Fact-checked by Grok 2 weeks ago

Maturation promoting factor

Maturation-promoting factor (MPF), also known as M-phase promoting factor, is a universal complex in eukaryotic cells that induces entry into the M phase of the , specifically driving the G2/M transition to trigger or . It consists of a catalytic subunit, (CDK1, formerly Cdc2), bound to a regulatory subunit, which together exhibit histone kinase activity essential for breakdown, condensation, and spindle assembly. MPF was first identified in 1971 through experiments in which cytoplasm from mature Rana pipiens oocytes induced maturation in immature oocytes, revealing a transferable activity independent of new protein synthesis. Subsequent studies in laevis and oocytes confirmed its role as an intracellular regulator, with purification in 1988 identifying the 32-kDa CDK1 and 45-kDa components that correlate with MPF activity. This discovery laid foundational insights into control, linking MPF to the broader cyclin-CDK oscillator mechanism that governs periodic progression through the cell cycle phases. MPF activity is tightly regulated to ensure precise timing of M-phase entry, primarily through cyclin B accumulation during , followed by activating of CDK1 at threonine 161 and inhibitory phosphorylations at tyrosine 15 and threonine 14 that are relieved by phosphatases. Inactivation occurs via ubiquitin-mediated degradation of by the anaphase-promoting complex/cyclosome (APC/C), allowing exit from M phase. Additional regulators, such as Greatwall , enhance MPF function by inhibiting 2A-B55 to stabilize CDK1 substrates. Dysregulation of MPF contributes to disorders, including cancer, underscoring its critical role in cellular proliferation and development.

History and Discovery

Early Observations in Oocyte Maturation

In the 1960s and 1970s, studies on amphibian maturation shifted from a primary emphasis on hormonal triggers to the recognition of intracellular regulatory factors, particularly in species like the frog Rana pipiens. Early had established that hormones such as progesterone, released from follicular cells, were essential for initiating maturation in intact oocytes, but experiments involving defolliculation revealed that isolated oocytes could still respond to the hormone, suggesting downstream cytoplasmic mediators. This transition highlighted the role of oocyte-intrinsic components in controlling nuclear and cytoplasmic changes during resumption.21002-2) Pioneering experiments by Yoshio Masui and Clement L. Markert in the early 1970s provided direct evidence for such an intracellular factor. Using Rana pipiens oocytes, they demonstrated that cytoplasm extracted from progesterone-treated, maturing oocytes could induce maturation when microinjected into untreated, immature recipient oocytes, even in the absence of exogenous hormones. This transfer bypassed the need for hormonal stimulation, showing that the inducing activity resided in the donor cytoplasm and was sufficient to trigger the maturation process. These findings, reported in 1971, marked the initial observation of what would later be termed (MPF). Oocyte maturation in amphibians involves distinct stages, with germinal vesicle breakdown (GVBD) serving as a hallmark event signaling the resumption of I. GVBD entails the dissolution of the and the disappearance of the within the large germinal vesicle of the prophase-arrested , leading to condensation and formation. In Rana pipiens, progesterone treatment of defolliculated s typically induces GVBD within 8-10 hours, reflecting the of intracellular pathways that coordinate these events with cytoplasmic rearrangements, such as pigment migration and cortex reorganization. The assay for MPF activity relied on quantitative cytoplasmic transfer experiments, where small volumes (approximately 50-100 nl) of from maturing donor oocytes were injected into recipients, and the percentage of induced GVBD was measured as a proxy for activity levels. This method revealed that MPF-like activity appeared in the shortly after progesterone exposure and could be modulated by inhibitors like , which blocked and thus prevented activity buildup if applied early. studies further localized the factor to the region of the ooplasm, confirming its cytoplasmic nature. Later work identified MPF as a , but these early assays established its phenomenological role in maturation.

Identification of the Factor and Key Experiments

The identification of maturation-promoting factor (MPF) as a distinct biochemical entity advanced significantly in the through collaborative efforts between Yoshio Masui's , which refined assays into immature laevis oocytes to quantify MPF activity based on germinal vesicle breakdown and maturation progression, and James Maller's , which utilized assays to probe its enzymatic properties. These assays established that MPF induces oocyte maturation with high efficiency, where one unit of activity corresponds to 50% maturation in recipient s, and demonstrated its species-nonspecific nature across amphibian species. Early biochemical by Maller's group revealed that MPF activation correlates with a marked increase in during oocyte maturation, prompting the hypothesis that MPF functions as a or kinase activator; this was confirmed through assays showing MPF-dependent labeling of oocyte proteins, particularly histones and lamins.90352-6)90251-2) A breakthrough enabling studies occurred in when Manfred Lohka, working with Masui, developed cell-free extracts from activated Rana pipiens eggs that recapitulated nuclear remodeling events; these extracts induced pronuclear formation and condensation in added Xenopus sperm chromatin, providing a to monitor MPF-driven processes without intact cells. Subsequent refinement by Lohka and Maller in 1985 demonstrated that adding active MPF to extracts triggered breakdown, premature condensation, and microtubule spindle assembly, directly linking MPF to mitotic morphology in a cell-free context. Purification efforts culminated in 1988, when Lohka, Hayes, and Maller isolated MPF from mature laevis eggs using , ion-exchange , and gel filtration, yielding a highly active fraction that induced formation in cell-free systems; SDS-PAGE analysis revealed two predominant polypeptides of 32 kDa and 45 kDa, with the complex exhibiting histone activity essential for its function. The 32 kDa subunit was a whose state modulated activity, though its identity as was a later clarification tied to parallel discoveries. Initial nomenclature distinguished MPF from the extracellular maturation-inducing progesterone, as experiments showed MPF resides in and resists diffusion like small molecules, with activity abolished by proteinase treatment but retained after , resolving any perceived overlap through direct activity transfer assays. Parallel work by in 1982 identified periodic 45-50 kDa phosphoproteins in eggs whose synthesis and degradation oscillate with cell divisions, providing the conceptual framework that these "cyclins" regulate MPF, later confirmed in systems.90358-9) Cell-free extracts further revealed MPF's dynamic nature, with Gerhart and colleagues demonstrating in 1984 that MPF activity oscillates between high (M-phase-like) and low (interphase-like) states in cycling Xenopus egg extracts, mimicking embryonic cell cycle phases through periodic activation and inactivation.

