Induced cell cycle arrest

Last updated

Induced cell cycle arrest is the use of a chemical or genetic manipulation to artificially halt progression through the cell cycle. Cellular processes like genome duplication and cell division stop. [1] It can be temporary or permanent. [1] It is an artificial activation of naturally occurring cell cycle checkpoints, induced by exogenous stimuli controlled by an experimenter.

Contents

Model organisms

Some induced cell cycle arrests are done in Xenopus (frog) oocytes Xenopusoocyte.jpg
Some induced cell cycle arrests are done in Xenopus (frog) oocytes

In an academic research context, cell cycle arrest is typically performed in model organisms and cell extracts, such as Saccharomyces cervisiae (yeast) or Xenopus oocytes (frog eggs). [2] [3] Frog egg cell extracts have been used extensively in cell cycle research because they are relatively large, reaching a diameter of 1mm, and so contain large amounts of protein, making protein levels more easily measurable. [4]

Purposes

There are a variety of reasons a researcher may want to temporarily or permanently prevent progress through the cell cycle.

Cell cycle synchronization

In some experiments, a researcher may want to control and synchronize the time when a group of cells progress to the next phase of the cell cycle. [5] The cells can be induced to arrest as they arrive (at different time points) at a certain phase, so that when the arrest is lifted (for instance, rescuing cell cycle progression by introducing another chemical) all the cells resume cell cycle progression at the same time. In addition to this method acting as a scientific control for when the cells resume the cell cycle, this can be used to investigate necessity and sufficiency.

Another reason synchrony is important is the control for amount of DNA content, which varies at different parts of the cell cycle based on whether DNA replication has occurred since the last round of completed mitosis and cytokinesis. [6]

Furthermore, synchronization of large numbers of cells into the same phase allows for the collection of large enough groups of cells in the same cycle for the use in other assays, such as western blot and RNA sequencing. [7]

DNA damage repair

Researchers may be investigating mechanisms of DNA damage repair. Given that some of the mechanisms below of inducing cell cycle arrest involve damaging the DNA, this allows investigation into how the cell responds to damage of its genetic material. [8]

Identification of in vivo protein function

Genetic engineering of cells with specific gene knockouts can also result in cells that arrest at different phases of the cell cycle. Examples include:

G1 phase arrest

Phases of the cell cycle Cell Cycle 3-3.svg
Phases of the cell cycle

G1 phase is the first of the four phases of the cell cycle, and is part of interphase. While in G1 the cell synthesizes messenger RNA (mRNA) and proteins in preparation for subsequent steps of interphase leading to mitosis. In human somatic cells, the cell cycle lasts about 18 hours, and the G1 phase makes up about 1/3 of that time. [13] On the other hand, in frog, sea urchin, and fruit fly embryos, the G1 phase is extremely brief and instead is a slight gap between cytokinesis and S phase. [13]

Alpha Factor

α-factor is a pheromone secreted by Saccharomyces cervisiae that arrests the yeast cells in G1 phase. It does so by inhibiting the enzyme adenylate cyclase. [2] The enzyme catalyzes the conversion of adenosine triphosphate (ATP) to 3',5'-cyclic AMP (cAMP) and pyrophosphate. [14]

Contact inhibition

Contact inhibition is a method of arresting cells when neighboring cells come into contact with each other. It results in a single layer of arrested cells of arrested cells, and is a process that is notably missing in cancer cells. The suspected mechanism is dependent on p27Kip1, a cyclin-dependent kinase inhibitor. [15] p27Kip1 protein levels are elevated in arresting cells. This natural process can be mimicked in a lab through the overexpression of p27Kip1, which results in induced cell cycle arrest in G1 phase. [16]

Mimosine

Mimosine is a plant amino acid that has been shown to reversibly inhibit progression beyond G1 phase in some human cells, including lymphoblastoid cells. [5] Its proposed mechanism of action is an iron/zinc chelator that depletes iron within the cell. This induces double-strand breaks in the DNA, inhibiting DNA replication. This may involve blocking the action of an iron-dependent ribonucleotide reductase. It may also inhibit transcription of serine hydroxymethyltransferase, which has zinc dependence. [17]

