Fact-checked by Grok 2 weeks ago

Cell cycle

The cell cycle is the ordered series of events in which a eukaryotic cell grows, duplicates its DNA, and divides to produce two genetically identical daughter cells, ensuring accurate transmission of genetic information across generations. This process is fundamental to organismal growth, tissue repair, and reproduction in multicellular organisms, while in unicellular species, it represents the primary mechanism of population increase. The cell cycle consists of two major phases: interphase and the mitotic (M) phase. Interphase, which occupies the majority of the cycle, includes three subphases—G1 (gap 1), S (synthesis), and G2 (gap 2)—during which the cell increases in size, replicates its DNA, and prepares for division. In the G1 phase, the cell assesses environmental conditions and synthesizes proteins and organelles; the S phase involves precise duplication of the genome to produce sister chromatids; and the G2 phase checks for DNA replication errors before proceeding. The M phase encompasses mitosis, where chromosomes segregate, and cytokinesis, which physically divides the cytoplasm. Some cells may enter a quiescent G0 phase, temporarily exiting the cycle in response to signals like nutrient limitation. Progression through the cell cycle is tightly regulated by cyclin-dependent kinases (CDKs), which are activated by binding to regulatory proteins called cyclins whose levels oscillate periodically. These cyclin-CDK complexes phosphorylate target proteins to drive phase transitions, such as promoting in or chromosome condensation in . Checkpoints act as surveillance mechanisms at key points—G1/S, G2/M, and —to detect DNA damage, replication errors, or spindle assembly defects, halting the cycle for repairs or triggering if irreparable. Dysregulation of these controls, often through mutations in cyclins, CDKs, or checkpoint genes, can lead to uncontrolled proliferation and is a hallmark of cancer. The cell cycle's fidelity is crucial for maintaining genomic stability, enabling embryonic development, and responding to injury, with evolutionary conservation across eukaryotes underscoring its biological significance. In humans, disruptions contribute to diseases beyond cancer, including developmental disorders and aging-related pathologies.

Overview

Definition and significance

The cell cycle is a highly regulated sequence of events in eukaryotic cells that coordinates growth, , and to produce two genetically identical daughter cells. This process ensures the accurate duplication and equitable distribution of genetic material, primarily during —comprising the G1, , and phases for preparation—and the mitotic (M) phase for . In mammalian cells, the typical duration of the cell cycle under optimal conditions is approximately 24 hours, though this varies significantly by cell type and physiological context. The significance of the cell cycle extends to fundamental biological processes, including organismal , where coordinated builds complex and organs from a single fertilized . It also supports maintenance and repair by replacing damaged or senescent cells, such as in skin renewal or , thereby preserving in multicellular organisms. Dysregulation of the cell cycle, often through genetic , can lead to uncontrolled as seen in cancer or premature contributing to degenerative diseases. Cell cycle length exhibits notable variations across cell types; for instance, early embryonic cells in mammals divide rapidly with cycles of approximately 12 hours to facilitate rapid tissue formation, in contrast to the longer 24-hour cycles typical of somatic cells in adults. These dynamics are often visualized in a cyclical diagram depicting the phases in a circular or linear progression, with occupying the majority of the timeline and the M phase as a brief culminating event, highlighting the preparatory nature of growth relative to division.

Historical background

The study of the cell cycle began with early observations of cell division in the late . In 1882, German anatomist published detailed descriptions of , identifying the process through chromatin staining in larval cells, which revealed the longitudinal splitting of chromosomes during division. This work, detailed in his book Zellsubstanz, Kern und Zelltheilung, marked a foundational shift from vague notions of cell reproduction to precise cytological documentation, emphasizing the continuity of threads across generations of cells. A major advance occurred in the early 1950s with the introduction of radioactive labeling techniques to track cellular processes biochemically. In 1953, Alma Howard and Stephen Pelc coined the term "cell cycle" while studying root meristem cells of Vicia faba using phosphorus-32 to label DNA synthesis. Their experiments demonstrated that DNA replication occurs during a discrete period distinct from mitosis, leading to the identification of interphase subphases: G1 (pre-DNA synthesis gap), S (synthesis), and G2 (post-synthesis gap). This framework revealed the cell cycle's ordered progression, with DNA duplication confined to approximately 6-8 hours in their model system, separate from the roughly 4-hour mitotic phase. The 1950s also saw the adoption of tritiated thymidine as a more specific tracer for DNA replication, enabling finer resolution of the S phase. Developed shortly after World War II, this isotope allowed researchers to pulse-label dividing cells and track their progression via autoradiography, confirming the timing of DNA synthesis and distinguishing cycling from non-cycling populations. By the 1960s, these methods had solidified the G1, S, and G2 phases as standard, while the recognition of a quiescent G0 phase emerged for non-dividing cells, such as neurons and hepatocytes, where cells exit the cycle reversibly or permanently. Lajtha's 1963 studies on hematopoietic cells proposed G0 as a resting state outside the proliferative loop, highlighting variability in cycle commitment. Post-World War II advancements in radioisotope availability facilitated this transition from descriptive cytology to quantitative biochemistry, transforming the cell cycle from a morphological curiosity into a measurable kinetic process. These milestones laid the groundwork for understanding in and , though molecular details awaited later decades.

Phases

G0 phase

The , often referred to as the quiescent or resting phase, is a distinct state outside the active cell cycle where cells temporarily or permanently halt . In this non-dividing condition, cells maintain viability but do not prepare for or division, distinguishing it from the preparatory growth occurring in G1. Quiescence in G0 can be reversible, allowing cells like lymphocytes to re-enter the cycle upon immune stimulation, or irreversible in post-mitotic cells such as neurons and cardiomyocytes, which remain in this state for the organism's lifetime. Entry into G0 typically occurs from the in response to environmental or developmental signals, including nutrient deprivation, high cell density leading to contact inhibition, or cues promoting terminal differentiation. The duration of G0 varies widely: it may last only hours in response to transient stress or become permanent in differentiated tissues. For instance, under nutrient limitation, cells sense depleted or glucose levels, rapidly arresting to conserve resources while enacting protective against damage. During G0, cells undergo notable physiological adaptations, including reduced metabolic activity, compaction of into a more condensed heterochromatic state, and diminished synthesis to minimize energy expenditure. These changes enable long-term survival in adverse conditions, with transcription limited to genes essential for maintenance rather than growth. A prominent example is following partial , where the majority of hepatocytes reside in G0 under normal conditions but synchronously re-enter the cycle from this quiescent state to restore tissue mass within days. In contrast to G1 phase cells, which actively assess conditions for progression toward DNA synthesis, G0 cells exhibit downregulated expression of proliferation-associated factors and require specific external stimuli, such as growth factors, to transition back into the cycle. Without these signals, G0 cells remain arrested and do not advance to the S phase, underscoring the phase's role in preventing inappropriate division.

G1 phase

The G1 phase, the first gap phase following mitosis, is a period of cellular growth and preparation for DNA replication in the cell cycle. In mammalian cells, it typically lasts 8-12 hours, though this duration varies by cell type and environmental conditions, representing a significant portion of the overall 24-hour cycle in proliferating cells. During this phase, cells increase in size through active protein synthesis and , which support the metabolic demands of impending division. duplication, including mitochondria and other structures, also occurs to ensure equitable distribution to daughter cells. A critical event in G1 is the , located in mid-to-late G1 approximately 2-3 hours before entry into , where cells make an irreversible to complete the division process. This , first described by Arthur Pardee in 1974, renders cells independent of external mitogenic signals thereafter, marking a transition from reversible to unidirectional progression. In yeast, an analogous point called START serves a similar function, committing cells to division upon nutrient availability. Progression through G1 involves sensing environmental cues, particularly growth factors such as (PDGF) and (EGF), which activate signaling pathways to promote cell cycle advancement. These factors are essential prior to the , enabling cells to assess proliferative potential; their absence halts progression in early G1. As G1 nears its end, cells evaluate their readiness for via checkpoints that monitor DNA damage and nutrient status, preventing faulty replication if issues are detected. This assessment ensures genomic integrity and metabolic sufficiency before the transition to .

S phase

The S phase, or synthesis phase, of the cell cycle is the period during which the cell's DNA is replicated to ensure each daughter cell receives an identical copy of the genome. In mammalian cells, this phase typically lasts 6-8 hours, comprising a significant portion of the 24-hour cell cycle in rapidly dividing cells. During S phase, DNA replication proceeds via a semi-conservative mechanism, where each parental DNA strand serves as a template for synthesizing a complementary daughter strand, effectively doubling the DNA content from the diploid 2N level to 4N. This process is tightly regulated to occur only once per cell cycle, preventing over-replication that could lead to genomic instability. DNA replication initiates at multiple origins of replication distributed throughout the , with human cells utilizing approximately 10,000 to 50,000 such sites to complete the duplication of their 6 billion base pairs within the allotted time. At each origin, the (ORC) binds to specific DNA sequences, recruiting additional proteins to unwind and form bidirectional replication forks that progress in opposite directions. These forks advance at speeds of 50-100 base pairs per second, driven by a complex that includes MCM to unwind the DNA, δ and ε for synthesizing the leading and lagging strands, and (part of α) to generate short primers for initiating on the lagging strand. As the forks proceed, topoisomerases relieve torsional stress ahead of the replication machinery, while single-strand binding proteins maintain the stability of unwound DNA. To enforce the once-per-cycle rule, replication origins are "licensed" during G1 phase by the loading of inactive MCM helicase complexes onto DNA by ORC, Cdc6, and Cdt1 proteins; high cyclin-dependent kinase activity in S phase then activates these complexes while preventing re-licensing until the next G1. This licensing system ensures initiation occurs only at authorized sites and only once. Concurrently, S phase coordinates the synthesis of histones, the core proteins of chromatin, which must double to package the newly replicated DNA; histone genes are transcriptionally activated at the G1/S transition, with mRNA stability enhanced to match replication timing, thereby maintaining nucleosome density and epigenetic marks. Replication fidelity is exceptionally high, with an overall error rate of about 1 mismatch per 10^9 bases incorporated, achieved through multiple safeguards. DNA polymerases exhibit base selectivity to favor correct nucleotide insertion, followed by 3'→5' exonuclease proofreading activity that removes misincorporated bases with 99-99.9% efficiency. Any residual errors are corrected post-replication by the mismatch repair system, which scans for and excises mismatched base pairs using proteins like MSH2-MSH6 and MLH1-PMS2. Checkpoints briefly monitor replication progress and DNA integrity during S phase to halt progression if damage or stalled forks are detected.

G2 phase

The G2 phase follows the completion of in and serves as a preparatory interval before entry into , allowing the to grow and finalize internal structures essential for division. In mammalian cells, this phase typically lasts 3–4 hours, during which the cell volume approximately doubles through continued of proteins and , ensuring adequate cytoplasmic resources for the daughter cells. duplication, initiated during , completes in G2, yielding two fully mature centrosomes that will organize the mitotic spindle. Key events in G2 include the synthesis of mitotic proteins such as tubulins, which are critical components of , and the onset of microtubule reorganization to form the precursors of the . The is maintained intact throughout G2, preserving the integrity of the replicated while the cell tests for stress responses, such as oxidative or metabolic perturbations, to confirm readiness for division. Additionally, organelles like mitochondria and the Golgi apparatus undergo final maturation and positioning. A primary function of G2 involves the genome integrity check, where any residual DNA damage from S phase—such as double-strand breaks or replication errors—is repaired through pathways like or , preventing propagation of mutations into . In stressed cells, such as those exposed to or UV light, G2 can be prolonged for hours or even days to facilitate repair, exemplified by arrests in and mammalian models under genotoxic conditions. The transition from to mitosis is orchestrated by the accumulation of mitotic cyclins, particularly , which binds to and activates (CDK1), driving phosphorylation events that initiate breakdown and condensation. This cyclin buildup, first identified in embryos, ensures timely progression only after preparatory events are complete.