Naming and Initial Characterization

The term "maturation promoting factor" (MPF) originated from studies on amphibian oocyte maturation in the early 1970s, when Yoshio Masui and Clement Markert demonstrated that a cytoplasmic factor from mature frog eggs could induce germinal vesicle breakdown (GVBD) and meiotic resumption in immature oocytes upon microinjection. This factor was initially identified in Rana pipiens oocytes, where it was shown to act independently of the nucleus, highlighting its cytoplasmic nature and role in triggering meiotic progression without requiring ongoing protein synthesis after initial activation. By the mid-1970s, similar activities were observed in other species, leading to the recognition that MPF could also promote mitotic events in cleaving embryos, prompting its generalization as "mitosis promoting factor" in somatic cells. Initial biochemical characterization in the revealed MPF as a heat-stable activity, resistant to incubation at 42°C for up to 30 minutes, yet sensitive to treatment such as pronase, which inactivated it rapidly, indicating a proteinaceous component. Further assays demonstrated that MPF possessed activity, particularly toward , distinguishing it from cAMP-dependent pathways, as its induction by progesterone in oocytes occurred without changes in intracellular levels, and purified MPF was unaffected by cAMP modulators or the heat-stable inhibitor of cAMP-dependent . Unlike signaling, which acts upstream to initiate maturation, MPF was positioned as a downstream effector that directly drove breakdown and condensation. In 1988, the connection between MPF and the cdc2 kinase from fission yeast was established, with William Dunphy and colleagues showing that the homolog of cdc2 (p34cdc2) co-purified with MPF activity and exhibited properties essential for mitotic entry. Paul Nurse's parallel work on cdc2+ further unified these findings, demonstrating that this conserved kinase regulated both G1/S and G2/M transitions across eukaryotes, positioning MPF as the active B-cdc2 complex central to control. Debates on species-specificity arose in the late 1980s, as initial studies suggested potential variations, but by 1990, cross-species experiments confirmed MPF's functional conservation, with and MPF inducing GVBD in oocytes, and vice versa, extending to both vertebrates and . This universality underscored MPF's role as a core regulator beyond , resolving earlier concerns about taxon-specific mechanisms.

Molecular Composition

Core Components

Maturation promoting factor (MPF) is a heterodimeric protein complex composed of cyclin B as the regulatory subunit and cyclin-dependent kinase 1 (CDK1, also known as cdc2) as the catalytic subunit. This complex forms the core of MPF's ability to drive meiotic and mitotic progression in eukaryotic cells. Cyclin B, with a molecular weight of approximately 45-55 kDa depending on the isoform and species, serves as the regulatory component that confers substrate specificity and temporal control to the complex. Among its isoforms, cyclin B1 is the primary form associated with mitosis, where it is synthesized during the G2 phase of the cell cycle through regulated transcription and translation, leading to its accumulation prior to mitotic entry. In contrast, CDK1, the 34 kDa catalytic subunit, is highly conserved across eukaryotes, from yeast cdc2 homologs to vertebrate forms, enabling its universal role in phosphorylating target proteins to initiate M-phase events. Prior to complex formation, CDK1 exists as an inactive monomer incapable of significant kinase activity without cyclin binding. Similarly, cyclin B is inherently unstable on its own and relies on association with CDK1 for functional persistence, as unbound cyclin B is rapidly turned over. The stoichiometric ratio of the MPF complex is 1:1, with one molecule of binding to one molecule of CDK1 to form the active heterodimer. levels fluctuate cyclically, rising via in and declining through at the end of , which directly modulates MPF activity. This composition ensures that MPF activity is tightly coupled to progression.

Assembly and Stoichiometry

The assembly of maturation promoting factor (MPF), composed of and (CDK1), initiates in the late through the binding of newly translated to CDK1 monomers. This interaction is facilitated by the conserved cyclin box domain in , a that specifically recognizes and docks onto a hydrophobic cleft on the CDK1 surface, inducing conformational changes necessary for complex stability. The resulting MPF holoenzyme exhibits a strict equimolar of one molecule per CDK1 subunit, ensuring precise activity control; excess unbound is rapidly degraded via ubiquitin-mediated to prevent aberrant . Prior to full assembly, CDK1 undergoes pre-phosphorylation at 161 (Thr161) by CDK-activating (CAK, typically the CDK7-cyclin H complex), which partially activates the monomer and primes it for binding, although full activity requires subsequent events. Complex formation peaks immediately before the G2/M transition, accumulating pre-MPF reserves that remain inactive due to inhibitory by Wee1 kinase on CDK1 residues Thr14 and Tyr15, thereby enforcing temporal control until cellular conditions permit mitotic entry. This regulated assembly ensures that MPF activation aligns with completion and checkpoint satisfaction.

Structural Features

Cyclin-Dependent Kinase Domain

The (CDK1) serves as the catalytic core of maturation promoting factor (MPF), exhibiting a canonical bilobal fold that is characteristic of the eukaryotic superfamily. This architecture consists of an N-terminal lobe, primarily composed of β-sheets and responsible for binding, and a C-terminal lobe, dominated by α-helices and involved in positioning and . The ATP-binding cleft is located at the interface between these lobes, formed by conserved structural elements such as an extended hairpin from residues 40–46 in the N-lobe, which shapes the pocket for ATP coordination. A hallmark of CDK1 is the PSTAIRE , a (residues 45–51) embedded within the αC-helix of the N-lobe, which contributes to the overall fold stability and serves as a key for regulatory interactions. The activation loop, spanning residues 146–173, includes substrate recognition sites that, in the inactive conformation, obstruct access to the but can reposition to facilitate phosphotransfer. Critical residues within this domain include Thr14 and Tyr15 in the glycine-rich loop (G-loop) of the N-lobe, which are sites of inhibitory that distort the ATP-binding geometry, and Asp128 in the C-lobe, which acts as the catalytic base to abstract a proton from the substrate's hydroxyl group during transfer. CDK1 displays high evolutionary conservation across eukaryotes, sharing approximately 65% sequence identity with CDK2 and homology to yeast Cdc28, reflecting its ancient origin as the primordial cell cycle kinase. Despite this homology, CDK1 has specialized for mitotic functions through unique structural features, such as differences in key loops and a disordered activation segment in its monomeric form.