Serum deprivation

In cell culture, serum is the growth medium in which the cells are grown and contains vital nutrients. The use of serum deprivation - partially or completely removing the serum and its nutrients - has been shown to arrest and synchronize cell cycle progression in G0 phase, for example in neonatal mammalian astrocytes [18] and human foreskin fibroblasts. [19]

Amino acid starvation is a similar approach. When grown in a media without some essential amino acids, such as methionine, some cells arrest in early G1 phase. [5]

S phase arrest

S phase follows G1 phase via the G1/S transition and precedes G2 phase in interphase and is the part of the cell cycle in which DNA is replicated. Since accurate duplication of the genome is critical to successful cell division, the processes that occur during S-phase are tightly regulated and widely conserved. Pre-replication complexes assembled before S phase are converted into active replication forks. [20] Driving this conversion is Cdc7 and S-phase cyclin-dependent kinases, which are both upregulated after the G1/S transition. [20]

Aphidicolin

Aphidicolin is an antibiotic isolated from the fungus Cephalosporum aphidicola. It is a reversible inhibitor of eukaryotic nuclear DNA replication that blocks progression past the S phase. Its mechanism is the inhibition of DNA polymerase A and D. A structural study found that this is thought to occur through binding the alpha active site of the polymerase and "rotating the template guanine," which prevents deoxycytidine triphosphate (dCTP) from binding. [21] This S phase block induces apoptosis in HeLa cells. [5]

Hydroxyurea

Hydroxyurea (HU) is a small molecule drug that inhibits the enzyme ribonucleotide reductase (RNR), preventing the catalysis of converting deoxyribonucleotides (DNTs) to ribonucleotides. It is hypothesized that there is tyrosyl free radical within RNR that is disabled by HU. [6] [22] The free radicals are necessary for the reduction of the DNTs and are scavenged by HU instead. [23] HU has been shown to arrest cells in both S phase (healthy cells) and immediately before cytokinesis (mutant cells). [22]

2,3-DCPE

2[[3-(2,3-dichlorophenoxy)propyl] amino]ethanol (2,3-DCPE) is a small-molecule that induces S phase arrest. [24] This was demonstrated in cancer cell lines and downregulates expression of B-cell lymphoma-extra large (Bcl-XL), an anti-apoptotic protein that prevents the release of mitochondrial contents like cytochrome c.

G2 phase arrest

G2 phase is the final part of interphase and directly precedes mitosis. It will only be entered in regular cells if the DNA replication in S phase is completed successfully. It is a period of rapid cell growth and protein synthesis during which the cell prepares itself for mitosis.

Destruction of cyclin mRNA

Cyclins are proteins that control progression through the cell cycle by activating cyclin-dependent kinases. Destruction of a cell's endogenous cyclin messenger RNA can arrest frog egg extracts in interphase and prevent them from entering mitosis. [3] Introduction of exogenous cyclin mRNA is also sufficient to rescue cell cycle progression. [3] One method of this destruction is through the use of antisense oligonucleotides, pieces of RNA that bind to the cyclin mRNA and prevent the mRNA from being translated into cyclin protein. [25] This can actually be used to destroy phase-specific cyclins beyond just G2 - for instance, destruction of cyclin D1 mRNA by antisense oligonucleotides prevents progression from G1 phase to S phase. [26]

Mitotic arrest

Mitosis is the non-interphase part of the cell cycle and generates two daughter cells Mitosis Stages.svg
Mitosis is the non-interphase part of the cell cycle and generates two daughter cells

Mitosis is the final part of the cell cycle and follows interphase. It is composed of four phases - prophase, metaphase, anaphase, and telophase - and involves the condensation of the chromosomes in the nucleus, the dissolution of the nuclear envelope, and the separation of sister chromatids by spindle fibers. As mitosis concludes, the spindle fibers disappear and the nuclear membrane reforms around each of the two sets of chromosomes. After successful mitosis, the cell physically splits into two identical daughter cells in a process called cytokinesis, and this concludes a full round of the cell cycle. Each of these new cells could then potentially re-enter G1 phase and begin the cell cycle again. [27]