Mitotic phase

The mitotic phase, also known as M phase, is the stage of the cell cycle in which the replicated chromosomes are precisely segregated to produce two daughter nuclei, each receiving an identical set of 2N chromosomes. This process ensures genetic stability across cell generations and typically lasts 30-60 minutes in mammalian cells, representing a brief but critical portion of the overall cell cycle. The phase is divided into five sequential stages—prophase, , , , and —characterized by dynamic changes in chromosome structure, nuclear architecture, and cytoskeletal organization. Central to mitosis is the mitotic spindle, a bipolar array of that forms from microtubule-organizing centers and facilitates chromosome movement through interactions with motor proteins such as kinesins and dyneins. In , the first stage, chromosomes begin to condense from their extended form into compact, rod-like structures visible under light microscopy, facilitated by condensins and modifications. The mitotic spindle starts to assemble, with emanating from centrosomes that migrate to opposite poles of the cell, establishing the spindle poles. proteins assemble on the centromeres of each sister chromatid pair, preparing attachment sites for spindle , while the remains intact. Prometaphase follows, marked by the breakdown of the nuclear envelope, which allows direct access of spindle microtubules to chromosomes. Microtubules from the spindle poles probe the cytoplasm and capture kinetochores, leading to initial attachments that are often unstable and corrected through trial-and-error dynamics. Kinesin and dynein motor proteins contribute to these attachments by generating forces that align chromosomes and resolve improper connections. During , chromosomes achieve bipolar attachment to the , with sister kinetochores connected to from opposite poles. This results in the alignment of chromosomes at the metaphase plate, an equatorial plane midway between the spindle poles, ensuring balanced distribution. The configuration is stabilized by tension across the kinetochores, powered by polymerization/ and activity. Anaphase initiates the separation of , triggered by the anaphase-promoting complex (APC/C), an E3 ubiquitin ligase that targets securin for degradation, thereby activating separase to cleave proteins holding chromatids together. Chromatids are then pulled toward opposite poles: in A, kinetochores lead the movement via shortening, while in anaphase B, poles separate further due to elongation and motor-driven sliding. Kinesins and dyneins play key roles in these poleward forces and spindle elongation. Telophase concludes with the arrival of chromatids at the spindle poles, where chromosomes decondense back to chromatin fibers. The reforms around each set of chromosomes, nucleoli reappear, and the mitotic spindle disassembles as depolymerize. This stage restores the nuclear architecture of , preparing the daughter nuclei for subsequent cell cycle progression. Mitosis exhibits variations across eukaryotes; in animals, it is typically "open," involving complete breakdown to enable broad spindle-chromosome interactions, whereas in yeasts like , it is "closed," with the envelope remaining intact and spindle formation occurring within the nucleus. These differences reflect evolutionary adaptations to cell size and organization.

Cytokinesis

is the final stage of in which the and its contents are partitioned between two daughter cells, ensuring each receives a complete set of organelles and cellular components. This process overlaps with the late stages of , commencing during when the chromosomes have segregated and concluding during as the nuclear envelopes reform. In most eukaryotic cells, lasts from several minutes to about an hour, depending on the cell type and organism, with typical durations of 10-30 minutes observed in mammalian cells such as . In animal cells, proceeds through the formation of a cleavage furrow at the cell's , driven by the of a cortical actomyosin ring composed of filaments and II motors. This ring assembles under the control of spatiotemporal RhoA signaling, where the RhoA is activated at the equatorial cortex by guanine nucleotide exchange factors like ECT2, promoting polymerization via formins and activation through ROCK kinase. The contractile ring constricts progressively, pinching the plasma membrane inward in an "outside-in" manner until the daughter cells separate. In plant cells, lacking a rigid during division, cytokinesis occurs via the formation of a that expands centrifugally to partition the and build a new . This process is mediated by the , a microtubule-based structure that forms during late anaphase-telophase and guides the delivery of Golgi-derived vesicles to the division site. Vesicles fuse at the phragmoplast midzone to form the , which matures into a as it reaches the parental plasma membrane, typically completing in tens of minutes to an hour in higher . During , cellular organelles such as mitochondria and the () are partitioned between the daughter cells to maintain metabolic and synthetic functions. Mitochondria, which replicate independently, are distributed somewhat randomly but often actively recruited to the division site via or interactions, ensuring equitable inheritance. The ER network, continuous across the cell, fragments and reallocates during and reforms in each daughter cell post-cytokinesis. In animal cells, following actomyosin ring contraction, a transient midbody structure forms from compacted microtubules and plasma membrane at the intercellular bridge, serving as an organizing center for the final abscission step. Abscission involves the endosomal sorting complex required for transport (ESCRT) machinery, which severs the bridge by membrane remodeling and severing, completing the physical separation of daughter cells. This culminates the division process, allowing the daughter cells to enter G1 phase of the next cell cycle. Failure of can result in binucleate cells, where the remains undivided despite nuclear separation, leading to or tetraploidy. Such cells may undergo subsequent mitotic errors, promoting genomic instability and , which are development. In experimental models, cytokinesis failure has been linked to centrosome amplification and tumorigenesis, underscoring its role in maintaining genomic integrity.

Molecular Mechanisms of Regulation

Cyclins and cyclin-dependent kinases

Cyclins are a family of regulatory proteins that act as oscillating subunits essential for controlling the progression through the eukaryotic . These proteins, including types such as A, B, D, and E, are synthesized and degraded in a periodic manner, with their levels fluctuating to ensure timely activation of downstream processes. The degradation of cyclins occurs primarily through the ubiquitin-proteasome pathway, which targets them for destruction at specific points, thereby resetting the cycle and preventing untimely re-entry into phases. Cyclin-dependent kinases (CDKs) are serine/ protein , numbered CDK1 through CDK9 in mammals, that remain enzymatically inactive in the absence of bound . of CDKs requires to their cognate partner, which induces a conformational change, followed by on a conserved residue in the T-loop by CDK-activating (CAK), such as the CDK7- H . This dual mechanism— and CAK-mediated —fully activates the CDK holoenzyme, enabling it to phosphorylate substrate proteins. The cyclin-CDK complexes play a central role in driving cell cycle phase transitions by phosphorylating key targets, such as the () to promote G1/S progression and nuclear lamins to facilitate . This regulatory system is highly conserved across eukaryotes, originating from the Cdc28 kinase in budding yeast, which shares functional homology with human CDK1. Cyclin expression is tightly phase-specific; for instance, levels peak during the in response to mitogenic signals, initiating early events in cell cycle commitment.

Cyclin-CDK complex specificity and activity

The specificity of -CDK complexes arises from the combinatorial pairing of distinct isoforms with particular cyclin-dependent kinases (CDKs), which directs to phase-specific and ensures ordered cell cycle progression. Cyclins not only activate CDKs but also confer substrate selectivity through structural motifs that facilitate and of targets. This targeted activity enables precise control over transitions between cell cycle phases, preventing premature or erroneous events. In the G1 phase, associates with CDK4 or CDK6 to form complexes that initiate progression toward by phosphorylating the (). This phosphorylation disrupts Rb's inhibitory binding to transcription factors, thereby releasing E2F to activate genes required for , such as cyclin E and components. The sequential hyperphosphorylation of Rb by cyclin D-CDK4/6 sets a for commitment to the cell cycle, ensuring that cells respond appropriately to growth signals. During S phase, E- and A-bound CDK2 complexes drive by phosphorylating key components of the replication machinery. E-CDK2 promotes the of replication at origins by targeting proteins like CDC6, which facilitates loading, while also preventing re-replication through inhibition of origin licensing factors such as and MCM. A-CDK2 further supports replication fork progression and elongation by activating δ and other polymerases, ensuring complete and accurate duplication. These activities maintain replication fidelity and block redundant firing of origins. In the G2/M transition, partners with CDK1 to form the (MPF), which orchestrates mitotic entry through phosphorylation of numerous substrates. MPF phosphorylates nuclear lamins, leading to their disassembly and breakdown, which is essential for chromosome condensation and spindle formation. Additionally, -CDK1 activity prepares substrates for degradation by the anaphase-promoting complex/cyclosome (APC/C), including securin, whose ubiquitination triggers sister chromatid separation during . The activity of cyclin-CDK complexes is dynamically regulated by activation thresholds and feedback loops that generate switch-like transitions. Low initial CDK activity is amplified through , where active cyclin B-CDK1 phosphorylates and activates phosphatases while inhibiting Wee1 kinases, which otherwise phosphorylate and inactivate CDK1. This double-negative loop establishes a bistable switch, ensuring rapid and irreversible commitment to once thresholds are surpassed. Cyclin levels are tightly controlled, with periodic synthesis and ubiquitin-mediated degradation by the /C providing temporal precision; for instance, APC/C-mediated destruction of inactivates CDK1 at mitotic exit, resetting the cycle. Mathematical models of -CDK oscillations illustrate how these feedback mechanisms produce robust, switch-like phase transitions. In computational simulations, interlinked bistable switches involving synthesis, activation loops, and /C degradation generate autonomous oscillations that mimic observed cell cycle periodicity, with abrupt rises in CDK activity driving irreversible commitments. Such models highlight the role of ultrasensitive in insulating , allowing modular control despite interconnected regulatory pathways.

Cell cycle inhibitors

Cell cycle inhibitors, also known as cyclin-dependent kinase inhibitors (CKIs), are critical regulatory proteins that impose brakes on cell cycle progression by binding to and inactivating (CDK) complexes, thereby preventing of key substrates and halting transitions such as G1 to . These inhibitors ensure fidelity in by responding to stress signals, and they are classified into two main families: the INK4 family, which specifically targets CDK4 and CDK6 to block their association with , and the Cip/Kip family, which broadly inhibits multiple cyclin-CDK complexes. The INK4 proteins, including p15^INK4B, ^INK4A, p18^INK4C, and p19^INK4D, act by competitively binding to monomeric CDK4 or CDK6, thereby inhibiting binding and subsequent activation. Among the Cip/Kip family members, p21^Cip1 (also known as CDKN1A) is transcriptionally induced by the tumor suppressor p53 in response to DNA damage, leading to G1 arrest by binding to cyclin E-CDK2 and cyclin A-CDK2 complexes to block access to substrates like the retinoblastoma protein (pRb). Similarly, p27^Kip1 (CDKN1B) promotes G1 arrest through dual mechanisms: direct inhibition of cyclin E-CDK2 activity by binding to the CDK subunit and sequestration of cyclin E away from CDK2, which is particularly evident in response to transforming growth factor-β (TGF-β) signaling or contact inhibition. These CKIs often function redundantly; for instance, both p21 and p27 contribute to cell cycle arrest following genotoxic stress, with p21 playing a more prominent role in p53-dependent pathways. In the context of cellular senescence, an irreversible form of cell cycle arrest, accumulates progressively with replicative stress or oncogenic signaling, enforcing G1 arrest by inhibiting CDK4/6 and maintaining pRb in a hypophosphorylated state that represses E2F-dependent transcription. This accumulation is a hallmark of (SASP) and helps prevent propagation of damaged cells during aging or processes. CKIs also facilitate in lineages such as erythroid cells, where upregulation coincides with terminal cell cycle exit. Synthetic cell cycle inhibitors, primarily small-molecule CDK antagonists, have been developed to pharmacologically mimic endogenous CKIs for therapeutic purposes, particularly in hyperproliferative diseases like cancer. Flavopiridol, a first-generation pan-CDK , targets multiple CDKs including CDK1, CDK2, CDK4, CDK6, and CDK9 by competing with ATP binding, inducing G1 and G2 but with limited clinical success due to . More selective second- and third-generation inhibitors focus on CDK4/6, such as (approved in 2015), which binds the ATP site of CDK4/6 to prevent association, thereby blocking pRb and S-phase entry in hormone receptor-positive cancers. (approved in 2017) offers enhanced selectivity and brain penetrance, inhibiting CDK4/6 as well as CDK9 to suppress proliferation and transcription of oncogenic drivers. These synthetic agents integrate with cellular checkpoints by amplifying DNA damage responses, similar to endogenous CKIs, to enforce in aberrant cells.

