using specific drugs to induce glioma cell differentiation to glia-like cells. However, the molecular mechanisms underlying glioma differentiation remain poorly understood. In this study, we built an experiment-integrated mathematical model for glioma differentiation signaling pathways. Our modeling and experimental analysis revealed that a "one-way-switch" bifurcation of cyclin D1 dynamics was critical for controlling the phenotypic transition of glioma cells. We also quantitatively evaluated drug combinations toward a synergistic therapeutic effect. These results provide insights into the molecular mechanisms underlying glioma differentiation and implications for the design of novel therapeutic targets in anti-cancer therapy.
Collapse
Affiliation(s)
- Xiaoqiang Sun
- Research Center of Bioinformatics, Zhong-shan School of Medicine, Sun Yat-Sen University, Guangzhou 510089, China; School of Mathematical and Computational Science, Sun Yat-Sen University, Guangzhou 510000, China.
| | - Xiaoke Zheng
- First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510000, China
| | - Jiajun Zhang
- School of Mathematical and Computational Science, Sun Yat-Sen University, Guangzhou 510000, China
| | - Tianshou Zhou
- School of Mathematical and Computational Science, Sun Yat-Sen University, Guangzhou 510000, China
| | - Guangmei Yan
- Department of Pharmacology, Zhong-shan School of Medicine, Sun Yat-Sen University, Guangzhou 510089, China
| | - Wenbo Zhu
- Department of Pharmacology, Zhong-shan School of Medicine, Sun Yat-Sen University, Guangzhou 510089, China.
| |
Collapse
|
19
|
Cogoi S, Zorzet S, Shchekotikhin AE, Xodo LE. Potent Apoptotic Response Induced by Chloroacetamidine Anthrathiophenediones in Bladder Cancer Cells. J Med Chem 2015; 58:5476-85. [DOI: 10.1021/acs.jmedchem.5b00409] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
- Susanna Cogoi
- Department
of Medical and Biological Sciences, University of Udine, P.le Kolbe
4, 33100 Udine, Italy
| | - Sonia Zorzet
- Department
of Life Science, University of Trieste, Via Giorgieri 7-9, 34100 Trieste, Italy
| | | | - Luigi E. Xodo
- Department
of Medical and Biological Sciences, University of Udine, P.le Kolbe
4, 33100 Udine, Italy
| |
Collapse
|
20
|
Dubon MJ, Park KS. Substance P enhances the proliferation and migration potential of murine bone marrow-derived mesenchymal stem cell-like cell lines. Exp Ther Med 2015; 9:1185-1191. [PMID: 25780407 PMCID: PMC4353778 DOI: 10.3892/etm.2015.2291] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2014] [Accepted: 02/03/2015] [Indexed: 01/14/2023] Open
Abstract
Due to the therapeutic characteristics of bone marrow (BM)-derived mesenchymal stem cells (MSCs), clinical trials are testing the use of autologous or allogeneic MSCs for the treatment of several conditions. These therapies require large numbers of MSCs and numerous studies are attempting to find substances that could enhance the egression of endogenous MSCs from the BM into the periphery and increase their proliferation in vivo and in vitro. It has been reported that substance P (SP) has the potential to increase the expansion of MSCs in vivo and to induce their mobilization from the BM into the periphery. The aim of the present study was to investigate the effects of SP on the migration and proliferation potential of two BM-derived MSC-like cell lines, ST2 and OP9. SP was found to induce the migration potential of ST2 cells in vitro. Furthermore, SP increased the proliferation of the MSCs cell line, OP9 cell line. Cyclin D1 expression was observed to increase in the OP9 cells, indicating the activation of the cell cycle in response to SP. The upstream signals involved in these phenomena have yet to be elucidated, although previous studies have proposed the activation of the extracellular signal-regulated kinase-1/2 and Wingless/β-catenin pathways as possible mediators of the cellular proliferation of human MSCs in response to SP. The present results therefore suggest that SP would facilitate the obtainment of higher numbers of endogenous MSCs from patients or donors and/or shorten the process of in vitro expansion that could cause the MSCs to undergo changes in their innate therapeutic characteristics prior to their use in therapy.
Collapse
Affiliation(s)
- Maria Jose Dubon
- Graduate School of Biotechnology, Kyung Hee University, Yong-In, Gyeonggi 466-701, Republic of Korea
| | - Ki-Sook Park
- East-West Medical Research Institute, Kyung Hee University, Seoul 130-701, Republic of Korea
| |
Collapse
|
21
|
Davis DM, Purvis JE. Computational analysis of signaling patterns in single cells. Semin Cell Dev Biol 2014; 37:35-43. [PMID: 25263011 DOI: 10.1016/j.semcdb.2014.09.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 09/11/2014] [Accepted: 09/13/2014] [Indexed: 01/19/2023]
Abstract
Signaling proteins are flexible in both form and function. They can bind to multiple molecular partners and integrate diverse types of cellular information. When imaged by time-lapse microscopy, many signaling proteins show complex patterns of activity or localization that vary from cell to cell. This heterogeneity is so prevalent that it has spurred the development of new computational strategies to analyze single-cell signaling patterns. A collective observation from these analyses is that cells appear less heterogeneous when their responses are normalized to, or synchronized with, other single-cell measurements. In many cases, these transformed signaling patterns show distinct dynamical trends that correspond with predictable phenotypic outcomes. When signaling mechanisms are unclear, computational models can suggest putative molecular interactions that are experimentally testable. Thus, computational analysis of single-cell signaling has not only provided new ways to quantify the responses of individual cells, but has helped resolve longstanding questions surrounding many well-studied human signaling proteins including NF-κB, p53, ERK1/2, and CDK2. A number of specific challenges lie ahead for single-cell analysis such as quantifying the contribution of non-cell autonomous signaling as well as the characterization of protein signaling dynamics in vivo.
Collapse
Affiliation(s)
- Denise M Davis
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599-7264, United States
| | - Jeremy E Purvis
- Department of Genetics and Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, 120 Mason Farm Road, Chapel Hill, NC 27599-7264, United States.
| |
Collapse
|
22
|
PP2A-mediated regulation of Ras signaling in G2 is essential for stable quiescence and normal G1 length. Mol Cell 2014; 54:932-945. [PMID: 24857551 DOI: 10.1016/j.molcel.2014.04.023] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2013] [Revised: 03/04/2014] [Accepted: 04/07/2014] [Indexed: 12/23/2022]
Abstract
Quiescence (G0) allows cycling cells to reversibly cease proliferation. A decision to enter quiescence is suspected of occurring early in G1, before the restriction point (R). Surprisingly, we have identified G2 as an interval during which inhibition of the protein phosphatase PP2A results in failure to exhibit stable quiescence. This effect is accompanied by shortening of the ensuing G1. The PP2A subcomplex required for stable G0 contains the B56γ B subunit. After PP2A inhibition in G2, aberrant overexpression of cyclin E occurs during mitosis and is responsible for overriding quiescence. Strikingly, suppression of Ras signaling re-establishes normal cyclin E levels during M and restores G0. These data point to PP2A-B56γ-driven Ras signaling modulation in G2 as essential for suppressing aberrant cyclin E expression during mitosis and thereby achieving normal G0 control. Thus, G2 is an interval during which the length and growth factor dependence of the next G1 interval are established.