Nocodazole

Nocodazole is a chemical agent that interferes with the polymerization of microtubules. [28] Cells treated with nocodazole arrest with a G2 or M phase DNA content, which can be verified with flow cytometry. From microscopy it has been determined they do enter mitosis but they cannot form the spindles necessary for metaphase because the microtubules cannot polymerize. [29] Research into the mechanism has hinted at it potentially preventing tubulin from forming its alpha/beta heterodimer. [30]

Taxol

Taxol works in the opposite way of nocodazole, instead stabilizing the microtubule polymer and preventing it from disassembly. It also causes M phase arrest, as the spindle that is supposed to pull apart sister chromatids is unable to disassemble. [31] [32] It acts through a specific binding site on the microtubule polymer, and as such does not require GTP or other cofactors to induce tubulin polymerization. [33]

Temperature

Temperature has been shown to regulate HeLa cell cycle progression. Mitosis was found to be the most temperature-sensitive part of the cell cycle. [34] Pre-cytokinesis mitotic arrest was visible through accumulation of cells in mitosis in below-normal temperatures between 24 and 31 °C (75.2-87.8 °F). [34]

Verification

There are several methods that can be used to verify that cells have been arrested in the proper phase.

Flow cytometry

Flow cytometry is a technique of measuring physical and chemical characteristics of a population of cells using lasers and fluorophore dyes covalently linked to protein markers. [35] The stronger the signal, the more of a particular protein is present. Staining with DNA dyes propidium iodide or 4',6'-diamidino-2-phenylindole (DAPI) allows delineation or sorting of cells between G1, S, or G2/M phases. [36]

Immunoblotting

Immunoblotting is the detection of specific proteins in a tissue sample or extract. Primary antibodies recognize and bind the protein in question, and secondary antibodies are added that recognize the primary antibodies. The secondary antibody is then visualized through staining or immunofluorescence, allowing indirect detection of the original target protein.

Immunoblotting can be performed to detect the presence of cyclins, proteins that regulate the cell cycle. [37] Different classes of cyclins are up- and down-regulated at different parts of the cell cycle. Measurement of the cyclins from an extract of an arrested cell can determine what phase the cell is in. For example, a peak of cyclin E protein would indicate the G1/S transition, a cyclin A peak would indicate late G2 phase, and a cyclin B peak would indicate mitosis. [38]

Fluorescence ubiquitination-based cell cycle indication (FUCCI)

FUCCI is a system that takes advantage of cell cycle phase-specific expression of proteins and their degradation by the ubiquitin-proteasome pathway. Two fluorescent probes - Cdt1 and Geminin conjugated to fluorescent proteins - allow for real-time visualization of the cell cycle phase a cell is in. [39]

Related Research Articles

<span class="mw-page-title-main">Cell cycle</span> Series of events and stages that result in cell division

The cell cycle, or cell-division cycle, is the series of events that take place in a cell that causes it to divide into two daughter cells. These events include the duplication of its DNA and some of its organelles, and subsequently the partitioning of its cytoplasm, chromosomes and other components into two daughter cells in a process called cell division.

<span class="mw-page-title-main">Cell division</span> Process by which living cells divide

Cell division is the process by which a parent cell divides into two daughter cells. Cell division usually occurs as part of a larger cell cycle in which the cell grows and replicates its chromosome(s) before dividing. In eukaryotes, there are two distinct types of cell division: a vegetative division (mitosis), producing daughter cells genetically identical to the parent cell, and a cell division that produces haploid gametes for sexual reproduction (meiosis), reducing the number of chromosomes from two of each type in the diploid parent cell to one of each type in the daughter cells. Mitosis is a part of the cell cycle, in which, replicated chromosomes are separated into two new nuclei. Cell division gives rise to genetically identical cells in which the total number of chromosomes is maintained. In general, mitosis is preceded by the S stage of interphase and is followed by telophase and cytokinesis; which divides the cytoplasm, organelles, and cell membrane of one cell into two new cells containing roughly equal shares of these cellular components. The different stages of mitosis all together define the M phase of an animal cell cycle—the division of the mother cell into two genetically identical daughter cells. To ensure proper progression through the cell cycle, DNA damage is detected and repaired at various checkpoints throughout the cycle. These checkpoints can halt progression through the cell cycle by inhibiting certain cyclin-CDK complexes. Meiosis undergoes two divisions resulting in four haploid daughter cells. Homologous chromosomes are separated in the first division of meiosis, such that each daughter cell has one copy of each chromosome. These chromosomes have already been replicated and have two sister chromatids which are then separated during the second division of meiosis. Both of these cell division cycles are used in the process of sexual reproduction at some point in their life cycle. Both are believed to be present in the last eukaryotic common ancestor.