Transcriptional and post-transcriptional controls

plays a pivotal role in coordinating cell cycle progression by activating specific gene sets at distinct phases. The family of transcription factors, particularly E2F1-3, drives the expression of genes required for S-phase entry, including those involved in and nucleotide synthesis, ensuring timely progression from G1 to . In the G2/M transition, FoxM1 acts as a key activator, inducing transcription of genes essential for , such as those encoding proteins and cyclins, thereby promoting chromosome segregation and mitotic fidelity. In simpler eukaryotes like , oscillatory transcriptional networks underpin cell cycle dynamics, with approximately 800 genes exhibiting periodic expression synchronized to the cell cycle, forming a core regulatory circuit that maintains temporal order across phases. Post-transcriptional mechanisms further refine cell cycle timing through RNA and protein modifications. MicroRNAs, such as the miR-15/16 cluster, downregulate cyclins like cyclin D1 by targeting their mRNAs for degradation or translational repression, thereby inhibiting G1/S progression and preventing uncontrolled proliferation. Ubiquitination by E3 ligases, including the SCF complex and APC/C, provides precise control over protein degradation; SCF targets G1/S regulators for timely removal in early phases, while APC/C ensures mitotic exit by degrading securin and cyclins during anaphase, thus linking degradation kinetics to phase transitions. Feedback loops integrate these controls for robust regulation. Positive feedback amplifies CDK1 activity through auto-phosphorylation and nuclear translocation of cyclin B1-CDK1 complexes, rapidly committing cells to once initiated. Negative feedback, exemplified by the -Mdm2 loop, limits expression—a induced by —to resolve DNA damage responses and allow cell cycle re-entry, balancing growth arrest with recovery. Spatial compartmentalization adds another layer, with nuclear accumulation of CDK1-cyclin B1 preceding cytoplasmic activation, ensuring onset propagates from to for coordinated cytoskeletal remodeling. Recent advances in single-cell RNA sequencing (scRNA-seq) have illuminated cell cycle heterogeneity, revealing asynchronous waves across individual cells within populations, which underlie variable progression rates and contribute to developmental robustness or pathological states like cancer. These studies, post-2010, highlight how transcriptional bursts in cell cycle genes amplify differences in timing, offering insights beyond analyses. Additionally, as of 2025, research has identified evolutionarily recent transcription factors, such as Krüppel-associated box zinc-finger proteins (KZFPs) including ZNF519 and ZNF274, that add lineage-specific regulation to the conserved cell cycle machinery by targeting rhythmic genes and influencing progression in and mammals.

Checkpoints

G1/S checkpoint

The G1/S checkpoint functions as the primary regulatory barrier that evaluates cellular growth status, nutrient availability, and DNA integrity prior to commitment to DNA replication during S phase. This checkpoint integrates the restriction point, a commitment mechanism originally identified in mammalian fibroblasts where cells become independent of external mitogenic signals to complete the cell cycle. If conditions are unfavorable—such as insufficient cell mass or the presence of DNA lesions—the checkpoint halts progression, preventing the initiation of replication on damaged templates that could lead to genomic instability. In normal cells, passage through this checkpoint requires hyperphosphorylation of the retinoblastoma protein (Rb) by cyclin-dependent kinase 2 (CDK2) complexes, releasing E2F transcription factors to drive S-phase gene expression. DNA damage sensors, primarily the ATM and ATR kinases, play a central role in activating the checkpoint upon detection of lesions like double-strand breaks or replication stress. responds to double-strand breaks by phosphorylating and stabilizing , a tumor suppressor , while ATR handles single-strand DNA exposures. Activated then transcriptionally induces p21 (CDKN1A), a potent that binds and suppresses E-CDK2 activity, maintaining Rb in its hypophosphorylated state and blocking E2F-dependent transcription. This p53-p21 axis ensures that damaged cells do not proceed to , allowing time for repair pathways such as to act. Upon checkpoint activation, outcomes include transient G1 arrest for , prolonged , or via if irreparable damage persists. For instance, ultraviolet (UV) radiation induces G1/S arrest through p53-dependent p21 upregulation, which not only inhibits CDK2 but also protects cells from subsequent apoptosis by preventing replication of UV-induced . This response is conserved across mammalian cells and underscores the checkpoint's role in safeguarding stability. The G1/S checkpoint exhibits notable variations across developmental stages and organismal complexity. In early embryos of vertebrates, such as or mice, the checkpoint is absent or rudimentary, enabling rapid divisions with minimal G1 phases to facilitate swift during embryogenesis. This feature evolves during development, with full checkpoint establishment occurring in somatic s of multicellular organisms to enforce tighter control over in response to tissue demands and . Such developmental highlights the checkpoint's adaptability in balancing with fidelity in complex eukaryotes.

Intra-S phase checkpoint

The intra-S phase checkpoint is a surveillance mechanism activated during DNA replication to detect and respond to replication stress, such as single-strand DNA breaks or stalled replication forks, thereby maintaining genomic stability. This checkpoint operates within S phase to modulate replication dynamics, preventing further progression until damage is addressed. Activation of the intra-S phase checkpoint primarily involves the ATR (ataxia-telangiectasia and Rad3-related) , which senses regions of single-stranded DNA generated at stalled replication forks through recruitment by the RPA ()-coated ssDNA and the ATRIP (ATR-interacting protein) complex. Upon , ATR phosphorylates and activates the effector Chk1 (checkpoint kinase 1), which propagates the signal to downstream targets. This ATR-Chk1 pathway inhibits the firing of new replication origins by suppressing the activity of pre-replication complexes, ensuring that replication resources are directed toward resolving existing stress rather than initiating new forks. Key mechanisms include the Chk1-mediated phosphorylation of Cdc25A phosphatase, leading to its ubiquitination and proteasomal degradation via the SCF^β-TRCP E3 ligase complex, which reduces Cdc25A levels and sustains inhibitory phosphorylation on CDK2 (cyclin-dependent kinase 2). Downregulation of CDK2 activity in turn prevents the activation of late-firing replication origins and stabilizes stalled forks by limiting excessive helicase unwinding that could cause fork collapse. These processes collectively protect replication forks from collapse into double-strand breaks, preserving replication fork integrity during stress. The consequences of intra-S phase checkpoint activation include a controlled slowing of S phase progression, which provides time for DNA repair pathways, such as translesion synthesis or fork restart, to resolve lesions without complete replication arrest. For instance, chemotherapeutic agents like hydroxyurea or induce replication stress by depleting dNTP pools or trapping topoisomerase-DNA complexes, respectively, triggering the checkpoint to delay S phase completion and enhance cell survival under therapeutic pressure. Defects in this checkpoint, such as ATR or Chk1 inhibition, exacerbate collapse and genomic in response to such drugs. Recent studies have highlighted the checkpoint's role in preventing under-replication at common fragile sites (CFSs), late-replicating genomic regions prone to instability under mild replication stress. The ATR-Chk1 pathway stabilizes forks at CFSs by promoting dormant origin firing and limiting ultrafine bridge formation, as evidenced in post-2000 analyses of aphidicolin-treated cells where ATR deficiency led to increased CFS expression and incomplete replication. These insights underscore the checkpoint's function in averting chromosomal aberrations at fragile loci during physiological or pathological replication challenges.

G2/M checkpoint

The G2/M checkpoint serves as a critical surveillance mechanism at the end of the , preventing cells from entering until is complete and any damage is repaired, thereby maintaining genomic stability. This checkpoint integrates signals from DNA damage response pathways to halt cell cycle progression, primarily by inhibiting the activation of the B-CDK1 complex, which is essential for mitotic entry. Triggers for the G2/M checkpoint activation include DNA double-strand breaks (DSBs), which are primarily sensed by the ataxia-telangiectasia mutated (ATM) kinase; ATM phosphorylates and activates the checkpoint kinase Chk2 (CHEK2). Unrepaired DNA lesions, such as those from replication stress, engage the ataxia-telangiectasia and Rad3-related (ATR) kinase and Chk1 (CHEK1), providing an overlapping pathway. Persistent or severe damage also activates the tumor suppressor p53, which transcriptionally induces p21 (CDKN1A), a potent inhibitor of CDK1 activity, thereby enforcing prolonged arrest. Key effectors of the checkpoint converge on direct regulation of . Phosphorylation of on tyrosine 15 (Tyr15) by () inhibits its activity, preventing premature mitotic initiation; this is enhanced by Chk1 and Chk2 signaling. Conversely, the family of phosphatases (, , ), which normally dephosphorylate to activate it, are inactivated through multisite by Chk1 and Chk2, leading to their binding by 14-3-3 proteins and sequestration in the . This dual inhibition—via activation and sequestration—ensures robust suppression of until damage is resolved. Resolution of the checkpoint involves DNA repair pathways active in G2, particularly homologous recombination (HR), which utilizes the sister chromatid as a template to accurately mend DSBs; ATM and Chk2 promote HR by facilitating end resection and recruitment of repair factors like BRCA1. Upon successful repair, phosphatase activity resumes, dephosphorylating Cdc25 to release it from 14-3-3 binding and allowing Wee1 degradation, thereby reactivating CDK1 and permitting mitotic entry. If the checkpoint is bypassed with unrepaired damage, cells proceed to aberrant mitosis, resulting in mitotic catastrophe characterized by chromosome fragmentation and cell death. The stringency of the G2/M checkpoint varies by cell type, being highly enforced in cells to prevent propagation of mutations, as evidenced by efficient arrest and repair in response to . In contrast, it is relaxed in germ cells and early embryos, such as oocytes and one-cell embryos, where attenuated responses to DNA damage allow rapid divisions essential for reproduction, though this increases mutagenesis risk in aged gametes.