Cyclin Binding and Conformational Changes

The binding of to CDK1, forming the maturation promoting factor (MPF), is mediated by the hydrophobic cyclin box motif within , which engages the αC-helix (containing the PSTAIRE sequence) of CDK1 through extensive hydrophobic interactions. This interface, encompassing approximately 1,200 Ų of buried surface area, positions key residues such as Phe153 of CDK1's activation segment into a hydrophobic groove on , stabilizing the complex. The interaction induces a and repositioning of CDK1's T-loop (the activation segment spanning residues 145–171), displacing it from the catalytic cleft and relieving steric hindrance that blocks access in the monomeric CDK1. This cyclin B binding triggers a profound conformational shift in CDK1, reorienting the αC-helix and displacing the DFG motif (Asp-Phe-Gly) at the start of the activation segment to form a short β-hairpin . The shift stabilizes an open conformation of the ATP-binding , enhancing affinity and enabling proper alignment of catalytic residues. Overall, these changes prime CDK1 for activation by exposing the Thr161 phosphorylation to CDK-activating (CAK), although the unphosphorylated CDK1-cyclin B complex retains low kinase activity. Allosterically, cyclin B binding exposes the Thr161 residue in CDK1's activation segment, positioning it for phosphorylation by CDK-activating kinase (CAK), which inserts into a solvent-filled cleft created by the conformational rearrangement. This exposure is critical, as the T-loop in free CDK1 occludes the site, preventing efficient CAK access. The resulting Thr161 phosphorylation further rigidifies the activation segment, increasing complex stability. Insights into these dynamics derive from 1990s X-ray crystal structures, such as that of the CDK2-cyclin A complex at 2.9 resolution, which showed cyclin binding causing a ~20° rotation between the N- and C-terminal lobes of the kinase, opening the active site cleft—a conserved mechanism extrapolated to MPF given the structural between CDK1-cyclin B and CDK2-cyclin A. Later structures of CDK1-cyclin B confirmed similar lobe reorientation and interface features, albeit with a smaller binding surface due to CDK1-specific twists in the C-helix.

Regulation of Activity

Activation Pathways

The activation of maturation promoting factor (MPF), composed of and (CDK1), involves a series of tightly regulated post-translational modifications that culminate in full enzymatic activity. Upon binding of to CDK1, the complex undergoes initial by CDK-activating kinase (CAK), which adds a phosphate group to threonine 161 (Thr161) on the activation loop (T-loop) of CDK1, enhancing its catalytic competence and affinity. This activating is essential but insufficient on its own, as the cyclin B-CDK1 complex remains inactive due to inhibitory phosphorylations at threonine 14 (Thr14) and tyrosine 15 (Tyr15), introduced by Wee1 and Myt1 kinases during to prevent premature mitotic entry. Full activation requires the removal of these inhibitory phosphates by members of the phosphatase family, particularly Cdc25B and Cdc25C, which dephosphorylate Thr14 and Tyr15 in a stepwise manner at the G2/M transition. This dephosphorylation is triggered by upstream signals, such as DNA damage checkpoints being satisfied, and initiates a powerful loop: newly activated MPF phosphorylates and activates phosphatases while simultaneously inhibiting Wee1 kinase activity, thereby accelerating the removal of inhibitory phosphates from additional cyclin B-CDK1 molecules and amplifying MPF activity exponentially. This feedback mechanism ensures rapid and irreversible commitment to mitosis once initiated. Spatial regulation further refines MPF by controlling the subcellular localization of cyclin B-CDK1. During , cyclin B contains a () that promotes its cytoplasmic retention, preventing untimely nuclear access and maintaining low nuclear MPF activity; at the onset of G2/M, of the by CDK1 masks it, facilitating nuclear import of the complex and enabling targeted within the . The overall process operates via a , where MPF activity remains low until levels or initial signals surpass a critical , after which the drives an abrupt, all-or-nothing surge in activity to ensure synchronized mitotic entry across the . This bistable switch-like , modeled mathematically in early studies, integrates multiple to safeguard against partial or erroneous .

Inactivation Mechanisms

The inactivation of maturation promoting factor (MPF), composed of and (CDK1), is essential for mitotic exit and preventing premature re-entry into the . The primary mechanism involves ubiquitin-mediated proteasomal degradation of , orchestrated by the anaphase-promoting complex/cyclosome (APC/C), a multi-subunit . This process targets the N-terminal destruction box motif in , marking it for polyubiquitination and subsequent rapid breakdown by the 26S , which dissociates from CDK1 and thereby abolishes MPF activity. The APC/C remains inactive during early to allow progression but is activated precisely at the metaphase-anaphase transition to initiate this degradation. Activation of the for cyclin B degradation requires its co-activator Cdc20, which binds directly to the and enhances its ubiquitin ligase activity toward and other substrates. Cdc20 recruitment is tightly regulated by the spindle assembly (SAC), a surveillance that inhibits ^Cdc20 until all chromosomes achieve proper bipolar attachment to the mitotic spindle. Upon SAC satisfaction, unattached kinetochores cease generating inhibitory signals (involving Mad1, Mad2, and BubR1 proteins that sequester Cdc20 into the mitotic checkpoint complex), allowing free Cdc20 to activate the and trigger ubiquitination after alignment. This ensures timely onset and mitotic progression without errors in chromosome . Following cyclin B degradation, a secondary inhibitory mechanism reinforces MPF inactivation through re-phosphorylation of CDK1 on inhibitory sites Thr14 and Tyr15. This phosphorylation is mediated by the kinases Myt1 (which targets both residues) and Wee1 (primarily Tyr15), whose activities increase as CDK1 levels drop and counterbalance the phosphatase during the M-to-G1 transition. Such post-degradation phosphorylation prevents residual CDK1 activity from reactivating prematurely, stabilizing the G1 state and contributing to irreversible mitotic exit in systems like egg extracts. Inhibition of Wee1/Myt1 allows rapid re-entry into mitosis even after prolonged G1 arrest, underscoring their role in maintaining low CDK1 activity. The oscillatory nature of MPF activity is restored in the subsequent cell cycle through de novo synthesis of cyclin B during G2 phase, which binds free CDK1 to reform MPF and initiate the next round of activation. This resynthesis, coupled with initial inhibitory phosphorylations on CDK1, sets up the periodic peaks and troughs of MPF characteristic of cell cycle progression in early embryonic divisions.