<span class="mw-page-title-main">Interphase</span> G1, S and G2 phases of the cell cycle

Interphase is the portion of the cell cycle that is not accompanied by visible changes under the microscope, and includes the G1, S and G2 phases. During interphase, the cell grows (G1), replicates its DNA (S) and prepares for mitosis (G2). A cell in interphase is not simply quiescent. The term quiescent would be misleading since a cell in interphase is very busy synthesizing proteins, copying DNA into RNA, engulfing extracellular material, processing signals, to name just a few activities. The cell is quiescent only in the sense of cell division. Interphase is the phase of the cell cycle in which a typical cell spends most of its life. Interphase is the 'daily living' or metabolic phase of the cell, in which the cell obtains nutrients and metabolizes them, grows, replicates its DNA in preparation for mitosis, and conducts other "normal" cell functions.

G<sub>1</sub> phase First growth phase in the eukaryotic cell cycle

The G1 phase, gap 1 phase, or growth 1 phase, is the first of four phases of the cell cycle that takes place in eukaryotic cell division. In this part of interphase, the cell synthesizes mRNA and proteins in preparation for subsequent steps leading to mitosis. G1 phase ends when the cell moves into the S phase of interphase. Around 30 to 40 percent of cell cycle time is spent in the G1 phase.

<span class="mw-page-title-main">Anaphase-promoting complex</span> Cell-cycle regulatory complex

Anaphase-promoting complex is an E3 ubiquitin ligase that marks target cell cycle proteins for degradation by the 26S proteasome. The APC/C is a large complex of 11–13 subunit proteins, including a cullin (Apc2) and RING (Apc11) subunit much like SCF. Other parts of the APC/C have unknown functions but are highly conserved.

<span class="mw-page-title-main">Cell growth</span> Increase in the total cell mass

Cell growth refers to an increase in the total mass of a cell, including both cytoplasmic, nuclear and organelle volume. Cell growth occurs when the overall rate of cellular biosynthesis is greater than the overall rate of cellular degradation.

<span class="mw-page-title-main">Cyclin</span> Group of proteins

Cyclin is a family of proteins that controls the progression of a cell through the cell cycle by activating cyclin-dependent kinase (CDK) enzymes or group of enzymes required for synthesis of cell cycle.

<span class="mw-page-title-main">S phase</span> DNA replication phase of the cell cycle, between G1 and G2 phase

S phase (Synthesis phase) is the phase of the cell cycle in which DNA is replicated, occurring between G1 phase and G2 phase. Since accurate duplication of the genome is critical to successful cell division, the processes that occur during S-phase are tightly regulated and widely conserved.

G<sub>2</sub> phase Second growth phase in the eukaryotic cell cycle, prior to mitosis

G2 phase, Gap 2 phase, or Growth 2 phase, is the third subphase of interphase in the cell cycle directly preceding mitosis. It follows the successful completion of S phase, during which the cell’s DNA is replicated. G2 phase ends with the onset of prophase, the first phase of mitosis in which the cell’s chromatin condenses into chromosomes.

<span class="mw-page-title-main">Cell cycle checkpoint</span> Control mechanism in the eukaryotic cell cycle

Cell cycle checkpoints are control mechanisms in the eukaryotic cell cycle which ensure its proper progression. Each checkpoint serves as a potential termination point along the cell cycle, during which the conditions of the cell are assessed, with progression through the various phases of the cell cycle occurring only when favorable conditions are met. There are many checkpoints in the cell cycle, but the three major ones are: the G1 checkpoint, also known as the Start or restriction checkpoint or Major Checkpoint; the G2/M checkpoint; and the metaphase-to-anaphase transition, also known as the spindle checkpoint. Progression through these checkpoints is largely determined by the activation of cyclin-dependent kinases by regulatory protein subunits called cyclins, different forms of which are produced at each stage of the cell cycle to control the specific events that occur therein.