Mitotic spindle assembly checkpoint

The mitotic spindle assembly checkpoint (SAC), also known as the , serves as the final surveillance mechanism in to ensure that all chromosomes achieve proper bipolar attachment to the mitotic spindle before the onset of . This checkpoint monitors kinetochore-microtubule interactions, preventing premature activation of the anaphase-promoting complex/cyclosome (/C), which would otherwise lead to the ubiquitination and degradation of securin and , thereby triggering sister chromatid separation and mitotic exit.01189-X) Unattached or improperly attached kinetochores generate a diffusible "wait-anaphase" signal that inhibits /C activity, allowing time for error correction and stable attachments to form. The core mechanism involves unattached kinetochores recruiting and activating SAC proteins, which catalyze the formation of the mitotic checkpoint (MCC). Key components include Mad1, Mad2, Bub1, Bub3, BubR1 (also known as Mad3 in ), and the Mps1, which localize to unattached kinetochores via initial recognition by the KMN (KNL1/Mis12/Ndc80 ). Mad2 undergoes a conformational change at kinetochores to bind Cdc20 (the APC/C co-activator), forming an open-Mad2:Cdc20 intermediate that promotes MCC assembly; BubR1 then joins this complex along with Bub3, creating a potent inhibitor that sequesters Cdc20 and directly blocks APC/C substrate binding. This MCC diffuses from kinetochores to inhibit cytoplasmic APC/C, halting progression. Upon bi-orientation, where sister kinetochores attach to microtubules from opposite spindle poles and experience tension, the SAC signal is silenced through stripping of checkpoint proteins from kinetochores by microtubule motors like dynein and PP1-mediated dephosphorylation, leading to MCC disassembly and APC/C activation. If misalignments persist, the SAC typically delays anaphase onset for 20-30 minutes in mammalian cells, providing a window for attachment stabilization; however, prolonged activation can lead to checkpoint slippage, where cells exit mitosis without division. Failure of the SAC results in chromosome missegregation and aneuploidy, a hallmark of genomic instability. The is highly conserved across most eukaryotes, reflecting its essential role in safeguarding integrity during chromosome segregation, with core components like Mad2 and BubR1 present from to humans. However, it is absent or rudimentary in some simple eukaryotes, such as trypanosomes, which lack a SAC despite having kinetochores, possibly due to their unique spindle architecture and closed .

Experimental and Imaging Techniques

Fluorescence-based methods

Fluorescence-based methods enable the and tracking of cell cycle progression in living cells by incorporating genetically encoded fluorescent proteins or dyes that label key regulatory components or structural elements. These techniques allow researchers to observe dynamic changes in , such as the accumulation and degradation of cyclins or the condensation of , without disrupting cellular processes. By fusing fluorescent tags like (GFP) or to cell cycle proteins, scientists can monitor phase transitions spatially and temporally, providing insights into the orchestration of division events. A prominent class of markers involves (FRET)-based sensors that detect (CDK) activity, which drives cell cycle transitions. These sensors typically consist of a CDK domain flanked by donor and acceptor fluorophores; by CDK alters the conformation, changing FRET efficiency and thus ratios to report activity levels. For instance, FRET biosensors have been used to quantify CDK1 activation thresholds at the G2/M transition in fission yeast, revealing a sharp rise in activity that commits cells to . Translocation-based CDK sensors, where shifts the protein's localization (e.g., import), offer single-color alternatives for live imaging with reduced . GFP-tagged cyclins serve as direct reporters for phase-specific expression and localization. Cyclin B1-GFP, for example, accumulates in the during and translocates to the at , enabling tracking of mitotic entry; its subsequent degradation marks onset. This has been instrumental in visualizing dynamics in mammalian cells and embryos, confirming that nuclear import requires specific sites on the . Such tags preserve endogenous while allowing quantitative of protein levels across the cycle. Time-lapse combined with these markers facilitates continuous observation of cell cycle events. (PCNA), a sliding clamp for , forms distinct foci during replication sites, detectable via PCNA-GFP or , distinguishing from G1 or G2 where it remains diffuse. H2B-mCherry fusions label , revealing condensation in and decondensation in ; this has been used to track changes during cycle progression. These approaches often employ confocal or spinning-disk to minimize in multi-color setups. In tissues and multicellular models, captures real-time cell cycle progression amid complex environments. For example, in embryos, multi-photon or light-sheet with GFP-tagged cyclins and H2B-mCherry has mapped asynchronous divisions, highlighting spatial coordination of G2/M transitions across the early lineage. These methods reveal how extrinsic cues, like cell-cell contacts, influence timing without averaging over populations. Advances in super-resolution techniques, such as depletion (STED) microscopy since the 2010s, have enhanced resolution beyond the diffraction limit to visualize fine structures like mitotic dynamics. STED imaging of or markers in fixed or live cells has resolved attachments and error corrections at the spindle checkpoint, providing sub-50 nm details of segregation. These innovations update earlier widefield approaches by enabling precise quantification of length and orientation fluctuations during .

Flow cytometry and other quantitative approaches

Flow cytometry is a cornerstone quantitative technique for assessing cell cycle distribution in cell populations by measuring DNA content. Cells are typically fixed and stained with a DNA-intercalating dye such as propidium iodide (PI), which binds stoichiometrically to DNA and fluoresces upon excitation, allowing discrimination of cell cycle phases based on fluorescence intensity. In this approach, cells in G0/G1 phases exhibit a diploid DNA content (2N), those in S phase show intermediate values between 2N and 4N as DNA replication progresses, and G2/M phase cells display a tetraploid content (4N). This method, pioneered in the 1970s, enables rapid analysis of thousands of cells per sample to quantify the percentages in each phase via histogram deconvolution software. To refine S-phase analysis and distinguish active DNA synthesis from other phases, bromodeoxyuridine (BrdU) pulse labeling is combined with DNA content staining in bivariate . BrdU, a analog, is incorporated into newly synthesized DNA during ; following a brief exposure, cells are fixed, denatured to expose BrdU, and stained with anti-BrdU antibodies conjugated to a , alongside PI for total DNA. This reveals the proportion of cells entering over time, with BrdU-positive cells showing increased green fluorescence (or equivalent) correlated with intermediate DNA content, allowing estimation of S-phase duration and progression rates. The technique, developed in the early 1980s, has become standard for kinetic studies in asynchronous populations. Ki-67 staining provides an additional quantitative marker for identifying proliferating cells across the cell cycle. The Ki-67 antigen is expressed in nuclei of cells in G1, S, G2, and M phases but absent in quiescent G0 cells, making it useful for flow cytometry to estimate the growth fraction when combined with DNA staining. Cells are permeabilized and stained with anti-Ki-67 antibodies, yielding a binary readout: positive cells represent the actively cycling population (typically 20-50% in growing cultures), while negative cells indicate quiescence. First characterized in the 1980s, this marker complements DNA-based methods by excluding non-cycling cells without requiring pulse labeling. Mass cytometry (CyTOF) extends these approaches to high-dimensional analysis by using metal isotope-tagged antibodies and DNA intercalators, enabling simultaneous measurement of up to 50 parameters per cell without spectral overlap issues of fluorescence. For cell cycle assessment, iridium-based DNA intercalators quantify content akin to PI, while metal-tagged probes for cyclins, Ki-67, or phospho-histone H3 delineate phases in multiparametric panels. This allows dissection of cycle progression alongside functional states (e.g., signaling pathways) in heterogeneous samples like tumors, with data analyzed via algorithms for phase assignment. Introduced in the late 2000s, CyTOF has facilitated quantitative insights into cycle regulation in immune and cancer cells. Quantitative metrics from these methods include cell cycle fractions (%G1, %S, %G2/M), derived from flow histograms using software like ModFit or FlowJo, which model sub-G1 debris, aggregates, and phase peaks for accurate (e.g., S-phase fractions often 20-40% in exponentially growing mammalian cells). Synchronization techniques enhance precision: , a microtubule-depolymerizing agent, arrests cells at the G2/M transition by activating the spindle assembly checkpoint, accumulating >80% of cells in 4N; upon washout, synchronous progression is monitored over 8-12 hours via serial to calculate phase transit times (e.g., G1 duration ~6-8 hours in cells). These metrics establish baseline cycle dynamics and responses to perturbations. In the 2010s, single-cell RNA sequencing (scRNA-seq) integrated cell cycle scoring tools like , which assign phases by comparing expression of paired marker genes (e.g., high PCNA/TOP2A for ) against reference profiles via and correlation scoring. Cyclone classifies cells into G1, S, G2/M, or intermediate states with ~90% accuracy in benchmark datasets, enabling pseudotime ordering of cycle progression without physical synchronization. This computational approach, leveraging datasets from synchronized cells, quantifies cycle heterogeneity in tissues and links it to transcriptional states, as validated in studies.

Cell Cycle in Disease and Development

Role in tumorigenesis

Dysregulation of the cell cycle is a central driver of tumorigenesis, primarily through mutations or alterations in key regulators that promote uncontrolled proliferation and genomic instability. Overactive cyclins, such as , play a pivotal role by accelerating the ; amplification of the CCND1 gene encoding is observed in approximately 15-20% of breast cancers, contributing to estrogen-driven tumor growth and poor prognosis in hormone receptor-positive subtypes. Similarly, inactivation of the pathway, which normally restrains cell cycle progression via sequestration, occurs in nearly all human cancers through diverse mechanisms including direct RB1 mutations, viral oncoprotein binding, or upstream disruptions like hyperactivity. Loss of function, the most frequent genetic alteration in cancers affecting about 50% of tumors, impairs checkpoint integrity and DNA damage response, allowing cells with unrepaired genetic lesions to proliferate and accumulate mutations. These alterations manifest as , including evasion of that leads to uncontrolled G1/S progression, heightened genomic instability from bypassed mechanisms, and resultant that fuels tumor heterogeneity and evolution. For instance, inactivation not only permits survival of cells with damaged DNA but also enables tolerance of chromosomal aberrations, exacerbating instability across tumor types. In the Rb pathway, loss of restraint on transcription factors drives aberrant expression of genes promoting S-phase entry and , often coupling with checkpoint defects to propagate aneuploid cells. A classic example is in , where human papillomavirus (HPV) E7 oncoprotein binds and degrades Rb, releasing to initiate while transforming host cells into a proliferative state that underpins nearly all HPV-associated malignancies. Therapeutic strategies targeting these dysregulations have advanced cancer management, exemplified by CDK4/6 inhibitors that restore Rb-mediated control. , approved by the FDA in 2015 for hormone receptor-positive, HER2-negative advanced , inhibits D-CDK4/6 complexes to induce G1 arrest, improving when combined with endocrine therapy in clinical trials. Recent post-2020 studies highlight synergies with , where inducing cell cycle arrest in tumors reduces immunosuppressive signaling and enhances T-cell infiltration and anti-tumor responses, as seen in preclinical models of and .