Role in the Cell Cycle

Entry into Mitosis

The entry into mitosis, or the G2/M transition, is primarily triggered by the accumulation of active maturation promoting factor (MPF), a complex of cyclin B and cyclin-dependent kinase 1 (CDK1), reaching a critical threshold that drives the cell into prophase. This threshold ensures that once surpassed, MPF rapidly phosphorylates numerous substrates—estimated at over 100 proteins involved in mitotic restructuring—initiating key events such as nuclear envelope breakdown and chromosome condensation precursors. The process is facilitated by positive feedback loops where active MPF further activates itself by promoting the dephosphorylation of inhibitory sites on CDK1 via Cdc25 phosphatases, amplifying the signal for irreversible commitment to mitosis. To safeguard genomic integrity, the G2/M transition integrates checkpoints that delay MPF activation in response to DNA damage or incomplete replication. Specifically, and ATR kinases detect such stresses and phosphorylate downstream effectors like Chk1 and Chk2, which in turn inhibit phosphatases by promoting their sequestration or degradation, thereby preventing the removal of inhibitory phosphates from CDK1 and blocking MPF maturation. This mechanism ensures that cells do not proceed to with unrepaired DNA, with experimental evidence from mammalian cells showing that overriding inhibition leads to checkpoint failure and premature mitotic entry. In mammalian oocytes, MPF plays a specialized role in regulating meiotic , particularly through interactions with the kinase pathway. Fully grown oocytes are held in I arrest until hormonal signals trigger MPF activation for meiotic resumption, but post-ovulation, MPF activity is modulated by , which activates MAPK to stabilize levels and maintain II arrest until fertilization. This pathway highlights species-specific adaptations, as knockout in mice results in spontaneous meiotic resumption and loss of , underscoring MPF's integration with upstream regulators for oocyte competence. Quantitatively, MPF activation is modeled as a bistable switch, where the system exhibits two stable states—low () and high () activity—separated by an unstable , ensuring a sharp, all-or-nothing transition. Pioneering computational models by Tyson and colleagues demonstrate that mutual activation (via ) and inhibition (via Wee1) loops create , making the G2/M commitment irreversible even if levels slightly decline, as validated in egg extracts and fission yeast. This framework explains the rapid onset of observed experimentally, with switch-like behavior confirmed across eukaryotic systems.

Progression Through Mitotic Phases

During , maturation promoting factor (MPF), the cyclin B1-CDK1 complex, maintains high kinase activity to facilitate the formation and stabilization of kinetochore- attachments. This sustained activity is essential as chromosomes initially attach laterally or via error-prone end-on connections to the assembling mitotic spindle; MPF phosphorylates kinetochore-associated proteins such as CLASP2, in coordination with (Plk1), to create a phospho-switch that promotes the transition to stable end-on attachments. Additionally, B1 localizes to unattached kinetochores, where it contributes to efficient microtubule capture and attachment by locally enhancing CDK1 activity, thereby correcting initial errors and ensuring bipolar orientation. In , MPF's plateaued activity supports congression to the equator by phosphorylating motor proteins and regulators that balance pulling and ejection forces. For instance, MPF phosphorylates the kinesin-like protein CENP-E at its C-terminal tail, relieving auto-inhibition and restoring its plus-end-directed motility to transport toward the equator while counteracting poleward forces from and polar ejection forces along arms generated by chromokinesins. This phosphorylation-mediated regulation ensures oscillatory movements that refine alignment, with MPF also targeting microtubule-associated proteins to modulate dynamics and maintain tension across sister kinetochores. The checkpoint (SAC) indirectly regulates MPF activity through Mad2, which, upon unresolved kinetochore-microtubule attachments, forms a mitotic checkpoint complex that inhibits the -promoting complex/cyclosome (APC/C), thereby preventing cyclin B1 ubiquitination and sustaining high MPF levels to delay onset. This checkpoint override mechanism ensures progression only after bi-orientation is achieved, as persistent Mad2 signaling at unattached kinetochores blocks MPF inactivation. MPF's temporal profile during these phases features a sharp increase through , peaking in due to loops involving phosphatases, followed by a plateau through until SAC satisfaction triggers APC/C activation.

Exit from Mitosis

The exit from is critically dependent on the inactivation of maturation promoting factor (MPF), a B-CDK1 complex, which triggers a cascade of events leading to chromosome decondensation, reformation, and completion. At the onset of , partial inactivation of MPF removes its inhibitory on separase, thereby allowing separase and subsequent of subunits that hold together. This process ensures the physical separation of chromosomes, marking the transition from to . During , the full reversal of mitotic states is mediated by protein phosphatase 2A (PP2A), particularly the B55 regulatory subunit variant (PP2A-B55), which numerous MPF-phosphorylated substrates to restore the cellular architecture. Activated upon MPF decline, PP2A-B55 targets over 2,900 phosphosites, including those on proteins involved in spindle disassembly and reassembly, such as NUP153 and PRC1, with kinetics determined by substrate-specific motifs that prioritize rapid of key regulators. This coordinated prevents premature progression and ensures orderly mitotic exit. The anaphase-promoting complex/cyclosome (APC/C) contributes to MPF inactivation through ubiquitination and degradation. The decline in MPF activity also coordinates by enabling the assembly of the central and contractile ring ingression. High MPF levels during phosphorylate components of the central machinery, such as PRC1 and ECT2, inhibiting their localization and function; upon MPF inactivation via degradation, dephosphorylation of these targets allows bundling of antiparallel at the midzone and recruitment of RhoA-GEFs to drive actin-myosin contractility for furrow formation and progression. This temporal control ensures initiates only after chromosome segregation, avoiding errors in daughter cell partitioning. Finally, the low MPF levels post-mitosis provide negative feedback to establish the by permitting dephosphorylation of the (), which in its hypophosphorylated form binds and represses transcription factors to silence genes required for S-phase entry. Rb remains hyperphosphorylated from earlier phases during ; the subsequent drop in CDK1 activity allows phosphatases like PP1 to reverse this, reactivating Rb to maintain E2F repression and enforce a G1 checkpoint until mitogenic signals reinitiate the cycle. This mechanism links mitotic completion to control, preventing unscheduled .