<span class="mw-page-title-main">G1/S transition</span> Stage in cell cycle

The G1/S transition is a stage in the cell cycle at the boundary between the G1 phase, in which the cell grows, and the S phase, during which DNA is replicated. It is governed by cell cycle checkpoints to ensure cell cycle integrity and the subsequent S phase can pause in response to improperly or partially replicated DNA. During this transition the cell makes decisions to become quiescent, differentiate, make DNA repairs, or proliferate based on environmental cues and molecular signaling inputs. The G1/S transition occurs late in G1 and the absence or improper application of this highly regulated checkpoint can lead to cellular transformation and disease states such as cancer.

Cyclin A is a member of the cyclin family, a group of proteins that function in regulating progression through the cell cycle. The stages that a cell passes through that culminate in its division and replication are collectively known as the cell cycle Since the successful division and replication of a cell is essential for its survival, the cell cycle is tightly regulated by several components to ensure the efficient and error-free progression through the cell cycle. One such regulatory component is cyclin A which plays a role in the regulation of two different cell cycle stages.

<span class="mw-page-title-main">Cyclin-dependent kinase inhibitor protein</span> Protein which inhibits cyclin-dependent kinase

A cyclin-dependent kinase inhibitor protein(also known as CKIs, CDIs, or CDKIs) is a protein which inhibits the enzyme cyclin-dependent kinase (CDK) and Cyclin activity by stopping the cell cycle if there are unfavorable conditions, therefore, acting as tumor suppressors. Cell cycle progression is stopped by Cyclin-dependent kinase inhibitor protein at the G1 phase. CKIs are vital proteins within the control system that point out whether the process of DNA synthesis, mitosis, and cytokines control one another. If a malfunction prevents the successful completion of DNA synthesis during the G1 phase, a signal is sent to delay or stop the progression to the S phase. Cyclin-dependent kinase inhibitor proteins are essential in the regulation of the cell cycle. If cell mutations surpass the cell cycle checkpoints during cell cycle regulation, it can result in various types of cancer.

Cell synchronization is a process by which cells in a culture at different stages of the cell cycle are brought to the same phase. Cell synchrony is a vital process in the study of cells progressing through the cell cycle as it allows population-wide data to be collected rather than relying solely on single-cell experiments. The types of synchronization are broadly categorized into two groups; physical fractionization and chemical blockade.

<span class="mw-page-title-main">Cyclin-dependent kinase 1</span> Mammalian protein found in Homo sapiens

Cyclin-dependent kinase 1 also known as CDK1 or cell division cycle protein 2 homolog is a highly conserved protein that functions as a serine/threonine protein kinase, and is a key player in cell cycle regulation. It has been highly studied in the budding yeast S. cerevisiae, and the fission yeast S. pombe, where it is encoded by genes cdc28 and cdc2, respectively. With its cyclin partners, Cdk1 forms complexes that phosphorylate a variety of target substrates ; phosphorylation of these proteins leads to cell cycle progression.

<span class="mw-page-title-main">Wee1</span> Nuclear protein

Wee1 is a nuclear kinase belonging to the Ser/Thr family of protein kinases in the fission yeast Schizosaccharomyces pombe. Wee1 has a molecular mass of 96 kDa and is a key regulator of cell cycle progression. It influences cell size by inhibiting the entry into mitosis, through inhibiting Cdk1. Wee1 has homologues in many other organisms, including mammals.

A series of biochemical switches control transitions between and within the various phases of the cell cycle. The cell cycle is a series of complex, ordered, sequential events that control how a single cell divides into two cells, and involves several different phases. The phases include the G1 and G2 phases, DNA replication or S phase, and the actual process of cell division, mitosis or M phase. During the M phase, the chromosomes separate and cytokinesis occurs.

Cdc14 and Cdc14 are a gene and its protein product respectively. Cdc14 is found in most of the eukaryotes. Cdc14 was defined by Hartwell in his famous screen for loci that control the cell cycle of Saccharomyces cerevisiae. Cdc14 was later shown to encode a protein phosphatase. Cdc14 is dual-specificity, which means it has serine/threonine and tyrosine-directed activity. A preference for serines next to proline is reported. Many early studies, especially in the budding yeast Saccharomyces cerevisiae, demonstrated that the protein plays a key role in regulating late mitotic processes. However, more recent work in a range of systems suggests that its cellular function is more complex.