Implications in development and tissue homeostasis

The cell cycle plays a pivotal role in embryonic by enabling rapid proliferation during early stages, such as in the blastula, where cycles are exceptionally short due to abbreviated G1 and G2 phases, often lasting as little as 25 minutes in species like Xenopus laevis. This rapid division supports the initial expansion of cell numbers without intervening growth periods, driven by maternally supplied factors that bypass transcriptional activation until the mid-blastula transition (MBT). As development progresses, cell cycles lengthen with the onset of zygotic transcription and , allowing cells to allocate time for and specialization, a shift observed across metazoans from fast, synchronous cleavages to slower, asynchronous ones. In s, further refines developmental patterning by producing one self-renewing and one committed , with cell cycle regulators like Numb influencing orientation and fate determinants to ensure unequal inheritance of cellular components. This process maintains pools while generating diverse lineages, as seen in neural and epithelial where polarity cues dictate cycle progression and daughter cell fates. Tissue in adults relies on tightly balanced cell cycle dynamics, where in or compartments offsets and to sustain organ function. In the , for instance, crypt-based cells undergo continuous division every 12-24 hours, driving a complete renewal of the villus every 3-5 days through a of Wnt signaling that promotes at the base and extrusion at the tip. This equilibrium is maintained by coordinated in transit-amplifying cells, preventing overgrowth while ensuring barrier integrity. Similarly, hematopoietic cells (HSCs) predominantly reside in quiescence (), cycling infrequently to preserve long-term repopulation capacity and avoid exhaustion, with only a fraction entering active in response to demand. Dysregulation of the cell cycle contributes to non-neoplastic pathologies, such as , where aberrant progression—often involving /M arrest in renal cells—promotes deposition and formation following injury. In aging, accumulation of senescent cells marked by p16^INK4a upregulation enforces permanent G1 arrest, limiting regenerative potential and contributing to tissue dysfunction across organs like the liver and skin. Examples of adaptive cell cycle modulation include , where shortening of the in accelerates re-epithelialization by enhancing expression and progression to . Recent single-cell sequencing studies from the 2010s have revealed heterogeneity in cycle states during development and , such as varying G0/G1 depths in neural cells that dictate reactivation timing and bias, underscoring how stochastic fluctuations influence tissue maintenance.

Evolution

Origins in prokaryotes

Prokaryotic cells, including and , reproduce through binary fission, a process that coordinates , segregation, and without the discrete phases characteristic of eukaryotic . In , initiates at the , oriC, where the initiator protein binds to specific DnaA box motifs in an ATP-dependent manner, unwinding the DNA to allow loading and bidirectional fork progression. segregation follows replication, mediated by the ParABS system in most : ParB proteins bind to centromere-like parS sites near oriC, forming a partition complex that interacts with the ParA to actively move duplicated origins toward opposite cell poles, ensuring equitable distribution to daughter cells. is executed by the divisome, anchored by the Z-ring formed through polymerization of , a that assembles at midcell to constrict the membrane and recruit peptidoglycan-synthesizing enzymes, culminating in formation and . In , chromosome segregation often involves systems like SegAB, where SegA binds to specific DNA sites and SegB acts as a DNA-binding adaptor to facilitate partitioning, showing functional parallels to bacterial ParABS but with distinct molecular components. Replication initiation in is more akin to eukaryotes, utilizing Orc1/Cdc6 proteins that bind origins and load MCM helicases, bridging prokaryotic and eukaryotic mechanisms. Regulation of binary fission is growth-dependent, integrating nutrient availability with replication and division to maintain cell size . The alarmone (p)ppGpp, synthesized during nutrient limitation via the RelA-SpoT pathway, senses amino acid starvation and inhibits replication initiation by reducing activity while delaying division, preventing over-initiation under stress. Unlike eukaryotes, prokaryotes lack cyclin-dependent kinases but employ checkpoints, such as occlusion and Min system in , or equivalent spatial controls in , to ensure replication completes before division, avoiding guillotining of unsegregated DNA. Key components of prokaryotic division show evolutionary conservation with eukaryotic counterparts, underscoring their foundational role. , the core of the Z-ring, is a structural and functional homolog of eukaryotic , sharing activity and filament-forming properties that ancestral forms likely contributed to evolution. Similarly, bacterial and archaeal Orc1/Cdc6 exhibit sequence and functional similarities to the eukaryotic (ORC), both binding origins to license replication, suggesting a common ancestral initiator across domains. Prokaryotic cell cycles are simpler than eukaryotic ones, with overlapping events rather than segregated phases; for instance, in fast-growing , replication forks from a prior cycle may persist into the next, enabling division every 20-60 minutes under optimal conditions. This multifork replication accommodates rapid proliferation but limits complexity, as processes like segregation and division occur concurrently without robust interphase gaps.

Eukaryotic innovations and diversification

The eukaryotic cell cycle originated approximately 2 billion years ago, coinciding with the endosymbiotic acquisition of mitochondria from an α-proteobacterium, which provided efficient aerobic respiration and enabled the energy demands of larger, more complex cells. This event, tied to the (GOE) around 2.4–2.2 billion years ago that increased atmospheric oxygen levels, facilitated the evolution of eukaryotic traits preserved in microfossils dating to about 1.8–2.1 billion years ago. The transition from prokaryotic circular chromosomes to multiple linear chromosomes in eukaryotes necessitated the development of to ensure accurate segregation, as linear ends posed replication and stability challenges absent in circular genomes. Key innovations in the eukaryotic cell cycle arose to accommodate the and complexity. The , a defining eukaryotic feature, requires partial or complete breakdown and reformation during —known as open or semi-open in many lineages—to allow access to , contrasting with prokaryotic division lacking such a barrier. The mitotic , composed of , evolved as a dedicated apparatus for segregation, with intranuclear spindles likely ancestral in the last eukaryotic common ancestor (LECA) before diversification into open and closed forms. Core regulators like cyclin-dependent kinases (CDKs) trace their origins to bacterial protein kinases, with binding providing oscillatory activation for phased progression; CDK1, in particular, is universally conserved across eukaryotes for driving G2/M transition. Diversification of the cell cycle occurred with increasing organismal complexity, particularly in multicellular lineages. The G1/S checkpoint evolved to integrate environmental cues and prevent premature DNA replication, with the retinoblastoma (Rb) protein emerging as a key repressor in both animal and plant lineages to enforce this control during multicellular development. In animals, D-type cyclins, which are also present in plants but absent in many unicellular eukaryotes such as yeast, fine-tune G1 progression in response to growth signals. Fungi exhibit variations such as a shortened or absent G1 phase in species like budding yeast, prioritizing rapid division over decision-making, unlike the extended G1 in animal cells that allows for size control and differentiation. Comparative genomics supports these innovations, revealing CDK1 as a universal eukaryotic regulator while D-type cyclins and Rb-related proteins expanded in multicellular clades, reflecting adaptations to tissue homeostasis. Fossil evidence links this diversification to post-GOE oxygenation, enabling mitochondrial integration and the phased cycles that underpin eukaryotic complexity.