Physiological Functions and Targets

Nuclear Envelope Breakdown

Maturation promoting factor (MPF), a complex of and (CDK1), plays a central role in initiating breakdown (NEBD) at the onset of by directly phosphorylating components of the . This process disassembles the nuclear barrier, enabling the mitotic spindle to access chromosomes during . The primary targets of MPF are the B-type lamins, particularly B1 and B2, which form the structural meshwork of the . MPF phosphorylates B1 at serine 23 in the N-terminal head domain and serine 393 in the C-terminal tail domain, with sites on B2 including serine 17 (N-terminal), 34, serine 37, and serine 405. These events, occurring rapidly upon MPF activation, lead to of filaments. The mechanism involves disruption of head-to-tail polymer interactions within the coiled-coil domains, which destabilizes the filamentous network and solubilizes the lamina into non-sedimentable forms. The disassembly of the has critical consequences for mitotic progression, as it fragments the into vesicles and allows from the forming to interact directly with . This facilitates proper chromosome alignment and . In , NEBD is reversed through of these sites by protein phosphatases such as PP1 and PP2A, which restores and promotes reassembly around daughter nuclei. Experimental evidence for MPF's role in NEBD comes from microinjection studies in Xenopus laevis embryos. Injection of purified MPF into cycloheximide-arrested embryos, which contain multiple nuclei in a shared , induces rapid nuclear envelope dispersal within 5 minutes, as visualized by staining of the . This breakdown occurs at MPF concentrations similar to those active during maturation, confirming MPF's sufficiency to trigger lamina disassembly independently of other events.

Chromosome Condensation

Maturation promoting factor (MPF), the cyclin B-CDK1 complex, plays a central role in initiating chromosome condensation during the entry into by phosphorylating key substrates that drive compaction. Among its primary targets are the structural maintenance of chromosomes (SMC) subunits SMC2 and SMC4 of the complexes, which form of condensin I and II. Phosphorylation of these subunits by MPF enhances the DNA-binding affinity and activity of condensins, enabling them to organize into higher-order looped structures that facilitate axial shortening and compaction of chromosomes. In parallel, of at serine 10 (Ser10), primarily by Aurora B kinase upon MPF activation, correlates closely with mitotic chromosome condensation and supports the structural changes necessary for proper packing. This aids in the dissociation of proteins and promotes the compaction of nucleosomes, working in concert with -mediated looping to achieve the rod-like morphology of mitotic chromosomes. The mechanism involves condensin activation, where phosphorylated SMC2/SMC4 subunits loop DNA segments into stable scaffolds, while H3 Ser10 further stabilizes these interactions by altering accessibility and rigidity. Chromosome condensation begins in upon MPF activation and progresses to completion by , coinciding with breakdown. This timing ensures that chromosomes are sufficiently compacted for alignment on the plate. The process requires synergy with Aurora B kinase, which reinforces H3 Ser10 and recruitment, amplifying MPF's effects to achieve full condensation. Defects in MPF-mediated , such as hypo- of condensins or Ser10, disrupt proper compaction and lead to the formation of bridges, where under-condensed trails between segregating daughter cells, resulting in genomic instability and segregation errors.

Spindle Assembly and

Maturation promoting factor (MPF), consisting of cyclin B1-bound Cdk1, plays a pivotal role in orchestrating assembly by phosphorylating key regulators of at kinetochores. Specifically, MPF phosphorylates targeting protein for Xklp2 (TPX2) at threonine 72, which enhances TPX2's ability to activate Aurora A kinase and promote and stabilization around chromosomes, facilitating the formation of a robust bipolar . Through these and other phosphorylations, MPF modulates dynamics to stabilize the bipolar . By phosphorylating stathmin/OP18, MPF inhibits its catastrophe-promoting activity, thereby suppressing depolymerization events and favoring the persistence of essential for capture and alignment. This stabilization is critical during , where sustained MPF activity maintains integrity against dynamic instabilities. As MPF activity declines toward , dephosphorylation events allow astral to elongate, enabling repositioning and orientation within the to specify the division plane. In , MPF coordinates the activation of the contractile ring via the ECT2/RhoA pathway. During early , MPF phosphorylates the Rho ECT2, initially sequestering it to prevent premature RhoA activation and contractile ring formation. The subsequent decline in MPF activity at onset releases this inhibition, allowing ECT2 to localize to the central , activate RhoA, and drive II recruitment for equatorial furrow ingression and contractile ring assembly. The spindle assembly checkpoint (SAC) further integrates MPF regulation, where unattached kinetochores generate a feedback signal via Aurora B kinase to sustain MPF levels. Aurora B, activated at error-prone attachments, phosphorylates kinetochore substrates to destabilize incorrect bindings, thereby maintaining SAC signaling that inhibits the anaphase-promoting complex/cyclosome (APC/C) and prevents B1 degradation, ensuring MPF persistence until all kinetochores achieve stable attachments.