<span class="mw-page-title-main">Control of chromosome duplication</span>

In cell biology, eukaryotes possess a regulatory system that ensures that DNA replication occurs only once per cell cycle.

<span class="mw-page-title-main">G2-M DNA damage checkpoint</span>

The G2-M DNA damage checkpoint is an important cell cycle checkpoint in eukaryotic organisms that ensures that cells don't initiate mitosis until damaged or incompletely replicated DNA is sufficiently repaired. Cells with a defective G2-M checkpoint will undergo apoptosis or death after cell division if they enter the M phase before repairing their DNA. The defining biochemical feature of this checkpoint is the activation of M-phase cyclin-CDK complexes, which phosphorylate proteins that promote spindle assembly and bring the cell to metaphase.

References

  1. 1 2 Li Y, Fan J, Ju D (Jan 1, 2019). "15 - Neurotoxicity concern about the brain targeting delivery systems". In Gao H, Gao X (eds.). Brain Targeted Drug Delivery System. Academic Press. pp. 377–408. doi:10.1016/B978-0-12-814001-7.00015-9. ISBN   978-0-12-814001-7. S2CID   105749019.
  2. 1 2 Liao H, Thorner J (April 1980). "Yeast mating pheromone alpha factor inhibits adenylate cyclase". Proceedings of the National Academy of Sciences of the United States of America. 77 (4): 1898–902. Bibcode:1980PNAS...77.1898L. doi: 10.1073/pnas.77.4.1898 . PMC   348616 . PMID   6246513.
  3. 1 2 3 Murray AW, Kirschner MW (May 1989). "Cyclin synthesis drives the early embryonic cell cycle". Nature. 339 (6222): 275–80. Bibcode:1989Natur.339..275M. doi:10.1038/339275a0. PMID   2566917. S2CID   4352582.
  4. Lodish H, Berk A, Zipursky SL, et al. (2000). "Section 13.2: Biochemical Studies with Oocytes, Eggs, and Early Embryos". Molecular Cell Biology (4th ed.). New York: W. H. Freeman. ISBN   0-7167-3136-3.
  5. 1 2 3 4 Krek W, DeCaprio JA (1995). "[7] Cell synchronization". Cell synchronization. Methods in Enzymology. Vol. 254. Elsevier. pp.  114–24. doi:10.1016/0076-6879(95)54009-1. ISBN   978-0-12-182155-5. PMID   8531680.
  6. 1 2 Koç A, Wheeler LJ, Mathews CK, Merrill GF (January 2004). "Hydroxyurea arrests DNA replication by a mechanism that preserves basal dNTP pools". The Journal of Biological Chemistry. 279 (1): 223–30. doi: 10.1074/jbc.M303952200 . PMID   14573610.
  7. Purcell M, Kruger A, Tainsky MA (2014-12-15). "Gene expression profiling of replicative and induced senescence". Cell Cycle. 13 (24): 3927–37. doi:10.4161/15384101.2014.973327. PMC   4615143 . PMID   25483067.
  8. Singh A, Xu YJ (November 2016). "The Cell Killing Mechanisms of Hydroxyurea". Genes. 7 (11): 99. doi: 10.3390/genes7110099 . PMC   5126785 . PMID   27869662.
  9. Mendenhall MD, Richardson HE, Reed SI (June 1988). "Dominant negative protein kinase mutations that confer a G1 arrest phenotype". Proceedings of the National Academy of Sciences of the United States of America. 85 (12): 4426–30. Bibcode:1988PNAS...85.4426M. doi: 10.1073/pnas.85.12.4426 . PMC   280442 . PMID   3288995.
  10. Uchiyama M, Galli I, Griffiths DJ, Wang TS (June 1997). "A novel mutant allele of Schizosaccharomyces pombe rad26 defective in monitoring S-phase progression to prevent premature mitosis". Molecular and Cellular Biology. 17 (6): 3103–15. doi:10.1128/MCB.17.6.3103. PMC   232163 . PMID   9154809.