Collapse
|
23
|
Zhang B, Dai M, Li QJ, Zhuang Y. Tracking proliferative history in lymphocyte development with cre-mediated sister chromatid recombination. PLoS Genet 2013; 9:e1003887. [PMID: 24204301 PMCID: PMC3814321 DOI: 10.1371/journal.pgen.1003887] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 09/03/2013] [Indexed: 11/18/2022] Open
Abstract
Tracking and isolating live cells based on their proliferative history in live animals remains a technical challenge in animal studies. We have designed a genetic marking system for tracking the proliferative frequency and history of lymphocytes during their development and homeostatic maintenance. This system is based on activation of a fluorescent marker after Cre-dependent recombination between sister chromatids at a specially designed tandem loxP site, named Tlox. We have demonstrated the utility of the Tlox system in tracking proliferative windows of B and T lymphocyte development. We have further applied the Tlox system in the analysis of the proliferative behavior and homeostatic maintenance of Vγ1.1 positive γδ T cells. Our data show that Vγ1.1 T cells generated in neonatal but not adult life are able to expand in the thymus. The expanded Vγ1.1 T cells are preferentially maintained in the liver but not in lymphoid organs. It has been shown that numbers of Vγ1.1 T cells were dramatically increased in the lymphoid organs of Id3 deficient mice. By combining BrdU and Tlox assays we show that this phenotype is primarily due to enhanced neonatal expansion and subsequent retention of Vγ1.1 T cells. Thus, the Tlox system provides a new genetic tool to track clonal expansion within a defined cell population or tissue type in live animals.
Collapse
Affiliation(s)
- Baojun Zhang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Meifang Dai
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
| | - Qi-Jing Li
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (QJL); (YZ)
| | - Yuan Zhuang
- Department of Immunology, Duke University Medical Center, Durham, North Carolina, United States of America
- * E-mail: (QJL); (YZ)
| |
Collapse
|
24
|
Spencer SL, Cappell SD, Tsai FC, Overton KW, Wang CL, Meyer T. The proliferation-quiescence decision is controlled by a bifurcation in CDK2 activity at mitotic exit. Cell 2013; 155:369-83. [PMID: 24075009 DOI: 10.1016/j.cell.2013.08.062] [Citation(s) in RCA: 483] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Revised: 06/04/2013] [Accepted: 08/29/2013] [Indexed: 01/21/2023]
Abstract
Tissue homeostasis in metazoans is regulated by transitions of cells between quiescence and proliferation. The hallmark of proliferating populations is progression through the cell cycle, which is driven by cyclin-dependent kinase (CDK) activity. Here, we introduce a live-cell sensor for CDK2 activity and unexpectedly found that proliferating cells bifurcate into two populations as they exit mitosis. Many cells immediately commit to the next cell cycle by building up CDK2 activity from an intermediate level, while other cells lack CDK2 activity and enter a transient state of quiescence. This bifurcation is directly controlled by the CDK inhibitor p21 and is regulated by mitogens during a restriction window at the end of the previous cell cycle. Thus, cells decide at the end of mitosis to either start the next cell cycle by immediately building up CDK2 activity or to enter a transient G0-like state by suppressing CDK2 activity.
Collapse
Affiliation(s)
- Sabrina L Spencer
- Department of Chemical and Systems Biology, Stanford University, Stanford, CA 94305, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Varshney S, Bhadada SK, Saikia UN, Sachdeva N, Behera A, Arya AK, Sharma S, Bhansali A, Mithal A, Rao SD. Simultaneous expression analysis of vitamin D receptor, calcium-sensing receptor, cyclin D1, and PTH in symptomatic primary hyperparathyroidism in Asian Indians. Eur J Endocrinol 2013; 169:109-16. [PMID: 23660642 DOI: 10.1530/eje-13-0085] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND To explore underlying molecular mechanisms in the pathogenesis of symptomatic sporadic primary hyperparathyroidism (PHPT). MATERIALS AND METHODS Forty-one parathyroid adenomas from patients with symptomatic PHPT and ten normal parathyroid glands either from patients with PHPT (n=3) or from euthyroid patients without PHPT during thyroid surgery (n=7) were analyzed for vitamin D receptor (VDR), calcium-sensing receptor (CASR), cyclin D1 (CD1), and parathyroid hormone (PTH) expressions. The protein expressions were assessed semiquantitatively by immunohistochemistry, based on percentage of positive cells and staining intensity, and confirmed by quantitative real-time PCR. RESULTS Immunohistochemistry revealed significant reductions in VDR (both nuclear and cytoplasmic) and CASR expressions and significant increases in CD1 and PTH expressions in adenomatous compared with normal parathyroid tissue. Consistent with immunohistochemistry findings, both VDR and CASR mRNAs were reduced by 0.36- and 0.45-fold change (P<0.001) and CD1 and PTH mRNAs were increased by 9.4- and 17.4-fold change respectively (P<0.001) in adenomatous parathyroid tissue. PTH mRNA correlated with plasma PTH (r=0.864; P<0.001), but not with adenoma weight, while CD1 mRNA correlated with adenoma weight (r=0.715; P<0.001). There were no correlations between VDR and CASR mRNA levels and serum Ca, plasma intact PTH, or 25-hydroxyvitamin D levels. In addition, there was no relationship between the decreases in VDR and CASR mRNA expressions and the increases in PTH and CD1 mRNA expressions. CONCLUSIONS The expression of both VDR and CASR are reduced in symptomatic PHPT in Asian Indians. In addition, CD1 expression was greatly increased and correlated with adenoma weight, implying a potential role for CD1 in adenoma growth and differential clinical expression of PHPT.