References

  1. [1]
    An Overview of the Cell Cycle - Molecular Biology of the Cell - NCBI
    The phases of the cell cycle. The cell grows continuously in interphase, which consists of three phases: DNA replication is confined to S phase; G1 is the gap ...
  2. [2]
    The Cell Cycle and Programmed Cell Death - NCBI - NIH
    This cycle of duplication and division, known as the cell cycle, is the essential mechanism by which all living things reproduce. In unicellular species, such ...
  3. [3]
    Cell Cycle - National Human Genome Research Institute
    Cell cycle is the name we give the process through which cells replicate and make two new cells. Cell cycle has different stages called G1, S, G2, and M. G1 is ...
  4. [4]
    6.1 The Cell Cycle – Human Biology
    The cell cycle is an ordered series of events involving cell growth and cell division that produces two new daughter cells.
  5. [5]
    The Eukaryotic Cell Cycle - NCBI - NIH
    The division cycle of most cells consists of four coordinated processes: cell growth, DNA replication, distribution of the duplicated chromosomes to daughter ...
  6. [6]
    The Roles of Cyclin-Dependent Kinases in Cell-Cycle Progression ...
    Mar 13, 2020 · The formation of cyclin/CDKs controls the cell-cycle progression via phosphorylation of the target genes, such as tumor suppressor protein ...
  7. [7]
    Cyclin-dependent protein kinases and cell cycle regulation in ...
    Jan 13, 2025 · Cyclin Dependent Kinases (CDKs) are closely connected to the regulation of cell cycle progression, having been first identified as the kinases able to drive ...
  8. [8]
    Cell Cycle Regulation by Checkpoints - PMC - PubMed Central - NIH
    Cell cycle checkpoints are surveillance mechanisms that monitor the order, integrity, and fidelity of the major events of the cell cycle.
  9. [9]
    How do genes control the growth and division of cells? - MedlinePlus
    Mar 26, 2021 · The cell cycle has checkpoints (also called restriction points), which allow certain genes to check for problems and halt the cycle for repairs ...
  10. [10]
    Cdks, cyclins and CKIs: roles beyond cell cycle regulation - PubMed
    Mammalian Cdks, cyclins and CKIs play indispensable roles in processes such as transcription, epigenetic regulation, metabolism, stem cell self-renewal, ...
  11. [11]
    INTRODUCTION to Cell Division - CalState-Pressbooks Network
    All multicellular organisms use cell division for growth, maintenance, and repair of tissues. Cell division is tightly regulated, and the occasional failure ...
  12. [12]
    Definition of cell cycle - NCI Dictionary of Cancer Terms
    The cell cycle consists of a series of steps during which the chromosomes and other cell material double to make two copies.
  13. [13]
    THE CELL CYCLE AND MYC INTERSECT WITH MECHANISMS ...
    Murine ESCs divide rapidly with generation times of ~8–10 hours and ... Cyclin synthesis drives the early embryonic cell cycle. Nature. 1989;339:275 ...
  14. [14]
    The Role of Model Organisms in the History of Mitosis Research - PMC
    Flemming described the behavior of chromosomes during mitosis with amazing accuracy in an 1882 collection entitled, “Cell substance, nucleus and cell division.” ...
  15. [15]
    Walther Flemming: pioneer of mitosis research - Nature
    These results led, in 1882, to the publication of Flemming?s comprehensive book Zellsubstanz, Kern und Zelltheilung (?Cell substance, nucleus and cell division ...Missing: primary | Show results with:primary
  16. [16]
    Tritiated Thymidine: A Breakthrough in DNA Replication and Repair
    Jul 1, 2014 · Tritium was discovered by Ernest Rutherford and colleagues in 1934 ... 1950s. As we chatted, she recalled my scintillation counter ...Missing: phase | Show results with:phase
  17. [17]
    A Journey through Time on the Discovery of Cell Cycle Regulation
    Feb 17, 2022 · In the 1960s, the cell cycle was thus defined as a succession of four phases—G1, S, G2, and M—plus an extra G0 phase (Figure 3).
  18. [18]
    Molecular regulation of stem cell quiescence - PMC - PubMed Central
    Somatic cells are able to enter reversible (quiescent) or irreversible (senescent and differentiated) G0 states from the G1 phase of the cell cycle before the ...
  19. [19]
    two stem cell populations in squamous epithelia, reserves and the ...
    Aug 27, 2019 · Overall, the G0 phase is poorly defined at the molecular level. Its occurrence is often described in conjunction with truly post-mitotic cells ...
  20. [20]
    Chromatin structure restricts origin utilization when quiescent cells ...
    Dec 24, 2020 · The chromatin structure around inactive origins in G0 cells showed increased H3 occupancy and decreased nucleosome positioning compared to the same origins in ...Missing: compact | Show results with:compact
  21. [21]
    A Model of Liver Regeneration - PMC - NIH
    After partial hepatectomy, hepatocytes reenter the cell cycle by going from the G0 state to the G1 phase. Cells in the early G1 phase progress, driven by ...
  22. [22]
    Commitment Point during G0→G1 That Controls Entry into the Cell ...
    As classically defined, G0 cells take longer to enter the S phase for the first time from quiescence than in subsequent cell divisions.
  23. [23]
    Regulation of Cell Cycle Progression by Growth Factor-Induced Cell ...
    Nov 26, 2021 · For a typical proliferating human cell, if we assume the total cycle time is 24 h, the duration of G1 phase is approximately 11 h, S phase ...
  24. [24]
  25. [25]
    The kinetic origins of the restriction point in the mammalian cell cycle
    The R‐point occurs in mid‐to‐late G1 phase and marks the transition between mitogendependent to mitogen‐independent progression of the cell cycle.
  26. [26]
    Start and the Restriction Point - PMC - NIH
    Aug 2, 2013 · A pre-Start cell exposed to mating pheromone immediately arrests in G1 while a post-Start cell proceeds once more through the cell cycle [1–3].
  27. [27]
  28. [28]
  29. [29]
    Duration of the Cell Cycle | SparkNotes
    Usually, cells will take between 5 and 6 hours to complete S phase. G2 is shorter, lasting only 3 to 4 hours in most cells. In sum, then, interphase generally ...
  30. [30]
    Pathways for Genome Integrity in G2 Phase of the Cell Cycle - PMC
    DNA repair pathways play a major role in the G2/M checkpoint pathway thereby blocking cell division as long as DNA lesions are present.
  31. [31]
    Evidence that the human cell cycle is a series of uncoupled ...
    Mar 19, 2019 · ... G2 phase cell cycle progression is inhibited in ultraviolet radiation‐induced G2 phase delay. J Biol Chem 274: 13961–13969 [DOI] [PubMed] ...
  32. [32]
    Manipulating cultured mammalian cells for mitosis research - PMC
    In mammalian cultured cells, the duration of mitosis is ~1 hr, and this occurs at a frequency of once every 24 hrs.
  33. [33]
    The Events of M Phase - The Cell - NCBI Bookshelf
    Mitosis is conventionally divided into four stages—prophase, metaphase, anaphase, and telophase—which are illustrated for an animal cell in Figures 14.23 ...
  34. [34]
    Mitosis | Learn Science at Scitable - Nature
    Mitosis consists of five morphologically distinct phases: prophase, prometaphase, metaphase, anaphase, and telophase. Each phase involves characteristic steps ...
  35. [35]
    The roles of microtubule-based motor proteins in mitosis
    Kinesins and dyneins play important roles during cell division. Using RNA interference (RNAi) to deplete individual (or combinations of) motors followed by.
  36. [36]
    Captivating Capture: How Microtubules Attach to Kinetochores
    The mitotic spindle is the molecular machine used to segregate chromosomes to the daughter cells during mitosis. Key to this process is the kinetochore, a ...
  37. [37]
    Spatiotemporal regulation of the anaphase-promoting complex in ...
    Jan 15, 2015 · The APC/C (anaphase-promoting complex; also known as the cyclosome) is an E3 ubiquitin ligase that has a crucial function in the regulation of the mitotic cell ...
  38. [38]
    Motor Function in the Mitotic Spindle Minireview - Cell Press
    Microtubule-based motor proteins provide essential forces for microtubule organization and chromosome movement.
  39. [39]
    The cell biology of open and closed mitosis - PMC - NIH
    Closed mitosis is considered to be the most ancient mechanism of eukaryotic cell division,1 whereas open mitosis appears to have been invented several times ...
  40. [40]
    Cytokinesis - Molecular Biology of the Cell - NCBI Bookshelf
    Cytokinesis occurs by a special mechanism in higher-plant cells—in which the cytoplasm is partitioned by the construction of a new cell wall, the cell plate, ...
  41. [41]
  42. [42]
    Spatiotemporal Regulation of RhoA during Cytokinesis - PMC
    The active form of the small GTPase RhoA is necessary and sufficient for formation of a cytokinetic furrow in animal cells.
  43. [43]
    Phragmoplast expansion: the four-stroke engine that powers plant ...
    The phragmoplast is a plant-specific secretory module that partitions daughter cells during cytokinesis by constructing a cell plate from membranes and ...
  44. [44]
    Organelle segregation during mitosis: Lessons from asymmetrically ...
    Feb 6, 2012 · Interestingly, these studies have revealed that segregation mechanisms frequently link organelle distribution to organelle growth and formation.
  45. [45]
    Understanding Cytokinesis Failure - PMC - PubMed Central - NIH
    Cytokinesis failure leads to both centrosome amplification and production of tetraploid cells, which may set the stage for the development of tumor cells.
  46. [46]
    Dynamics of Cdk1 Substrate Specificity during the Cell Cycle - NIH
    Jun 9, 2011 · Summary. Cdk specificity is determined by the intrinsic selectivity of the active site and by substrate docking sites on the cyclin subunit.
  47. [47]
    Cyclin-dependent protein kinases and cell cycle regulation ... - Nature
    Jan 13, 2025 · There are two Gap phases in somatic cell cycle: G1 separating the M and S phase, and G2 separating the S and M phase- absent in early embryonic ...
  48. [48]
    Multiple levels of cyclin specificity in cell-cycle control
    Cyclins regulate the cell cycle by binding to and activating cyclin-dependent kinases (Cdks). Phosphorylation of specific targets by cyclin–Cdk complexes ...<|control11|><|separator|>
  49. [49]
    Cyclin D-Cdk4,6 Drives Cell-Cycle Progression via the ... - NIH
    A well-known target of cyclin D-Cdk4,6 is the retinoblastoma protein Rb, which inhibits cell-cycle progression until its inactivation by phosphorylation.
  50. [50]
    CDK4/6 initiates Rb inactivation and CDK2 activity coordinates cell ...
    Oct 7, 2022 · We propose that CDK4/6 initiates Rb inactivation and CDK2 activation, which coordinates the timing of cell-cycle commitment and sequential G1/S transition.
  51. [51]
    Cyclin E/CDK2: DNA Replication, Replication Stress and Genomic ...
    Cyclin E/CDK2 complex mostly controls cell cycle progression and DNA replication through phosphorylation of specific substrates.
  52. [52]
    Control of DNA replication by cyclin-dependent kinases in ...
    Cyclin-dependent kinases (CDKs) are required for initiation of DNA replication in all eukaryotes, and appear to act at multiple levels to control replication ...
  53. [53]
    CCNB1 (cyclin B1)
    Jan 1, 2009 · Cyclin B1 interacts with CDK1 to form a complex known as the maturation-promoting factor (MPF), which is essential for cell cycle progression ...
  54. [54]
    Dissecting the mechanisms of cell division
    Jun 7, 2019 · These studies underscored that post-translational modifications and cyclin–kinase complexes play roles at the heart of the cell division program.Genetic Dissection Of Cell... · Proteomic Dissection Of Cell... · Future Perspectives
  55. [55]
    [PDF] The anaphase promoting complex/ cyclosome - UC Berkeley MCB
    Aug 9, 2006 · In metaphase, when all kinetochores are attached to microtubules, APC/CCdc20 ubiquitylates securin and cyclin B and thereby activates the ...
  56. [56]
    Switches and latches: a biochemical tug-of-war between the kinases ...
    This engages the positive and double-negative feedback loops, and leads to further Cdc25 activation and Wee1 inhibition, resulting in abrupt activation of Cdk1: ...
  57. [57]
    Mitotic progression becomes irreversible in prometaphase and ...
    Feb 16, 2011 · Cdk1AF short-circuits the Wee1 and Cdc25 feedback loops, causing Cdk1 activity to oscillate rapidly but with lower amplitude. Importantly, this ...
  58. [58]
    Mitotic kinase oscillation governs the latching of cell cycle switches
    May 2, 2022 · Novak and Tyson show that autonomous oscillations of S-phase kinase and mitotic exit-phosphatase activities are suppressed by a “latching-gate” mechanism.
  59. [59]
    Temporal self-organization of the cyclin/Cdk network driving ... - PNAS