Clinical and Research Implications

Dysregulation in Cancer

Dysregulation of maturation promoting factor (MPF), composed of B1 and CDK1, plays a significant role in tumorigenesis by disrupting normal control. Overexpression of B1, a key component of MPF, occurs in approximately 50% of solid tumors, with median prevalence rates around 48.78% across various studies. This upregulation is particularly noted in (about 42% of cases) and non-small cell (up to 58% in some cohorts), where it correlates with aggressive disease and poor , including reduced 3-year overall survival (odds ratio [OR] = 2.05, 95% CI = 1.20–3.50) and 5-year overall survival (OR = 2.11, 95% CI = 1.33–3.36). In , high B1 levels are an independent prognostic factor associated with tumor aggressiveness. Similarly, in , B1 overexpression predicts worse survival outcomes (3-year OS OR = 2.95, 95% CI = 1.60–5.44; 5-year OS OR = 2.60, 95% CI = 1.46–4.63). Mutations and pathway alterations leading to CDK1 hyperactivation further contribute to cancer progression and therapy resistance. of p53 function, common in over 50% of human cancers, impairs the G1/S checkpoint, causing cells to depend heavily on the /M checkpoint regulated by Wee1-mediated inhibitory phosphorylation of CDK1. This reliance promotes chemoresistance, as p53-deficient tumors evade DNA damage-induced arrest. Inhibition of Wee1 removes this brake, resulting in CDK1 hyperactivation and premature mitotic entry, which selectively sensitizes p53-mutant cells to by inducing . For instance, Wee1 inhibitors overcome cisplatin resistance in high-risk neuroblastoma models with p53 pathway defects. Therapeutic strategies targeting MPF dysregulation have shown promise in preclinical models. Selective CDK1 inhibitors, such as RO-3306, an ATP-competitive agent, exhibit potent anti-tumor activity by inducing /M arrest and in various cancers, including and . RO-3306 has demonstrated efficacy in models when combined with , reducing tumor growth through blockade of CDK1/PDK1/β-catenin signaling. Additionally, combining CDK1 inhibition with modulation of the anaphase-promoting complex/cyclosome (APC/C), which degrades to inactivate MPF, enhances therapeutic outcomes; APC/C activators like CDH1 promote destruction, synergizing with CDK1 inhibitors to enforce exit in tumor cells. Recent research up to 2025 highlights MPF as a synthetic lethal target in -mutant cancers via screens. Genome-wide / screens have identified vulnerabilities in the MPF pathway, where loss of sensitizes cells to perturbations in CDK1 or B1, leading to selective lethality. For example, dual inhibitors targeting A/B interactions (key for MPF activation) selectively kill cells with high activity and G1/S checkpoint defects, common in -mutant tumors. These findings underscore MPF's role in post-2000 studies of tumorigenesis and support its exploitation for precision therapies in -deficient malignancies.

Therapeutic Targeting

Direct inhibitors of maturation promoting factor (MPF), primarily targeting the CDK1 subunit, have been developed to disrupt uncontrolled in malignancies. Dinaciclib, an ATP-competitive inhibitor with activity against CDK1, CDK2, CDK5, and CDK9, has demonstrated encouraging single-agent activity in (CLL), a form of , in Phase II clinical trials conducted in the and reported through the 2020s. In these trials, dinaciclib achieved an objective response rate of approximately 58% in relapsed or refractory CLL patients, with manageable toxicity profiles including cytopenias. Purvalanol A, another ATP-competitive CDK1 blocker, has shown preclinical efficacy in pediatric (AML) models, particularly when combined with anti-apoptotic agents like ABT-737 to enhance in leukemic blasts. Indirect modulation of MPF activity through upstream regulators represents another therapeutic avenue, especially in hematologic cancers. Polo-like kinase 1 (PLK1) inhibitors, such as volasertib, interfere with the loop that activates MPF by phosphorylating phosphatases and inhibiting . Volasertib received orphan drug designation in the and for AML treatment and was evaluated in a Phase III trial for patients ineligible for standard induction , where it improved overall survival when added to low-dose cytarabine compared to cytarabine alone, though the trial did not meet its primary endpoint due to toxicity concerns like . Emerging approaches leverage proteolysis-targeting chimeras (PROTACs) to induce ubiquitin-mediated degradation of CDK1 or , bypassing the limitations of reversible inhibition. PROTACs such as those based on CDK1 ligands conjugated to E3 ligase recruiters (e.g., binders) have effectively degraded CDK1 in cell lines, leading to G2/M arrest and reduced proliferation without affecting non-dividing cells. These degraders offer enhanced selectivity and duration of action compared to small-molecule inhibitors. In reproductive contexts, nanoparticle-based delivery systems, including nanoparticles, have been employed to augment oocyte maturation and embryonic development in models by stabilizing MPF activity post-activation, improving formation rates in artificially activated oocytes. Key challenges in MPF-targeted therapies include achieving isoform-specific inhibition of CDK1 amid high structural homology with other CDKs in the ATP-binding pocket, which often results in off-target effects and dose-limiting toxicities like gastrointestinal issues and myelosuppression. Ongoing research addresses this through allosteric inhibitors and cyclin-specific degraders to minimize pan-CDK activity. As of November 2025, no selective CDK1 inhibitors have been approved for clinical use, with efforts focusing on combination therapies to improve efficacy and safety. Additionally, investigations into MPF dysregulation in non-oncologic conditions, such as CDK1-mediated tau hyperphosphorylation contributing to neurodegeneration in Alzheimer's disease, highlight potential broader applications, though clinical translation remains exploratory.