Collapse
Affiliation(s)
- Shweta Varshney
- Department of Endocrinology, Postgraduate Institute of Medical Education and Research, 4th Floor, F-Block, Room No. 2, Chandigarh 160012, India
| | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sellathurai J, Cheedipudi S, Dhawan J, Schrøder HD. A novel in vitro model for studying quiescence and activation of primary isolated human myoblasts. PLoS One 2013; 8:e64067. [PMID: 23717533 PMCID: PMC3662676 DOI: 10.1371/journal.pone.0064067] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 04/09/2013] [Indexed: 01/01/2023] Open
Abstract
Skeletal muscle stem cells, satellite cells, are normally quiescent but become activated upon muscle injury. Recruitment of resident satellite cells may be a useful strategy for treatment of muscle disorders, but little is known about gene expression in quiescent human satellite cells or the mechanisms involved in their early activation. We have developed a method to induce quiescence in purified primary human myoblasts isolated from healthy individuals. Analysis of the resting state showed absence of BrdU incorporation and lack of KI67 expression, as well as the extended kinetics during synchronous reactivation into the cell cycle, confirming arrest in the G0 phase. Reactivation studies showed that the majority (>95%) of the G0 arrested cells were able to re-enter the cell cycle, confirming reversibility of arrest. Furthermore, a panel of important myogenic factors showed expression patterns similar to those reported for mouse satellite cells in G0, reactivated and differentiated cultures, supporting the applicability of the human model. In addition, gene expression profiling showed that a large number of genes (4598) were differentially expressed in cells activated from G0 compared to long term exponentially proliferating cultures normally used for in vitro studies. Human myoblasts cultured through many passages inevitably consist of a mixture of proliferating and non-proliferating cells, while cells activated from G0 are in a synchronously proliferating phase, and therefore may be a better model for in vivo proliferating satellite cells. Furthermore, the temporal propagation of proliferation in these synchronized cultures resembles the pattern seen in vivo during regeneration. We therefore present this culture model as a useful and novel condition for molecular analysis of quiescence and reactivation of human myoblasts.
Collapse
Affiliation(s)
- Jeeva Sellathurai
- Institute of Clinical Research, SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense, Denmark
| | | | - Jyotsna Dhawan
- Institute for Stem Cell Biology and Regenerative Medicine (InStem), National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bangalore, India
| | - Henrik Daa Schrøder
- Institute of Clinical Research, SDU Muscle Research Cluster (SMRC), University of Southern Denmark, Odense, Denmark
- Department of Clinical Pathology, Odense University Hospital, Odense, Denmark
| |
Collapse
|
27
|
Anticancer effects of maple syrup phenolics and extracts on proliferation, apoptosis, and cell cycle arrest of human colon cells. J Funct Foods 2012. [DOI: 10.1016/j.jff.2011.10.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
28
|
Stacey DW. Three Observations That Have Changed Our Understanding of Cyclin D1 and p27 in Cell Cycle Control. Genes Cancer 2011; 1:1189-99. [PMID: 21779442 DOI: 10.1177/1947601911403475] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Our understanding of cell cycle control has been based largely upon studies of synchronized cultures, often focused upon the early stages of the cell cycle following stimulation of quiescent cultures. These studies showed that cyclin D1 and p27(Kip1) (p27) each respond to the growth environment of the cell and together control entry into the cell cycle. In contrast, all cell cycle phases were considered in these studies of actively growing cultures, including events leading to withdrawal from the cell cycle. This approach relies upon the techniques of microinjection, quantitative image analysis, and time-lapse microscopy. The results provide critical new detail to our understanding of the roles of cyclin D1 and p27 in cell cycle regulation. Three critical observations resulting from this work will be described here to demonstrate that 1) cyclin D1 levels oscillate through the normal cell cycle, 2) checkpoint kinases are able to suppress cyclin D1 during S phase, and 3) the level of p27 is determined by a dynamic interaction between cyclin D1 and p27 so as to determine the rate of cell cycle progression. Based upon these observations, a model of cell cycle control is presented in which ras activity stimulates cyclin D1 during G2 phase, resulting in commitment of the cell to continued cell cycle progression. During G1 phase, ras activity suppresses the level of p27 protein, most of which is bound to cyclin D1, resulting in regulation of the rate of proliferation. This model predicted the involvement of checkpoint kinases in regulating cyclin D1 and the role of checkpoint kinases in the protection of neural cells against reactive oxygen. The substantiation of these 2 predictions serves as general validation of the model.
Collapse
Affiliation(s)
- Dennis W Stacey
- Department of Molecular Medicine, Case Western Reserve University, and Department of Molecular Genetics, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
29
|
Contente S, Yeh TJA, Friedman RM. H-ras localizes to cell nuclei and varies with the cell cycle. Genes Cancer 2011; 2:166-72. [PMID: 21779490 PMCID: PMC3111243 DOI: 10.1177/1947601911405042] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Revised: 02/18/2011] [Accepted: 03/04/2011] [Indexed: 01/10/2023] Open
Abstract
H-Ras functions as a signal switch molecule in numerous signaling pathways in the cytoplasm, requiring H-Ras localization to the inner surface of the cytoplasmic membrane, and H-Ras is considered to be a cytoplasmic protein. Immunoblot studies of cells transformed by overexpression of c-H-ras indicated that H-Ras protein was present in both cytoplasmic and nuclear extracts, suggesting a possible correlation of nuclear H-Ras and cellular transformation. Unexpectedly, additional studies revealed that H-Ras protein was also present in the nuclei of nontransformed and primary mouse cells, which do not overexpress H-Ras. Mouse fibroblast NIH 3T3 cells, L cells, and a primary fibroblast line all had H-Ras present in both cytoplasmic and nuclear extracts. Nuclear extracts of cells synchronized by growth without serum displayed an increasing amount of H-Ras and cyclin D1 as cells grew after serum addition. Treatment with farnesyltransferase inhibitor caused loss of H-Ras from the nucleus. Immunofluorescence in situ studies of nuclei from synchronized cultures showed that H-Ras protein appeared in and disappeared from the nuclei as the cells moved through the growth cycle. This cycling occurred in both nontransformed and ras-transformed cells. Flow cytometry measurements on parallel cultures revealed that the time point at which the greatest percentage of cells were in S phase, for each line, corresponded to appearance of a noticeably stronger in situ signal for H-Ras. H-Ras may participate in nuclear signaling pathways associated with replication in addition to its cytoplasmic signaling functions.