    We propose an integrated computational model for the network of cyclin-dependent kinases (Cdks) that controls the dynamics of the mammalian cell cycle.
  60. [60]
    Ubiquitylation and proteasomal degradation of the p21Cip1 ... - NIH
    The Inhibitor of CDKs (INK4) family includes p15INK4B, p16INK4A, p18INK4C and p19INK4D; these CKIs specifically bind CDK4 and CDK6 and inhibit cyclin D ...
  61. [61]
    Inhibition of cyclin-dependent kinases by p21 - PubMed - NIH
    p21Cip1 is a cyclin-dependent kinase (Cdk) inhibitor that is transcriptionally activated by p53 in response to DNA damage. We have explored the interaction ...
  62. [62]
    Growth arrest by the cyclin-dependent kinase inhibitor p27Kip1 is ...
    Neither Myc itself nor other G1-cyclin/CDK complexes were directly responsible for p27 sequestration. ... Cdk inhibitors cooperate to induce cell cycle arrest in ...
  63. [63]
    Differential Roles for Cyclin-Dependent Kinase Inhibitors p21 and ...
    These data suggest that failure to phosphorylate pRb is a key mechanism for the cell cycle arrest of senescent cells. Phosphorylation of pRb during G1 phase is ...
  64. [64]
    The Molecular Balancing Act of p16INK4a in Cancer and Aging - NIH
    Long term p16 INK4a expression pushes cells to enter senescence, an irreversible cell cycle arrest that prevents the growth of would-be cancer cells, but also ...
  65. [65]
    P16INK4A—More Than a Senescence Marker - PMC - NIH
    P16 INK4A is a tumor suppressor and cell cycle regulator that has been linked to aging and senescence.
  66. [66]
    Cell cycle exit during terminal erythroid differentiation is associated ...
    Cell cycle exit during terminal erythroid differentiation is associated with accumulation of p27(Kip1) and inactivation of cdk2 kinase · Authors · Affiliation.<|separator|>
  67. [67]
    CDK inhibitors in cancer therapy, an overview of recent development
    Flavopiridol mainly inhibits the activities of CDK1, CDK2, CDK4, CDK6, CDK7 and CDK9 with IC50 values at 30, 170, 100, 60, 300 and 10 nM, respectively [17].
  68. [68]
    The Role of CDK4/6 Inhibition in Breast Cancer - PMC - NIH
    In this review, we outline the development of CDK inhibitory therapy in breast cancer, including the initial experience with the pan-CDK inhibitor flavopiridol ...
  69. [69]
    The Therapeutic Potential of CDK4/6 Inhibitors, Novel Cancer Drugs ...
    In recent years, the FDA has approved several CDK4/6 inhibitors, such as palbociclib, ribociclib and abemaciclib, for the treatment of HR-positive breast cancer ...
  70. [70]
    The Strange Case of CDK4/6 Inhibitors: Mechanisms, Resistance ...
    Jan 1, 2018 · This review focuses on the mechanism of action and biomarkers to direct the precision use of CDK4/6 inhibitors and rationally-developed combination therapies.
  71. [71]
    E2F-dependent transcription determines replication capacity and S ...
    Jul 14, 2020 · DNA replication timing is tightly regulated during S-phase. S-phase length is determined by DNA synthesis rate, which depends on the number ...
  72. [72]
    FOXM1, a typical proliferation-associated transcription factor - PubMed
    Accordingly, FOXM1 regulates genes that control G1/S-transition, S-phase progression, G2/M-transition and M-phase progression.
  73. [73]
    Comprehensive Identification of Cell Cycle–regulated Genes of the ...
    This procedure identified a total of 800 yeast genes as being periodically regulated.
  74. [74]
    Role of miR-15/16 in CLL | Cell Death & Differentiation - Nature
    Jun 27, 2014 · Two other recent reports demonstrated that miR-15/16 directly target critical cell cycle regulator Cyclin D1 in bladder cancer and osteosarcoma.
  75. [75]
    Control of cell growth by the SCF and APC/C ubiquitin ligases - NIH
    The SCF complex and the APC/C have been demonstrated to be the major E3s controlling the degradation of cell cycle regulators, although they play additional, ...
  76. [76]
    The MDM2-p53 pathway revisited - PMC - PubMed Central
    In normal cells, p53 is kept at low levels by murine double minute 2 (MDM2), an ubiquitin ligase. MDM2 and p53 form a negative-feedback loop, in which p53 ...
  77. [77]
    Spatiotemporal orchestration of mitosis by cyclin-dependent kinase
    Jun 25, 2025 · These results suggest that mitotic CDK activation occurs first in the nucleus before activation in other compartments, including the cytoplasm ...
  78. [78]
    Cell cycle expression heterogeneity predicts degree of differentiation
    Jul 22, 2024 · We developed a method in R (stemFinder) that predicts single cell differentiation time based on heterogeneity in cell cycle gene expression.
  79. [79]
    A restriction point for control of normal animal cell proliferation
    The restriction point control is proposed to permit normal cells to retain viability by a shift to a minimal metabolism upon differentiation in vivo and in ...
  80. [80]
    Cell cycle regulation: p53-p21-RB signaling - Nature
    Mar 31, 2022 · Furthermore, RB can be inactivated through binding of the MDM2 protein, which is often overexpressed in tumors. MDM2 causes proteolytic ...Missing: feedback | Show results with:feedback
  81. [81]
    unified model for the G1/S cell cycle transition - Oxford Academic
    Nov 9, 2020 · This unified model for the G1/S cell cycle transition combines the findings of decades of study, and provides an updated foundation for cell cycle research.
  82. [82]
    G1 Phase Cell Cycle Checkpoint - an overview | ScienceDirect Topics
    The G1 restriction point is a G1 stage marked by the inactivation of the retinoblastoma proteins at which cells are committed to divide independently on the ...
  83. [83]
    A reversible, p53-dependent G0/G1 cell cycle arrest induced by ...
    Abstract. Cells with a functional p53 pathway undergo a G0/G1 arrest or apoptosis when treated with gamma radiation or many chemotherapeutic drugs.
  84. [84]
    p21-induced cycle arrest in G1 protects cells from apoptosis ... - Nature
    Jul 14, 1998 · p21-induced cycle arrest in G1 protects cells from apoptosis induced by UV-irradiation or RNA polymerase II blockage. N Bissonnette &; D J ...
  85. [85]
    Role and Regulation of p53 during an Ultraviolet Radiation-induced ...
    These results indicate that UV can inhibit G 1 to S-phase progression through p53-dependent and independent mechanisms.
  86. [86]
    G1 checkpoint establishment in vivo during embryonic liver ...
    May 19, 2014 · During embryonic development, the characteristics of cell cycle and DNA damage checkpoint evolve from an extremely short G1 cell phase and ...
  87. [87]
    A short G1 phase imposes constitutive replication stress and fork ...
    Feb 15, 2016 · Furthermore, ESCs are reported to have a compromised G1–S checkpoint. The tumour suppressor protein retinoblastoma, which is required for ...
  88. [88]
    Evolution of opposing regulatory interactions underlies the ... - Nature
    May 27, 2021 · Evolution of opposing regulatory interactions underlies the emergence of eukaryotic cell cycle checkpoints ... G1/S transition. EMBO J. 13 ...
  89. [89]
    The Intra-S Checkpoint Responses to DNA Damage - PMC
    Cells activate the intra-S checkpoint in response to damage during S phase to protect genomic integrity and ensure replication fidelity.
  90. [90]
    The intra-S phase checkpoint directly regulates replication ... - PNAS
    Jun 9, 2021 · This study demonstrates that the intra-S phase checkpoint directly regulates replication elongation, reduces CMG helicase processivity, prevents CMG helicase ...
  91. [91]
    The essential kinase ATR: ensuring faithful duplication of a ... - NIH
    One crucial function of the ATR pathway is to arrest the cell cycle following DNA damage in S phase. This arrest is initiated by the phosphorylation of CHK1 by ...
  92. [92]
    Chk1 inhibits replication factory activation but allows dormant origin ...
    At low levels of replication stress, Chk1 favors resolving problems at stalled replication forks over initiating origin firing in unreplicated areas of the.
  93. [93]
    SCFβ-TRCP links Chk1 signaling to degradation of the Cdc25A ...
    IR during S phase leads to accelerated Cdc25A phosphorylation by Chk1 with a concomitant increase in turnover. Defects in this intra-S-phase checkpoint lead to ...
  94. [94]
    Article Chk1 regulates the S phase checkpoint by coupling the ...
    Here we report that Chk1 phosphorylates Cdc25A and thereby controls the Cdc25A protein turnover in unperturbed cell cycles.
  95. [95]
    Regulation of DNA replication by the S-phase DNA damage ...
    Jul 3, 2009 · The G1/S and G2/M DNA damage checkpoints prevent cell-cycle progression into S-phase and M-phase, respectively. Additionally, the S-M checkpoint ...
  96. [96]
    Exploiting DNA Replication Stress for Cancer Treatment
    Apr 15, 2019 · In addition to S phase checkpoint response proteins being the leading targets for cancer ... Table 1. Chemotherapeutics that increase DNA ...
  97. [97]
    ATR Regulates Fragile Site Stability - ScienceDirect.com
    Common fragile sites are loci that exhibit gaps and breaks on metaphase chromosomes of cells that have been cultured under conditions of replicative stress, ...
  98. [98]
    Insights into common fragile site instability: DNA replication ...
    Common fragile sites (CFS) are specific genomic regions prone to chromosomal instability under conditions of DNA replication stress. CFSs manifest as breaks, ...
  99. [99]
    Review The G2-phase DNA-damage checkpoint - ScienceDirect.com
    Here, we review current understanding of the organization and functions of the ATM–Chk2 and ATR–Chk1 pathways and the prospects for targeting DNA damage ...
  100. [100]
    The G2 checkpoint—a node‐based molecular switch - FEBS Press
    Feb 10, 2017 · In summary, the G2 checkpoint is an ingenious node-based molecular switch which outcome is determined by the interplay of the PLK1, CHK1, Wee1, ...
  101. [101]
    Kinases that Control the Cell Cycle in Response to DNA Damage
    The ATM-Chk2-Cdc25A checkpoint pathway guards against radioresistant DNA synthesis. ... G2/M checkpoint after ultraviolet radiation requires p38 kinase.
  102. [102]
    Regulating mammalian checkpoints through Cdc25 inactivation
    This review focuses on our understanding of the biochemical mechanisms that specifically inactivate Cdc25 (cell division cycle 25) phosphatases to achieve this.Missing: feedback | Show results with:feedback
  103. [103]
    p53 regulates a G2 checkpoint through cyclin B1 - PNAS
    We have found that p53 prevents G2/M transition by decreasing intracellular levels of cyclin B1 protein and attenuating the activity of the cyclin B1 promoter.
  104. [104]
    The G(2) DNA damage checkpoint targets both Wee1 and Cdc25
    In the fission yeast Schizosaccharomyces pombe, detection of DNA damage in G(2) activates a checkpoint that prevents entry into mitosis through the maintenance ...Missing: seminal M ATM Chk2
  105. [105]
    G2 checkpoint abrogation and checkpoint kinase-1 targeting in the ...
    Jan 29, 2008 · In response to DNA damage, the checkpoint kinases phosphorylate CDC25 phosphatases, resulting in CDC25 inactivation through either ubiquitin- ...
  106. [106]
    Mitotic death: a mechanism of survival? A review - PubMed Central
    Double strand breaks can be repaired by homologous recombination during G2 arrest. ... G2/M checkpoint. Thus, reaching the G2 compartment would appear crucial for ...
  107. [107]
    Targeting cell cycle regulation via the G2-M checkpoint for synthetic ...
    Bypassing the G2-M checkpoint leads to aberrant mitosis and mitotic catastrophe ... G2 checkpoint and inhibition of homologous recombinational DNA repair.Chk1 Inhibition · Wee1 Inhibition · Combination Of Chk1 And Wee1...
  108. [108]
    DNA Damage Mediated S and G2 Checkpoints in Human ... - NIH
    Somatic cells respond to DNA damage at different phases of the cell cycle by activating sets of checkpoint pathways. As cell cycle arrest in G1 prevents cells ...
  109. [109]
    Deficiency in the response to DNA double-strand breaks in mouse ...
    We precisely examined the G2/M checkpoint in one-cell embryos ... embryos, and in reduced quality and developmental potential of aged oocytes and embryos.
  110. [110]
    The APC/C maintains the spindle assembly checkpoint by targeting ...
    The Spindle Assembly Checkpoint (SAC) is required to block sister chromatid separation until all chromosomes are properly attached to the mitotic apparatus.Cdc20 Is Degraded By The... · An Inactive Cdc20 Cannot Be... · Bubr1 And Not Mad2 Is The...
  111. [111]
    The spindle checkpoint | Journal of Cell Science
    Oct 15, 2006 · The core spindle checkpoint proteins are Mad1, Mad2, BubR1 (Mad3 in yeast), Bub1, Bub3 and Mps1. The Mad and Bub proteins were first identified ...Activation Of The Checkpoint · Anaphase Delay · Silencing The Checkpoint
  112. [112]
  113. [113]
    Spatial-temporal model for silencing of the mitotic spindle assembly ...