References

  1. [1]
    Entry into mitosis: a solution to the decades-long enigma of MPF - NIH
    Maturation or M phase-promoting factor (MPF) is the universal inducer of M phase common to eukaryotic cells. MPF was originally defined as a transferable ...
  2. [2]
  3. [3]
    MPF-based meiotic cell cycle control: Half a century of lessons from ...
    The discovery and characterization of MPF derived from oocyte maturation was a major contributor to the development of this paradigm for cell cycle control ...
  4. [4]
  5. [5]
  6. [6]
  7. [7]
    From oocyte maturation to the in vitro cell cycle - PubMed
    This article briefly reviews the classical cell cycle studies using oocytes and zygotes of mainly amphibians in the past century.
  8. [8]
    Cytoplasmic control of nuclear behavior during meiotic ... - PubMed
    Cytoplasmic control of nuclear behavior during meiotic maturation of frog oocytes. ... Authors. Y Masui, C L Markert. PMID: 5106340; DOI: 10.1002/jez.1401770202.
  9. [9]
    [PDF] Masui Y & Markert C L. Cytoplasmic. control of nuclear be&ior during ...
    maturation of frog oocytes. J. Expo Zoof. 177:129-45, 1971. [Department of Biology, Yale University, New Haven, CT].Missing: Hiroko | Show results with:Hiroko
  10. [10]
    Partial purification and characterization of the maturation-promoting ...
    A quantitative in vivo assay of MPF is described and a unit of MPF activity is defined as that amount causing a 50% maturation frequency when oocytes are ...
  11. [11]
    Progesterone-stimulated meiotic cell division in Xenopus oocytes ...
    ... maturation promoting factor which upon transfer by micropipette to a ... protease- sensitive (12). However, a direct biochemical effect of MPF ...
  12. [12]
    The Xenopus cdc2 protein is a component of MPF, a ... - PubMed - NIH
    Jul 29, 1988 · The Xenopus cdc2 protein is a component of MPF, a cytoplasmic regulator of mitosis. Cell. 1988 Jul 29;54(3):423-31. ... MPF. Publication types.Missing: seminal paper Paul Nurse
  13. [13]
    Universal and essential role of MPF/cdc2+ - PubMed
    Universal and essential role of MPF/cdc2+. Nature. 1988 Dec 1;336(6198):430. doi: 10.1038/336430a0. Authors. O Niwa, M Yanagida.
  14. [14]
    MPF from starfish oocytes at first meiotic metaphase is a heterodimer ...
    MPF from starfish oocytes at first meiotic metaphase is a heterodimer containing one molecule of cdc2 and one molecule of cyclin B. · Abstract · Full text · Images ...
  15. [15]
    Cyclin-dependent protein kinases and cell cycle regulation ... - Nature
    Jan 13, 2025 · In the context of cancer CDK1, by phosphorylating multiple factors, plays a major role in promoting cell transformation and tumorigenesis.
  16. [16]
    G2/mitotic-specific cyclin-B1 - Homo sapiens (Human) | UniProtKB
    Interacts with the CDC2 protein kinase to form a serine/threonine kinase holoenzyme complex also known as maturation promoting factor (MPF).
  17. [17]
    CDK1 structures reveal conserved and unique features of ... - Nature
    Apr 13, 2015 · Relative to CDK2–cyclin A, CDK1–cyclin B is less thermally stable, has a smaller interfacial surface, is more susceptible to activation segment ...Missing: unstable | Show results with:unstable
  18. [18]
    Cell cycle-dependent binding between Cyclin B1 and Cdk1 ... - NIH
    Jun 29, 2022 · Although Cyclin B1 and Cdk1 are known to bind with high affinity, our results reveal that in living cells there is a pool of Cyclin B1 that is not bound to Cdk ...
  19. [19]
    The Cyclin-dependent kinase 1: more than a cell cycle regulator - NIH
    Oct 28, 2023 · The activity of CDK1 is modulated by its binding to cyclin B1 and by its phosphorylation on crucial residues. Specifically, during the late G2 ...Missing: box | Show results with:box
  20. [20]
    Why cyclin Y? A highly conserved cyclin with essential functions - PMC
    More specifically, all cyclins are related by having a domain called the cyclin box, which mediates binding to Cdks. Likewise, many Cdks have been ...
  21. [21]
    Differences in the Conformational Energy Landscape of CDK1 ... - NIH
    Nov 21, 2018 · To prepare the ternary CDK1–cyclin B-Cks2 complex, components were individually purified and then mixed in a molar ratio of 1:1.5:2, CDK1:cyclin ...
  22. [22]
    Thr-161 Phosphorylation of Monomeric Cdc2 - ScienceDirect.com
    Activation of Cdc2 depends on two key events: cyclin binding and an activating phosphorylation on Thr-161 residue located in the T-loop.
  23. [23]
    Spatiotemporal regulations of Wee1 at the G2/M transition
    Jan 13, 2011 · Wee1 is a protein kinase that negatively regulates mitotic entry in G2 phase by suppressing cyclin B–Cdc2 activity, but its spatiotemporal ...
  24. [24]
    The G2-to-M transition from a phosphatase perspective - Cell Division
    May 25, 2020 · At the end of the G2-phase, new Cyclin B synthesis is not required anymore, all components needed for mitosis are present but MPF is inactive.<|control11|><|separator|>
  25. [25]
    A quantitative analysis of the kinetics of the G2 DNA damage ... - PNAS
    I propose a general answer at this point: an antagonistic interaction between MPF and Wee1 exists (Wee1 inhibits MPF activity by tyrosine phosphorylation while ...The Mpf Subsystem · The Cdc25 Subsystem · The Wee1 Subsystem
  26. [26]
    CDK1 structures reveal conserved and unique features of the ... - NIH
    Apr 13, 2015 · The character of the CDK1 active site cleft beyond the ATP phosphate-binding site is shaped by residues 40–46 that precede the start of the ...Cdk1--Cyclin B And... · Protein Expression And... · Cdk Kinase AssaysMissing: PSTAIRE | Show results with:PSTAIRE
  27. [27]
    Cyclin-dependent kinases | Genome Biology | Full Text
    Jun 30, 2014 · For instance, Cdk1 is thought to be activated mainly by A- and B-type cyclins but can also bind to, and be activated by, D- or E-type cyclins in ...
  28. [28]
    Inhibitory Phosphorylation of Cyclin-Dependent Kinase 1 as a ... - NIH
    Before mitosis, cyclin B1-CDK1 complexes are kept in a CDK1Thr14/Tyr15-phosphorylated and inactive state by two kinases called WEE1 and MYT1. While WEE1 ...Missing: Asp128 catalysis
  29. [29]
    Cyclin-dependent kinase 1 (Cdk1) is essential for cell division and ...
    Cdk1 was the first Cdk identified (4, 5), is conserved in all organisms, plays important roles in mitosis, and can drive S phase in the absence of Cdk2 (6).
  30. [30]
    Mechanism of CDK activation revealed by the structure of a cyclinA-CDK2 complex - Nature
    ###Summary of Conformational Changes Upon Cyclin A Binding to CDK2
  31. [31]
    Distinct Activation Pathways Confer Cyclin Binding Specificity ... - NIH
    ... <1,000-fold), possibly due to the instability of Cdk1/cyclin B ... The effect of phosphorylation by CAK on cyclin binding by CDC2 and CDK2. Mol ...
  32. [32]
    Identification of Candidate Cyclin-dependent kinase 1 (Cdk1 ...
    In interphase, Cdk1 is inactivated through phosphorylation on threonine 14 (Thr14) and tyrosine 15 (Tyr15) by the dual specificity protein kinases Myt1 and Wee1 ...