Collapse
Affiliation(s)
- Sara Contente
- Department of Pathology, F. Edward Hébert School of Medicine, and United States Military Cancer Institute, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | | | | |
Collapse
|
30
|
Hitomi M, Stacey DW. The checkpoint kinase ATM protects against stress-induced elevation of cyclin D1 and potential cell death in neurons. Cytometry A 2010; 77:524-33. [DOI: 10.1002/cyto.a.20885] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
31
|
Burhans WC, Heintz NH. The cell cycle is a redox cycle: linking phase-specific targets to cell fate. Free Radic Biol Med 2009; 47:1282-93. [PMID: 19486941 DOI: 10.1016/j.freeradbiomed.2009.05.026] [Citation(s) in RCA: 261] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2009] [Revised: 05/19/2009] [Accepted: 05/22/2009] [Indexed: 11/22/2022]
Abstract
Reactive oxygen species (ROS) regulate the strength and duration of signaling through redox-dependent signal transduction pathways via the cyclic oxidation/reduction of cysteine residues in kinases, phosphatases, and other regulatory factors. Signaling circuits may be segregated in organelles or other subcellular domains with distinct redox states, permitting them to respond independently to changes in the oxidation state of two major thiol reductants, glutathione and thioredoxin. Studies in yeast, and in complex eukaryotes, show that oscillations in oxygen consumption, energy metabolism, and redox state are intimately integrated with cell cycle progression. Because signaling pathways play specific roles in different phases of the cell cycle and the hierarchy of redox-dependent regulatory checkpoints changes during cell cycle progression, the effects of ROS on cell fate vary during the cell cycle. In G1, ROS stimulate mitogenic pathways that control the activity of cyclin-dependent kinases (CDKs) and phosphorylation of the retinoblastoma protein (pRB), thereby regulating S-phase entry. In response to oxidative stress, Nrf2 and Foxo3a promote cell survival by inducing the expression of antioxidant enzymes and factors involved in cell cycle withdrawal, such as the cyclin-dependent kinase inhibitor (CKI) p27. In S phase, ROS induce S-phase arrest via PP2A-dependent dephosphorylation of pRB. In precancerous cells, unconstrained mitogenic signaling by activated oncogenes induces replication stress in S phase, which activates the DNA-damage response and induces cell senescence. A number of studies suggest that interactions of ROS with the G1 CDK/CKI network play a fundamental role in senescence, which is considered a barrier to tumorigenesis. Adaptive responses and loss of checkpoint proteins such as p53 and p16(INK4a) allow tumor cells to tolerate constitutive mitogenic signaling and enhanced production of ROS, leading to altered redox status in many fully transformed cells. Alterations in oxidant and energy metabolism of cancer cells have emerged as fertile ground for new therapeutic targets. The present challenge is to identify redox-dependent targets relevant to each cell cycle phase, to understand how these targets control fate decisions, and to describe the mechanisms that link metabolism to cell cycle progression.
Collapse
Affiliation(s)
- William C Burhans
- Department of Molecular & Cellular Biology, Roswell Park Cancer Institute, Buffalo, NY 14263, USA
| | | |
Collapse
|
32
|
Cilla A, González-Sarrías A, Tomás-Barberán FA, Espín JC, Barberá R. Availability of polyphenols in fruit beverages subjected to in vitro gastrointestinal digestion and their effects on proliferation, cell-cycle and apoptosis in human colon cancer Caco-2 cells. Food Chem 2009. [DOI: 10.1016/j.foodchem.2008.10.019] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
33
|
Sa G, Das T. Anti cancer effects of curcumin: cycle of life and death. Cell Div 2008; 3:14. [PMID: 18834508 PMCID: PMC2572158 DOI: 10.1186/1747-1028-3-14] [Citation(s) in RCA: 258] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2008] [Accepted: 10/03/2008] [Indexed: 01/14/2023] Open
Abstract
Increasing knowledge on the cell cycle deregulations in cancers has promoted the introduction of phytochemicals, which can either modulate signaling pathways leading to cell cycle regulation or directly alter cell cycle regulatory molecules, in cancer therapy. Most human malignancies are driven by chromosomal translocations or other genetic alterations that directly affect the function of critical cell cycle proteins such as cyclins as well as tumor suppressors, e.g., p53. In this respect, cell cycle regulation and its modulation by curcumin are gaining widespread attention in recent years. Extensive research has addressed the chemotherapeutic potential of curcumin (diferuloylmethane), a relatively non-toxic plant derived polyphenol. The mechanisms implicated are diverse and appear to involve a combination of cell signaling pathways at multiple levels. In the present review we discuss how alterations in the cell cycle control contribute to the malignant transformation and provide an overview of how curcumin targets cell cycle regulatory molecules to assert anti-proliferative and/or apoptotic effects in cancer cells. The purpose of the current article is to present an appraisal of the current level of knowledge regarding the potential of curcumin as an agent for the chemoprevention of cancer via an understanding of its mechanism of action at the level of cell cycle regulation. Taken together, this review seeks to summarize the unique properties of curcumin that may be exploited for successful clinical cancer prevention.
Collapse
Affiliation(s)
- Gaurisankar Sa
- Division of Molecular Medicine, Bose Institute, P-1/12 CIT Scheme VII M, Kolkata, 700054, India.
| | | |
Collapse
|
34
|
Abstract
When cell cycle studies are performed following cell cycle synchronization, it is possible that critical properties of an actively cycling cell will be overlooked. For this reason past studies have not revealed critical aspects of cell cycle control; such as how a cell determines when to exit the cell cycle, or how rapidly it should cycle. To address these challenging questions we have developed a procedure to quantitate fluorescent stains in a monolayer culture, where nuclear fluorescence and cell cycle history can be assessed with accuracy on a cell by cell basis. The cell cycle position of each cell can be determined by analyzing DNA and BrdU levels. The behavior of cells in a given cell cycle position can then be studied by quantitating up to two other stained markers. When the microinjection of siRNA, neutralizing antibodies, and expression plasmids are coupled with quantitative image analysis, these cell cycle studies can be conducted following alterations in the expression levels of selected cellular targets. With these techniques we have discovered critical aspects of cell cycle control; including how cyclin D1 levels vary through the cell cycle, the molecular mechanisms governing these changes, and the biological implications of changes in cyclin D1 concentration in various cell cycle stages. Our studies with cyclin D1, coupled with similar studies of p27Kip1, form the basis of an entirely new model of cell cycle control proposed here. This model explains how cell cycle progression is terminated, and how the length of the cell cycle is regulated.