    Sep 12, 2014 · This checkpoint surveillance mechanism is highly sensitive and efficient. A single unattached chromosome can delay anaphase onset for hours, ...
  114. [114]
  115. [115]
    Characterization of unconventional kinetochore kinases KKT10 and ...
    Apr 29, 2020 · Summary: Trypanosomes lack a canonical spindle checkpoint system, but kinetochore kinases KKT10/19 phosphorylate KKT4 and KKT7 and are ...
  116. [116]
    A concise guide to fluorescent cell cycle reporters for live-cell imaging
    Genetically encoded fluorescent cell cycle reporters have become indispensable tools for studying the cell cycle, providing invaluable insights into cell cycle ...Missing: papers | Show results with:papers
  117. [117]
    CDK activity sensors: genetically encoded ratiometric biosensors for ...
    CDK activity sensors are fluorescent biosensors that change location upon CDK activation, used to study the cell cycle in live cells. They are single-color, ...
  118. [118]
    Live-cell imaging defines a threshold in CDK activity at the G2/M ...
    A FRET biosensor is developed to monitor the CDK activity in living fission yeast. The sensor detects CDK activity, including a slight activation in the S ...
  119. [119]
    Capturing CDKs in Action: Live-Cell Biosensors Pioneer the New
    Mar 5, 2025 · modes of action in CDK activity biosensors—FRET-based and translocation-based biosensors— ... CDK-activity sensor marks cell cycle entry and ...
  120. [120]
    Translocation of cyclin B1 to the nucleus at prophase requires a ...
    We have used real-time imaging of cyclin B1–green fluorescent protein (GFP) fusion proteins as cells enter mitosis to address the mechanism of translocation. We ...
  121. [121]
    The disappearance of cyclin B at the end of mitosis is regulated ...
    To visualize the distribution and destruction of cyclin B directly during the cell cycle, we expressed a cyclin B–GFP fusion protein in Drosophila embryos using ...
  122. [122]
    Quantitative Imaging of B1 Cyclin Expression Across the Cell Cycle ...
    Jul 1, 2020 · In this article, we report the number of cyclin B1 proteins tagged with enhanced green fluorescent protein (eGFP) in fixed U-2 OS cells ...
  123. [123]
    Quantitative Cell Cycle Analysis Based on an Endogenous All-in ...
    May 30, 2017 · Overexpression of PCNA fused at the N terminus to a fluorescent protein is widely used to label cells in S phase based on the presence of ...
  124. [124]
    replication patterns are modulated by S‐phase checkpoint kinases ...
    PCNA fluorescence was diffuse during the M, G1 and G2 phases of the cell cycle, but became punctate in early S phase, which was identified by the cell's ...
  125. [125]
    Changes in Chromatin Compaction During the Cell Cycle Revealed ...
    Feb 8, 2012 · By visual inspection of the H2B-mCherry marked nuclei, we select a cell in the early stage of mitosis and follow it until early interphase. In ...
  126. [126]
    Live-cell imaging of mitosis in Caenorhabditis elegans embryos
    For imaging, C. elegans embryos provide large clear cells, an invariant pattern of cell division, only six chromosomes, a very short cell cycle, and remain ...
  127. [127]
  128. [128]
    Super-Resolution Imaging of Mitotic Spindle Microtubules Using ...
    Stimulated emission depletion (STED) microscopy is a powerful super-resolution imaging technique that only recently entered the field of mitosis, ...
  129. [129]
    Super-Resolution Imaging of Mitotic Spindle Microtubules Using ...
    Here, we describe immunofluorescence combined with STED microscopy as a method for analyzing microtubules and kinetochore-microtubule attachments in human ...Missing: cycle | Show results with:cycle
  130. [130]
    Cell cycle analysis of a cell proliferation-associated human nuclear ...
    The aim of the present investigation was to clarify whether the Ki-67 nuclear antigen is restricted in its expression to certain phases of the cell cycle.
  131. [131]
    Cell Cycle Analysis by Mass Cytometry - PubMed
    This method describes the use of mass cytometry for the analysis of cell cycle state for cells from three different sources.
  132. [132]
    Synchronization of cell populations in G1/S and G2/M phases of the ...
    Double thymidine blocks DNA synthesis for G1/S, then nocodazole inhibits mitosis for G2/M. Flow cytometry monitors cell cycle progression.
  133. [133]
    Kinase independent oncogenic cyclin D1 - PMC - NIH
    Jul 7, 2015 · Strong evidence implicates cyclin D1 overexpression as a driving force in breast cancer and many other types of human tumors.
  134. [134]
    The Rb Pathway and Cancer Therapeutics - PMC - NIH
    The intimate link between the Rb pathway and cancer development suggests that the status of Rb activity can potentially be used to develop targeted therapy.The Rb Pathway And Cancer... · Rb And E2f Family Proteins · Mammalian Rb Family Proteins
  135. [135]
    Understanding the function–structure and function–mutation ... - PNAS
    Somatic TP53 mutations are the most common (≈50%) genetic alteration in human cancer (12), and a large number of TP53 mutations have been assembled in two major ...
  136. [136]
    The Role of HPV E6 and E7 Oncoproteins in HPV-associated ... - NIH
    E7 binds to a region of the Rb protein commonly referred to as the 'pocket domains' (4). The 'pocket domain' sequences of Rb are essential for its tumor ...
  137. [137]
    Ibrance (Palbociclib): First CDK4 and CDK6 Inhibitor FDA Approved ...
    Ibrance (Palbociclib): First CDK4 and CDK6 Inhibitor FDA Approved for the Treatment of Postmenopausal Women with Metastatic Breast Cancer · Palbociclib Receives ...
  138. [138]
    Cell cycle regulation meets tumor immunosuppression - PMC
    May 28, 2025 · Recent studies indicate that enhanced cell cycle activity in cancer cells suppresses anti-tumor immunity. Here, we discuss potential mechanisms ...
  139. [139]
    A cell cycle-coordinated Polymerase II transcription compartment ...
    Feb 11, 2019 · Most metazoan embryos commence development with rapid, transcriptionally silent cell divisions, with genome activation delayed until the ...
  140. [140]
    Emerging mechanisms of asymmetric stem cell division
    Sep 19, 2018 · Asymmetric division of stem cells creates one stem cell and one differentiating cell, a simple yet elegant way to balance stem cell self-renewal ...
  141. [141]
    Apoptosis, Necrosis, and Necroptosis in the Gut and Intestinal ...
    Intestinal epithelial homeostasis is maintained by a strict equilibrium between cell proliferation in the crypt and cell shedding from the villus tip. In the ...
  142. [142]
    Regulation and plasticity of intestinal stem cells during homeostasis ...
    Oct 15, 2016 · When proliferation is inhibited in intestinal organoids, the Wnt gradient in the crypts collapses, implying that Wnt spreads by proliferative ...Wnt · Progenitor Cell Plasticity · Dedifferentiation Of...
  143. [143]
    Molecular regulation of hematopoietic stem cell quiescence - PMC
    LT-HSCs are largely cell-cycle-quiescent (G0 phase) with low biosynthetic activity, while ST-HSCs are broadly considered as activated stem cells. LT-HSCs and ST ...
  144. [144]
    Cell Cycle Dysregulation and Renal Fibrosis - PMC - PubMed Central
    Nov 25, 2021 · Cell cycle dysregulation is associated with many human diseases including renal fibrosis, a common process of various chronic kidney diseases progressing to ...Missing: shortened | Show results with:shortened
  145. [145]
    Cell-cycle Regulatory Proteins in Human Wound Healing - PubMed
    These data indicate that downmodulation of several G(1)/S-phase cyclins and a relative excess of CKIs may cooperate to ensure the quiescent state of migrating ...Missing: shortening | Show results with:shortening
  146. [146]
    Cell cycle heterogeneity directs the timing of neural stem ... - Science
    Apr 6, 2018 · We demonstrate that quiescent NSCs (qNSCs) are arrested in either G 2 or G 0. G 2 -G 0 heterogeneity directs NSC behavior: G 2 qNSCs reactivate before G 0 ...
  147. [147]
    Cell cycle proliferation decisions: the impact of single cell analyses
    Actively proliferating cells have at least two decision points, the first in G2 phase and the second in G1, whereas cells entering the cycle from G0 (quiescence) ...
  148. [148]
    Bacterial cell proliferation: from molecules to cells - PubMed Central
    DnaA binds to multiple sites (DnaA box motifs) within oriC in an ordered manner according to the affinity of each motif to DnaA in its ATP- or ADP-bound forms.
  149. [149]
    Bacterial chromosome segregation by the ParABS system - Journals
    Jun 17, 2020 · In the majority of bacterial species, faithful chromosome segregation is mediated by the tripartite ParABS system, consisting of an ATPase protein ParA, a ...Abstract · Introduction · The ParB-DNA and SMC... · The evolution of the ParABS...
  150. [150]
    At the heart of bacterial cytokinesis: the Z ring - PMC - NIH
    Bacterial cell division is mediated by the divisome which is organized by the Z ring, a cytoskeletal element formed by the polymerization of the tubulin ...
  151. [151]
    Effects of (p)ppGpp on the Progression of the Cell Cycle of ...
    Bacteria must control the progression of their cell cycle in response to nutrient availability. This regulation can be mediated by guanosine tetra- or ...
  152. [152]
    Tubulin and its Prokaryotic Homologue FtsZ: A Structural and ... - NIH
    FtsZ is a close structural homologue of tubulin within prokaryotes, and plays an important structural role during cell division. This article compares what is ...
  153. [153]
    Common domains in the initiators of DNA replication in Bacteria ...
    In this article new data concerning the presence of common features in the initiators of chromosomal replication in bacteria, archaea and eukaryotes are ...
  154. [154]
    New insights into the assembly and regulation of the bacterial ...
    In the case of fast-growing cells such as Escherichia coli, this act of binary fission needs to happen every ~20 minutes, and requires perfect coordination ...Conserved Divisome Proteins · Stages Of Divisome Assembly · Divisome Proteins As...<|separator|>
  155. [155]
    A look at the parameters that regulate cell‐cycle events: EMBO reports
    Aug 1, 2003 · At high growth rates, E. coli cell cycles are actually overlapping, so that the DNA replication of one cell cycle may occur before the ...Missing: limitations | Show results with:limitations<|control11|><|separator|>
  156. [156]
    Origin of Mitochondria | Learn Science at Scitable - Nature
    Mitochondria arose through a fateful endosymbiosis more than 1.45 billion years ago. Many mitochondria make ATP without the help of oxygen.
  157. [157]
    The origin of eukaryotes and rise in complexity were synchronous ...
    Sep 1, 2023 · The evolution of complex eukaryotic life has frequently been associated with the Great Oxidation Event (GOE), 2.43–2.22 Ga (Poulton et al., ...
  158. [158]
    On the Origin of the Eukaryotic Chromosome - NIH
    May 22, 2013 · The birth of multiple eukaryotic linear chromosomes was the key innovation that allowed adaptive evolution by means of transient aneuploidy ( ...
  159. [159]
    Ancestral Mitotic State: Closed Orthomitosis With Intranuclear ...
    The nuclear envelope can either remain intact during mitosis (closed mitosis) or it can be partly or completely dispersed (semi-open or open mitosis).
  160. [160]
    Recycling the Cell Cycle: Cyclins Revisited - ScienceDirect.com
    Jan 23, 2004 · I discuss how a cyclin/Cdk-based engine could have evolved to assume control of the cell cycle from other, older protein kinases.
  161. [161]
    The ancient function of RB-E2F Pathway - Biology Direct
    Sep 20, 2010 · The RB-E2F pathway is conserved in most eukaryotic lineages, including animals and plants. E2F and RB family proteins perform crucial functions ...
  162. [162]
    Molecular evolution of cyclin proteins in animals and fungi
    Jul 28, 2011 · The association of cyclins with CDKs is the condition requisite for their activation [1–3]. Cyclins have been discovered in sea urchin eggs as ...
  163. [163]
    Fungal Cell Cycle: A Unicellular versus Multicellular Comparison
    We compare and contrast morphological landmarks of G1, S, G2, and M phases, molecular mechanisms that drive cell cycle progression, and checkpoints in these ...Missing: variations | Show results with:variations
  164. [164]
    Comparative genomics of cyclin-dependent kinases suggest co ...
    Here we identify 123 CDK family members from animals, plants, yeasts, and four protists from which genome sequences have been completed, and 10 additional CDKs ...Missing: universal | Show results with:universal
  165. [165]
    Study suggests complex life was present on Earth 2.33 billion years ...
    Mar 6, 2017 · ... Great Oxidation Event. Summons says the possibility that eukaryotes may have existed around the same time makes sense, as they would have ...<|control11|><|separator|>