  11. al-Khodairy F, Carr AM (April 1992). "DNA repair mutants defining G2 checkpoint pathways in Schizosaccharomyces pombe". The EMBO Journal. 11 (4): 1343–50. doi:10.1002/j.1460-2075.1992.tb05179.x. PMC   556583 . PMID   1563350.
  12. Hwang LH, Murray AW (October 1997). "A novel yeast screen for mitotic arrest mutants identifies DOC1, a new gene involved in cyclin proteolysis". Molecular Biology of the Cell. 8 (10): 1877–87. doi:10.1091/mbc.8.10.1877. PMC   25633 . PMID   9348530.
  13. 1 2 Morgan DO (2007). The cell cycle : principles of control. New Science Press. ISBN   978-0-19-920610-0. OCLC   70173205.
  14. Zhang G, Liu Y, Ruoho AE, Hurley JH (March 1997). "Structure of the adenylyl cyclase catalytic core". Nature. 386 (6622): 247–53. Bibcode:1997Natur.386..247Z. doi:10.1038/386247a0. PMID   9069282. S2CID   4329051.
  15. Seluanov A, Hine C, Azpurua J, Feigenson M, Bozzella M, Mao Z, et al. (November 2009). "Hypersensitivity to contact inhibition provides a clue to cancer resistance of naked mole-rat". Proceedings of the National Academy of Sciences of the United States of America. 106 (46): 19352–7. Bibcode:2009PNAS..10619352S. doi: 10.1073/pnas.0905252106 . PMC   2780760 . PMID   19858485.
  16. Li J, Yang XK, Yu XX, Ge ML, Wang WL, Zhang J, Hou YD (August 2000). "Overexpression of p27(KIP1) induced cell cycle arrest in G(1) phase and subsequent apoptosis in HCC-9204 cell line". World Journal of Gastroenterology. 6 (4): 513–521. doi: 10.3748/wjg.v6.i4.513 (inactive 31 January 2024). PMC   4723549 . PMID   11819639.{{cite journal}}: CS1 maint: DOI inactive as of January 2024 (link)
  17. "Mimosine". www.drugbank.ca. Retrieved 2019-12-13.
  18. Langan TJ, Rodgers KR, Chou RC (2016-11-05). "Synchronization of Mammalian Cell Cultures by Serum Deprivation". Cell Cycle Synchronization. Methods in Molecular Biology. Vol. 1524. Springer New York. pp. 97–105. doi:10.1007/978-1-4939-6603-5_6. ISBN   978-1-4939-6602-8. PMID   27815898.
  19. Narasimha AM, Kaulich M, Shapiro GS, Choi YJ, Sicinski P, Dowdy SF (June 2014). "Cyclin D activates the Rb tumor suppressor by mono-phosphorylation". eLife. 3: e02872. doi: 10.7554/eLife.02872 . PMC   4076869 . PMID   24876129.
  20. 1 2 Takeda DY, Dutta A (April 2005). "DNA replication and progression through S phase". Oncogene. 24 (17): 2827–43. doi: 10.1038/sj.onc.1208616 . PMID   15838518.
  21. Baranovskiy AG, Babayeva ND, Suwa Y, Gu J, Pavlov YI, Tahirov TH (December 2014). "Structural basis for inhibition of DNA replication by aphidicolin". Nucleic Acids Research. 42 (22): 14013–21. doi:10.1093/nar/gku1209. PMC   4267640 . PMID   25429975.
  22. 1 2 Xu YJ, Singh A, Alter GM (November 2016). "Hydroxyurea Induces Cytokinesis Arrest in Cells Expressing a Mutated Sterol-14α-Demethylase in the Ergosterol Biosynthesis Pathway". Genetics. 204 (3): 959–973. doi:10.1534/genetics.116.191536. PMC   5105871 . PMID   27585850.
  23. Platt OS (March 2008). "Hydroxyurea for the treatment of sickle cell anemia". The New England Journal of Medicine. 358 (13): 1362–9. doi:10.1056/NEJMct0708272. PMID   18367739.
  24. Zhu H, Zhang L, Wu S, Teraishi F, Davis JJ, Jacob D, Fang B (June 2004). "Induction of S-phase arrest and p21 overexpression by a small molecule 23-(2,3-dichlorophenoxy)propyl] amino]ethanol in correlation with activation of ERK". Oncogene. 23 (29): 4984–92. doi: 10.1038/sj.onc.1207645 . PMID   15122344.