Collapse
Affiliation(s)
- Dennis W Stacey
- Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, Ohio 44195, USA.
| | | |
Collapse
|
35
|
Analysis of a molecular structured population model with possible polynomial growth for the cell division cycle. ACTA ACUST UNITED AC 2008. [DOI: 10.1016/j.mcm.2007.06.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
36
|
Bekkal Brikci F, Clairambault J, Ribba B, Perthame B. An age-and-cyclin-structured cell population model for healthy and tumoral tissues. J Math Biol 2007; 57:91-110. [DOI: 10.1007/s00285-007-0147-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2006] [Revised: 11/13/2007] [Indexed: 10/22/2022]
|
37
|
Hashem MA, Bhandari DP, Kang SK, Lee BC. Cell cycle analysis and interspecies nuclear transfer of in vitro cultured skin fibroblasts of the Siberian tiger (Panthera tigris Altaica). Mol Reprod Dev 2007; 74:403-11. [PMID: 17075834 DOI: 10.1002/mrd.20528] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
The present study was conducted to examine the effect of cell culture conditions, antioxidants, protease inhibitors (PI), and different levels of dimethylsulfoxide (DMSO) for the promotion of synchronization of different cell cycles of Siberian tiger skin fibroblasts. We also compared the ability of somatic cell nuclei of the Siberian tiger in pig cytoplasts and to support early development after reconstruction. Cell cycle synchronization between nuclear donor and recipient cells is considered to be one of the most crucial factors for successful cloning. Five experiments were performed each with a one-way completely randomized design involving three replicates of all treatments. Least significant difference (LSD) was used to determine variation among treatment groups. Experiment I focused in the effects of cycling, serum starved and fully confluent stages of Siberian tiger cells on different cell cycles. In Experiment II, the effects of different antioxidants like beta-Mercaptoethanol (beta-ME, 10 microM), cysteine (2 mM), and glutathione (2 mM) were examined after cells were fully confluent without serum starvation for 4 hr. In Experiment III, three PI, namely 6-dimethylaminopurine (6-DMAP, 2 mM), cycloheximide (7.5 microg/ml) and cytochalasin B (7.5 microg/ml) were used in the sane manner as in Experiment II. In Experiment IV, different levels of DMSO at 0%, 0.5%, 1.0%, and 2.5% were tested on different cell cycle stages of Siberian tiger examined by Flowcytometry (FACS). In Experiment I, 67.2% of the Siberian tiger skin fibroblasts reached the G0/G1 stage (2C DNA content) in fully confluent conditions which was more than the cycling (49.8%) and serum starved (SS) medium (65.5%; P < 0.05). Among the chemically treated group, glutathione (72.6%) and cycloheximide (71.3%) had little bit better results for the synchronization of G0 + G1 phases than serum starved and fully confluent. After nuclear transfer we did not see any significant differences on the development of tiger-porcine reconstructed embryos at cycling, SS and fully confluent. Data indicate that prolonged culture of cells in the absence of serum as well as using different chemicals for this experiment does not imply a shift in the percentage of cells that enter G0/G1 and that confluency is sufficient to induce quiescence. This finding can be beneficial in nuclear transfer programs in Siberian tiger, because there are negative effects, such as apoptosis associated with serum starvation.
Collapse
Affiliation(s)
- Md Abul Hashem
- Department of Theriogenology and Biotechnology, College of Veterinary Medicine, Seoul National University, Seoul, Korea
| | | | | | | |
Collapse
|
38
|
Yang K, Hitomi M, Stacey DW. Variations in cyclin D1 levels through the cell cycle determine the proliferative fate of a cell. Cell Div 2006; 1:32. [PMID: 17176475 PMCID: PMC1769361 DOI: 10.1186/1747-1028-1-32] [Citation(s) in RCA: 166] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2006] [Accepted: 12/18/2006] [Indexed: 12/02/2022] Open
Abstract
We present evidence that variations in cyclin D1 levels through the cell cycle are essential for continuing proliferation. Cyclin D1 levels must be high during G1 phase for a cell to initiate DNA synthesis, but then must be suppressed to low levels during S phase to allow for efficient DNA synthesis. This suppression during S phase is apparently regulated by cell cycle position alone and occurs automatically during each cell cycle. If the cell is to continue proliferating, cyclin D1 levels must be induced once again during G2 phase. This induction depends upon the activity of proliferative signaling molecules, and ensures that the extracellular environment continues to be conducive for growth. We propose that the suppression of cyclin D1 levels during each S phase ensures that the subsequent induction during G2 phase, and the resulting commitment to continuing proliferation, is closely linked to the cellular growth environment.
Collapse
Affiliation(s)
- Ke Yang
- The Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave. Cleveland OH, 44072, USA
| | - Masahiro Hitomi
- The Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave. Cleveland OH, 44072, USA
| | - Dennis W Stacey
- The Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Ave. Cleveland OH, 44072, USA
| |
Collapse
|
39
|
Zhang YA, Nemunaitis J, Samuel SK, Chen P, Shen Y, Tong AW. Antitumor Activity of an Oncolytic Adenovirus-Delivered Oncogene Small Interfering RNA. Cancer Res 2006; 66:9736-43. [PMID: 17018633 DOI: 10.1158/0008-5472.can-06-1617] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Despite successes in animal models, cancer gene therapy with small interfering RNAs (siRNA) is hindered by the lack of an optimal delivery platform. We examined the applicability of the replication-competent, oncolytic adenovirus, ONYX-411, to deliver a mutant K-ras siRNA transgene to human cancer cells. Proof-of-principle studies showed an additive tumor growth-inhibitory response through siRNA-mediated K-ras knockdown and ONYX-411-mediated cancer cell lysis. A novel construct, termed Internavec (for interfering RNA vector), was generated by cloning a K-ras(v12)-specific siRNA(ras-4) hairpin construct under the control of the human H1 promoter into the deleted E3b region of ONYX-411. Internavec acquired an increase in potency of approximately 10-fold in human cancer cells expressing the relevant K-ras(v12) mutation (H79, H441, and SW480), as defined by a reduction in the effective dose needed to achieve 50% growth inhibition (ED(50)). Internavec remained attenuated in nonmalignant epithelial cells. Daily intratumoral injections of Internavec (five daily injections of 1 x 10(8) plaque-forming units) significantly reduced the growth of s.c. H79 pancreatic cancer xenografts in nu/nu mice by 85.5%, including complete growth suppression in three of five mice. Parental ONYX-411 or ONYX-411-siRNA(GFP) was markedly less effective (47.8% growth reduction, P = 0.03; and 44.1% growth reduction, P = 0.03, respectively). siRNA(ras) transgene activity contributed to cell cycle blockage, increased apoptosis, and marked down-regulation of Ras signaling-related gene expression (AKT2, GSK3 beta, E2F2, and MAP4K5). These findings indicate that Internavec can generate a two-pronged attack on tumor cells through oncogene knockdown and viral oncolysis, resulting in a significantly enhanced antitumor outcome.
Collapse
Affiliation(s)
- Yu-An Zhang
- The Mary Crowley Medical Research Center, Dallas, Texas
| | | | | | | | | | | |
Collapse
|
40
|
Yang K, Guo Y, Stacey WC, Harwalkar J, Fretthold J, Hitomi M, Stacey DW. Glycogen synthase kinase 3 has a limited role in cell cycle regulation of cyclin D1 levels. BMC Cell Biol 2006; 7:33. [PMID: 16942622 PMCID: PMC1592484 DOI: 10.1186/1471-2121-7-33] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Accepted: 08/30/2006] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND The expression level of cyclin D1 plays a vital role in the control of proliferation. This protein is reported to be degraded following phosphorylation by glycogen synthase kinase 3 (GSK3) on Thr-286. We recently showed that phosphorylation of Thr-286 is responsible for a decline in cyclin D1 levels during S phase, an event required for efficient DNA synthesis. These studies were undertaken to test the possibility that phosphorylation by GSK3 is responsible for the S phase specific decline in cyclin D1 levels, and that this event is regulated by the phosphatidylinositol 3-kinase (PI3K)/AKT signaling pathway which controls GSK3. RESULTS We found, however, that neither PI3K, AKT, GSK3, nor proliferative signaling activity in general is responsible for the S phase decline in cyclin D1 levels. In fact, the activity of these signaling kinases does not vary through the cell cycle of proliferating cells. Moreover, we found that GSK3 activity has little influence over cyclin D1 expression levels during any cell cycle phase. Inhibition of GSK3 activity by siRNA, LiCl, or other chemical inhibitors failed to influence cyclin D1 phosphorylation on Thr-286, even though LiCl efficiently blocked phosphorylation of beta-catenin, a known substrate of GSK3. Likewise, the expression of a constitutively active GSK3 mutant protein failed to influence cyclin D1 phosphorylation or total protein expression level. CONCLUSION Because we were unable to identify any proliferative signaling molecule or pathway which is regulated through the cell cycle, or which is able to influence cyclin D1 levels, we conclude that the suppression of cyclin D1 levels during S phase is regulated by cell cycle position rather than signaling activity. We propose that this mechanism guarantees the decline in cyclin D1 levels during each S phase; and that in so doing it reduces the likelihood that simple over expression of cyclin D1 can lead to uncontrolled cell growth.
Collapse
Affiliation(s)
- Ke Yang
- From the Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland OH, USA
| | - Yang Guo
- From the Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland OH, USA
| | - William C Stacey
- The Department of Neurology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Jyoti Harwalkar
- From the Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland OH, USA
| | - Jonathan Fretthold
- From the Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland OH, USA
| | - Masahiro Hitomi
- From the Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland OH, USA
| | - Dennis W Stacey
- From the Department of Molecular Genetics, The Lerner Research Institute, The Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland OH, USA
| |
Collapse
|
41
|
Liu F, Lee WH. CtIP activates its own and cyclin D1 promoters via the E2F/RB pathway during G1/S progression. Mol Cell Biol 2006; 26:3124-34. [PMID: 16581787 PMCID: PMC1446954 DOI: 10.1128/mcb.26.8.3124-3134.2006] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cell cycle progression from G(1) to S phase is mainly controlled by E2F transcription factors and RB family proteins. Previously we showed that the presence of CtIP is essential for G(1)/S transition in primary mouse blastocysts, as well as in NIH 3T3 cells. However, how CtIP executes this function remains to be elucidated. Here we show that in NIH 3T3 cells the expression of CtIP is regulated by the E2F/RB pathway during late G(1) and S phases. The presence of wild-type CtIP, but not the E157K mutant form, which failed to interact with RB, enhanced its own promoter activity. Chromatin immunoprecipitation analysis indicated that the recruitment of CtIP to its promoter occurs concomitantly with TFIIB, a component of the RNA polymerase II complex, and with dissociation of RB from the promoter during late G(1) and G(1)/S transition. Similar positive regulation of cyclin D1 expression by CtIP was also observed. Consistently, cells expressing the CtIP(E157K) protein alone exhibited growth retardation, an increase in the G(1) population, and a decrease in the S-phase population. Taken together, these results suggest that, contrary to the postulated universal corepressor role, CtIP activates a subset of E2F-responsive promoters by releasing RB-imposed repression and therefore promotes G(1)/S progression.
Collapse
Affiliation(s)
- Feng Liu
- Department of Biological Chemistry, 839 Medical Science Court, 124 Sprague Hall, University of California, Irvine, CA 92697, USA
| | | |
Collapse
|
42
|
Chen BX, Erlanger BF. Cell cycle inhibition by an anti-cyclin D1 antibodychemically modified for intracellular delivery. Cancer Lett 2006; 244:71-5. [PMID: 16442707 DOI: 10.1016/j.canlet.2005.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2005] [Accepted: 12/04/2005] [Indexed: 11/21/2022]
Abstract
Antibodies, especially monoclonal antibodies, are highly specific for their target antigens and have found extensive clinical application in the treatment of infectious diseases and neoplasia. However, they have a major shortcoming which, if overcome, would greatly expand their utility: an inability to penetrate the outer membrane of cells and act on intracellular targets. We demonstrated previously that this deficiency could be overcome by covalent linkage of an oligoarginine sequence to the conserved carbohydrate moiety present in the CH2 region of immunoglobulins. Immune specificity was maintained but no attempt was made to test for biological activity related to specificity. Here, we report that a polyarginated monoclonal anti-cyclin D1 enters cells and inhibits cell cycle progression. We demonstrate this with NIH 3T3 cells and with two tumor cell lines, HT29 and SW480. As many tumors overexpress cyclin D1, an intracellular anti-cyclin D1, properly targeted, has the potential to be a novel broad range inhibitor of tumor cell multiplication. Moreover, success with intracellular anti-cyclin D1 suggests that polyarginated antibodies, in general, could be a new, widely applicable experimental tool to investigate and influence intracellular processes, whether native to cells or introduced into cells by outside entities such as viruses.