  25. "NCI Dictionary of Cancer Terms". National Cancer Institute. 2011-02-02. Retrieved 2019-12-13.
  26. Saikawa Y, Kubota T, Otani Y, Kitajima M, Modlin IM (October 2001). "Cyclin D1 antisense oligonucleotide inhibits cell growth stimulated by epidermal growth factor and induces apoptosis of gastric cancer cells". Japanese Journal of Cancer Research. 92 (10): 1102–9. doi:10.1111/j.1349-7006.2001.tb01065.x. PMC   5926617 . PMID   11676861.
  27. "Mitosis - an overview | ScienceDirect Topics". www.sciencedirect.com. Retrieved 2019-12-12.
  28. Kuhn M (March 1998). "The microtubule depolymerizing drugs nocodazole and colchicine inhibit the uptake of Listeria monocytogenes by P388D1 macrophages". FEMS Microbiology Letters. 160 (1): 87–90. doi: 10.1111/j.1574-6968.1998.tb12895.x . PMID   9495017.
  29. Kanthou C, Tozer GM (June 2009). "Microtubule depolymerizing vascular disrupting agents: novel therapeutic agents for oncology and other pathologies". International Journal of Experimental Pathology. 90 (3): 284–94. doi:10.1111/j.1365-2613.2009.00651.x. PMC   2697551 . PMID   19563611.
  30. Xu K, Schwarz PM, Ludueña RF (Feb 2002). "Interaction of nocodazole with tubulin isotypes". Drug Development Research. 55 (2): 91–96. doi:10.1002/ddr.10023. ISSN   0272-4391. S2CID   86235729.
  31. Choi YH, Yoo YH (December 2012). "Taxol-induced growth arrest and apoptosis is associated with the upregulation of the Cdk inhibitor, p21WAF1/CIP1, in human breast cancer cells". Oncology Reports. 28 (6): 2163–9. doi: 10.3892/or.2012.2060 . PMID   23023313.
  32. Ikui AE, Yang CP, Matsumoto T, Horwitz SB (October 2005). "Low concentrations of taxol cause mitotic delay followed by premature dissociation of p55CDC from Mad2 and BubR1 and abrogation of the spindle checkpoint, leading to aneuploidy". Cell Cycle. 4 (10): 1385–8. doi: 10.4161/cc.4.10.2061 . PMID   16138009.
  33. Horwitz SB (1994). "Taxol (paclitaxel): mechanisms of action". Annals of Oncology. 5 (Suppl 6): S3-6. PMID   7865431.
  34. 1 2 Rao PN, Engelberg J (May 1965). "He La Cells: Effects of Temperature on the Life Cycle". Science. 148 (3673): 1092–4. Bibcode:1965Sci...148.1092R. doi:10.1126/science.148.3673.1092. PMID   14289609. S2CID   27085343.
  35. Picot J, Guerin CL, Le Van Kim C, Boulanger CM (March 2012). "Flow cytometry: retrospective, fundamentals and recent instrumentation". Cytotechnology. 64 (2): 109–30. doi:10.1007/s10616-011-9415-0. PMC   3279584 . PMID   22271369.
  36. Pozarowski P, Darzynkiewicz Z (2004-07-01). "Analysis of cell cycle by flow cytometry". Checkpoint Controls and Cancer. Methods in Molecular Biology. Vol. 281. Humana Press. pp. 301–11. doi:10.1385/1-59259-811-0:301. ISBN   978-1-59259-811-3. PMID   15220539.
  37. Jenkins CW, Xiong Y (1996). "Immunoprecipitation and Immunoblotting in Cell Cycle Studies". Cell Cycle — Materials and Methods. Springer Berlin Heidelberg. pp. 250–263. doi:10.1007/978-3-642-57783-3_22. ISBN   978-3-540-58066-9.
  38. "Cyclin - an overview | ScienceDirect Topics". www.sciencedirect.com. Retrieved 2019-12-12.
  39. "Fluorescent Ubiquitination-based Cell Cycle Indicator (FUCCI)". MBL International. Retrieved 2019-12-12.