Collapse
Affiliation(s)
- Bi-Xing Chen
- Department of Microbiology, Columbia University, 701 West 168 Street, New York, NY 10032, USA
| | | |
Collapse
|
43
|
Choudhuri T, Pal S, Das T, Sa G. Curcumin Selectively Induces Apoptosis in Deregulated Cyclin D1-expressed Cells at G2 Phase of Cell Cycle in a p53-dependent Manner. J Biol Chem 2005; 280:20059-68. [PMID: 15738001 DOI: 10.1074/jbc.m410670200] [Citation(s) in RCA: 235] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Curcumin (diferuloylmethane) is known to induce apoptosis in tumor cells. In asynchronous cultures, with time-lapse video-micrography in combination with quantitative fluorescence microscopy, we have demonstrated that curcumin induces apoptosis at G(2) phase of cell cycle in deregulated cyclin D1-expressed mammary epithelial carcinoma cells, leaving its normal counterpart unaffected. In our search toward delineating the molecular mechanisms behind such differential activities of curcumin, we found that it selectively increases p53 expression at G(2) phase of carcinoma cells and releases cytochrome c from mitochondria, which is an essential requirement for apoptosis. Further experiments using p53-null as well as dominant-negative and wild-type p53-transfected cells have established that curcumin induces apoptosis in carcinoma cells via a p53-dependent pathway. On the other hand, curcumin reversibly inhibits normal mammary epithelial cell cycle progression by down-regulating cyclin D1 expression and blocking its association with Cdk4/Cdk6 as well as by inhibiting phosphorylation and inactivation of retinoblastoma protein. In addition, curcumin significantly up-regulates cell cycle inhibitory protein (p21Waf-1) in normal cells and arrests them in G(0) phase of cell cycle. Therefore, these cells escape from curcumin-induced apoptosis at G(2) phase. Interestingly, these processes remain unaffected by curcumin in carcinoma cells where cyclin D1 expression is high. Similarly, in ectopically overexpressed system, curcumin cannot down-regulate cyclin D1 and thus block cell cycle progression. Hence, these cells progress into G(2) phase and undergo apoptosis. These observations together suggest that curcumin may have a possible therapeutic potential in breast cancer patients.
Collapse
Affiliation(s)
- Tathagata Choudhuri
- Bose Institute, P-1/12 Calcutta Improvement Trust Scheme VII M, Kolkata, India
| | | | | | | |
Collapse
|
44
|
Burch PM, Heintz NH. Redox regulation of cell-cycle re-entry: cyclin D1 as a primary target for the mitogenic effects of reactive oxygen and nitrogen species. Antioxid Redox Signal 2005; 7:741-51. [PMID: 15890020 DOI: 10.1089/ars.2005.7.741] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Reactive oxygen and nitrogen species inhibit or promote cell proliferation by modulating the cell signaling pathways that dictate decisions between cell survival, proliferation, and death. In the growth factor-dependent pathways that regulate mitogenesis, numerous positive and negative effectors of signaling are influenced by physiological fluctuations of oxidants, including receptor tyrosine kinases, small GTPases, mitogen-activated protein kinases, protein phosphatases, and transcription factors. The same mitogenic pathways that are sensitive to oxidant levels also directly regulate the expression of cyclin D1, a labile factor required for progression through the G1 phase on the cell cycle. Because the transition from G0 to G1 is the only phase of the cell cycle that is not regulated by cyclin-dependent kinases, but rather by redox-dependent signaling pathways, expression of cyclin D1 represents a primary regulatory node for the dose-dependent effects of oxidants on the induction of cell growth. We suggest that expression of cyclin D1 represents a useful marker for assessing the integration of proliferative and growth inhibitory effects of oxidants on the redox-dependent signaling events that control reentry into the cell cycle.
Collapse
Affiliation(s)
- Peter M Burch
- Environmental Pathology Program and Vermont Cancer Center, University of Vermont College of Medicine, Burlington, VT 05405, USA
| | | |
Collapse
|
45
|
Guo Y, Harwalkar J, Stacey DW, Hitomi M. Destabilization of cyclin D1 message plays a critical role in cell cycle exit upon mitogen withdrawal. Oncogene 2005; 24:1032-42. [PMID: 15592507 DOI: 10.1038/sj.onc.1208299] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Cyclin D1 is critical for entry into, continuation of, and exit from the cell division cycle. Mitogen stimulation of quiescent cells induces cyclin D1 expression in a transcription-dependent manner. In actively cycling cells, on the other hand, fluctuation of cyclin D1 protein levels through the cell cycle is post-transcriptionally regulated. Cyclin D1 is expressed at low levels during S phase to allow efficient DNA synthesis, and induced to high levels in G2 phase through Ras activity to commit the cells to continuing cell cycle progression. Once induced in G2 phase, cyclin D1 expression becomes Ras independent through the next G1 phase, where it promotes G1/S transition. When mitogenic signaling is abrogated, however, cyclin D1 fails to increase during G2 phase and the cell becomes arrested in the next G1 phase. In this way, the expression levels of cyclin D1 in G2 phase determine the fate of the next cell cycle. Despite its importance of the mechanism of cyclin D1 suppression upon mitogen withdrawal is unknown. Using both quantitative fluorescence microscopy and biochemical analyses, we have found that, upon serum deprivation, cyclin D1 mRNA is downmodulated without any decline in its rate of transcription. Furthermore, cyclin D1 mRNA half-life becomes shorter when serum is removed. These results demonstrate that cyclin D1 message destabilization plays a critical role in cyclin D1 suppression during G2 phase of serum-deprived cultures, and therefore in the withdrawal from the cell cycle.
Collapse
Affiliation(s)
- Yang Guo
- The Department of Molecular Biology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | | | | | |
Collapse
|
46
|
De Falco M, Fedele V, De Luca L, Penta R, Cottone G, Cavallotti I, Laforgia V, De Luca A. Evaluation of cyclin D1 expression and its subcellular distribution in mouse tissues. J Anat 2005; 205:405-12. [PMID: 15575889 PMCID: PMC1571359 DOI: 10.1111/j.0021-8782.2004.00347.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Cyclin D1 is a key cell-cycle regulatory protein required for the cell to progress through G1 to S phase. We have shown by Western blot analysis that cyclin D1 has a wide distribution in adult mouse tissues, with its level of expression being tissue-dependent. Immunohistochemistry has also shown that cyclin D1 may be present in the cytoplasm, in the nucleus or in both these cell compartments: cytoplasmic staining was observed in both proliferating cells (e.g. kidney, intestine, stomach and salivary gland) and in the non-dividing cells (the mature neurons of adult brain), while nuclear staining was seen in the neurons of the embryonic nervous system. Immunoelectron microscopy results indicate that, in tissues where cyclin D1 is present in both compartments (e.g. intestinal enterocytes), it may move via nuclear pores from the nucleus to the cytoplasm, and vice versa. The findings as a whole suggest that cyclin D1 may play multiple roles within specific tissues, probably by interacting with different substrates, and that its transit between nuclear and cytoplasmic compartments may help maintain cell homeostasis.
Collapse
Affiliation(s)
- Maria De Falco
- Department of Evolutive and Comparative Biology, University of Naples ‘Federico II’Naples, Italy
| | - Valentina Fedele
- Department of Evolutive and Comparative Biology, University of Naples ‘Federico II’Naples, Italy
| | - Luca De Luca
- Department of Medicine and Public Health, Section of Clinical Anatomy, Second University of NaplesNaples, Italy
| | - Roberta Penta
- Department of Medicine and Public Health, Section of Clinical Anatomy, Second University of NaplesNaples, Italy
| | - Giuliano Cottone
- Laboratory ‘C’, Center for Experimental Research, Regina Elena InstituteRome, Italy
| | - Ivan Cavallotti
- Department of Medicine and Public Health, Section of Clinical Anatomy, Second University of NaplesNaples, Italy
| | - Vincenza Laforgia
- Department of Evolutive and Comparative Biology, University of Naples ‘Federico II’Naples, Italy
| | - Antonio De Luca
- Department of Medicine and Public Health, Section of Clinical Anatomy, Second University of NaplesNaples, Italy
- Laboratory ‘C’, Center for Experimental Research, Regina Elena InstituteRome, Italy
| |
Collapse
|
47
|
Sa G, Stacey DW. P27 expression is regulated by separate signaling pathways, downstream of Ras, in each cell cycle phase. Exp Cell Res 2004; 300:427-39. [PMID: 15475007 DOI: 10.1016/j.yexcr.2004.07.032] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2004] [Revised: 07/30/2004] [Indexed: 11/20/2022]
Abstract
The cyclin inhibitory protein p27Kip1 (p27) plays a vital role in regulating cell proliferation in response to the extracellular growth environment. Active proliferation requires the suppression of p27 levels throughout the cell cycle. Late in the cell cycle, p27 degradation requires phosphorylation of Thr 187 by cyclin dependent kinase 2, leading to recognition by the SCF ubiquitin ligase containing the Skp2 F-box protein. Suppression of p27 is also essential for cell proliferation early in the cell cycle, but this occurs independently of Skp2, whose expression is suppressed during G1 phase. In this study, we use a time lapse and quantitative imaging approach to study the connection between proliferative signaling and the degradation of p27 during each cell cycle period in actively cycling cells. Ras activity was required for the suppression of p27 levels throughout the cell cycle, but separate pathways downstream of Ras signaling were required in different cell cycle periods. For example, inhibitors of MEK and phosphatidylinositol-3-kinase induced p27 expression primarily in G1 phase, while inhibitors of AKT activity stimulated these levels primarily in S phase. Skp2 was expressed in a Ras-dependent manner at higher levels late in the cell cycle. Its ablation resulted in higher p27 levels primarily in G2 phase as expected. The fact that separate signaling pathways downstream of Ras function in each cell cycle phase to suppress p27 levels helps explain the vital connection between proliferative signaling, cell cycle control, and p27 expression.
Collapse
Affiliation(s)
- Gaurisankar Sa
- Department of Molecular Biology, The Lerner Research Institute, The Cleveland Clinic Foundation, Cleveland, OH 44195, USA
| | | |
Collapse
|
48
|
Stierlé V, Laigle A, Jollès B. The Reduction of P-Glycoprotein Expression by Small Interfering RNAs Is Improved in Exponentially Growing Cells. Oligonucleotides 2004; 14:191-8. [PMID: 15625914 DOI: 10.1089/oli.2004.14.191] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Small interfering RNAs (siRNAs) are powerful tools in specifically silencing gene expression. Nevertheless, their efficiency can be limited when targeting proteins with an unusually long half-life, such as P-glycoprotein (P-gp), which is involved in the multidrug resistance phenomenon. P-gp is characterized by a long half-life, which may vary depending on the cell line and, for some of them, on serum deprivation or high cell density. In the present paper, involvement of an exponential cell growth phase in the improvement of siRNA efficiency has been suggested. The doxorubicin-selected human line MCF7-R was shown to be a more adapted model than NIH-MDR-G185 cells stably transfected with human mdr1. Nonspecific effects occurring at moderate (100 nM) siRNA concentration have been shown. Two efficient siRNAs led to a very satisfactory P-gp extinction (only 20% P-gp expression remaining) with siRNA concentration as low as 20 nM.
Collapse
Affiliation(s)
- Vérène Stierlé
- Laboratoire de Physicochimie Biomoléculaire et Cellulaire, CNRS (UMR 7033) and Université P. et M. Curie, 75005 Paris, France
| | | | | |
Collapse
|
49
|
Coleman ML, Marshall CJ, Olson MF. RAS and RHO GTPases in G1-phase cell-cycle regulation. Nat Rev Mol Cell Biol 2004; 5:355-66. [PMID: 15122349 DOI: 10.1038/nrm1365] [Citation(s) in RCA: 268] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Mathew L Coleman
- Abramson Family Cancer Research Institute, BRB II/III, 421 Curie Boulevard, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6160, USA
| | | | | |
Collapse
|
50
|
Shojaei F, Gallacher L, Bhatia M. Differential gene expression of human stem progenitor cells derived from early stages of in utero human hematopoiesis. Blood 2004; 103:2530-40. [PMID: 14656878 DOI: 10.1182/blood-2003-09-3209] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractHematopoietic stem progenitor cells (HSPCs) are highly enriched in a rare subset of Lin-CD34+CD38- cells. Independent of stage of human development, HSPC function segregates to the subset of Lin-CD34+CD38- cells. However, fetal-derived HSPCs demonstrate distinct self-renewal and differentiation capacities compared with their adult counterparts. Here, to characterize the molecular nature of fetal HSPCs, suppressive subtractive hybridization was used to compare gene expression of HSPCs isolated from fetal blood (FB-HSPCs) versus adult mobilized peripheral blood (MPB-HSPCs). We identified 97 differentially expressed genes that could be annotated into distinct groups that include transcription factors, cell cycle regulators, and genes involved in signal transduction. Candidate regulators, such as Lim only domain-2 (LMO2), nuclear factor–kappa B (NF-κB), tripartite motif 28 (Trim28), and N-myc protooncogene (MYCN), and a novel homeobox gene product were among transcripts that were found to be differentially expressed and could be associated with specific proliferation and differentiation properties unique to FB-HSPCs. Interestingly, the majority of genes associated with signal transduction belong to Ras pathway, highlighting the significance of Ras signaling in FB-HSPCs. Genes differentially expressed in FB-HSPCs versus adult MPB-HSPCs were verified using quantitative real-time polymerase chain reaction (Q-PCR). This approach also resulted in the identification of a transcript that is highly expressed in FB-HSPCs but not detectable in more differentiated Lin-CD34+CD38+ FB progenitors. Our investigation represents the first study to compare phenotypically similar, but functionally distinct, HSPC populations and to provide a gene profile of unique human HSPCs with higher proliferative capacity derived from early in utero human blood development.
Collapse
Affiliation(s)
- Farbod Shojaei
- Robarts Research Institute, Stem Cell Biology and Regenerative Medicine, London, ON, Canada
| | | | | |
Collapse
|