  33. [33]
    Regulatory roles of cyclin dependent kinase phosphorylation in cell ...
    In interphase, newly synthesized Cyclin B interacts with Cdk1, followed subsequently by Wee1 kinase family-mediated phosphorylation at Thr14 and Tyr15 of Cdk1 ( ...
  34. [34]
    feedback and redundancy in the mitotic entry network | Journal of ...
    Apr 13, 2009 · In human cells, high cyclin B levels are temporally restricted to G2 phase and early mitosis by regulated transcription and protein degradation.
  35. [35]
    Triggering the all-or-nothing switch into mitosis - PMC - NIH
    A switch-like mechanism integrates signals of cellular status and commits the cell to mitosis by abruptly removing inhibitory phosphate from preformed cyclin: ...
  36. [36]
    Nuclear export of cyclin B1 and its possible role in the DNA damage ...
    The activity of MPF is regulated by phosphorylation/dephosphorylation of cdc2 and accumulation of cyclin B protein. During S and G2 phases, the B‐type cyclins ...
  37. [37]
  38. [38]
  39. [39]
    The Cyclin-dependent kinase 1: more than a cell cycle regulator
    Oct 28, 2023 · Despite the expected inhibitory effect on CDK1 activity, the phosphorylation of Tyr15 has been found to be increased in several cancer types ...
  40. [40]
    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.
  41. [41]
    Regulating mammalian checkpoints through Cdc25 inactivation - PMC
    This review focuses on our understanding of the biochemical mechanisms that specifically inactivate Cdc25 (cell division cycle 25) phosphatases to achieve this.
  42. [42]
    Mos in the Oocyte: How to Use MAPK Independently of Growth ... - NIH
    Mos would serve to activate MPF for entry into the first meiotic division, but also to stabilize MPF during the second meiotic division, inducing the arrest of ...
  43. [43]
    Maturation-Promoting Factor Governs Mitogen-Activated Protein ...
    Resumption of meiosis in oocytes is associated with activation of maturation-promoting factor (MPF) and mitogen-activated protein kinase (MAPK). The activity of ...Missing: collaboration | Show results with:collaboration
  44. [44]
    A model for a bistable biochemical trigger of mitosis - PubMed
    The activation of maturation promoting factor (MPF, cyclin B/Cdc2), which starts mitosis, is modeled as a bistable biochemical switch or trigger.
  45. [45]
    lessons from modeling regulation of the eukaryotic cell cycle - PMC
    Jul 1, 2015 · Our prediction that MPF activation is governed by a bistable switch differs radically from the simple interpretation of Solomon's observations ...
  46. [46]
    The bistable mitotic switch in fission yeast
    May 6, 2024 · “Bistability” of the mitotic switch means that the cyclin threshold for mitotic entry is larger than the threshold for mitotic exit; for cyclin ...
  47. [47]
    Cdk1 and Plk1 mediate a CLASP2 phospho-switch that stabilizes ...
    Oct 8, 2012 · Cdk1 phosphorylation of CLASP2 promotes Plk1 recruitment to kinetochores and is required for stabilization of kinetochore–microtubule ...Missing: MPF | Show results with:MPF
  48. [48]
    Cyclin B1 is localized to unattached kinetochores and contributes to ...
    Jan 15, 2008 · We conclude that cyclin B1 accumulates at kinetochores during prometaphase, where it contributes to the correct attachment of microtubules to kinetochores and ...Missing: MPF | Show results with:MPF
  49. [49]
    Mechanisms of Chromosome Congression during Mitosis - PMC
    This favors the balanced action of kinetochore pulling forces and polar ejection forces along chromosome arms that drive chromosome oscillatory movements during ...
  50. [50]
  51. [51]
    An overview of Cdk1-controlled targets and processes | Cell Division
    May 13, 2010 · In eukaryotic cells, the cell cycle is controlled by cyclin dependent kinases (CDKs). Six conserved CDKs exist in the budding yeast S.Missing: across original
  52. [52]
    Mitotic progression becomes irreversible in prometaphase and ... - NIH
    Cdk1 activity increases sharply during prophase and prometaphase. It is well established that the activity of Cdk1/cyclin B complex is low in interphase and ...Missing: profile | Show results with:profile
  53. [53]
  54. [54]
    A New Model for Nuclear Envelope Breakdown - PMC
    The process of nuclear envelope breakdown is triggered by active maturation-promoting factor (MPF), which is thought to be a complex of cyclin B and cdc2/cdk1 ...
  55. [55]
    Intermediate, but not average: The unusual lives of the nuclear lamin ...
    Lamina disassembly is achieved by phosphorylation of sites flanking the central coiled coil by Cdk1 and other kinases; these phospho-modifications disrupt head- ...
  56. [56]
    Novel Mode of Phosphorylation-triggered Reorganization of the ...
    The CDK1 phosphorylation sites of the lamin types are part of conserved sequence stretches. For lamin B, the respective phosphorylated residues are Ser23 ...
  57. [57]
    Phosphorylation of lamins determine their structural properties and ...
    Cyclin dependent kinase 1 (Cdk1; also known as cdc2) has been identified to directly phosphorylate both A- and B-type lamins during mitosis. Phosphorylation of ...Missing: Ser416 | Show results with:Ser416
  58. [58]
    Dephosphorylation in nuclear reassembly after mitosis - PMC
    Oct 4, 2022 · Nuclear Reassembly (NR) at the end of open mitosis requires the dephosphorylation of several targets. At least two groups of serine/threonine ...
  59. [59]
    PP2A-B55 promotes nuclear envelope reformation after mitosis in ...
    Our results suggest that PP2A-Tws dephosphorylates multiple proteins, including BAF and Lamin, to promote the reassembly of nuclei after mitosis. Results. A ...
  60. [60]
    Maturation-promoting factor induces nuclear envelope breakdown in ...
    We have studied the effect of maturation-promoting factor (MPF) on embryonic nuclei during the early cleavage stage of Xenopus laevis development.Missing: Ser22 Ser416
  61. [61]
    The initial phase of chromosome condensation requires Cdk1 ...
    Apr 15, 2011 · Here we report that mitotic chromosome assembly in prophase depends on Cdk1-mediated phosphorylation of the condensin II complex.
  62. [62]
    Cell cycle regulation of condensin Smc4 - PMC - NIH
    Budding yeast condensin is composed of an Smc2-Smc4 ... The initial phase of chromosome condensation requires Cdk1-mediated phosphorylation of the CAP-D3 subunit ...
  63. [63]
    Mitotic kinase anchoring proteins: the navigators of cell division - PMC
    Many substrates have been reported for CDK1-Cyclin B during this transition, including histones, whose phosphorylation by CDK1 promotes chromosome condensation, ...<|control11|><|separator|>
  64. [64]
  65. [65]
    Condensins: Organizing and Segregating the Genome - ScienceDirect
    (A) SMC2 and SMC4 form the core of the condensin complexes. Each SMC ... Upon NEBD, cyclin B–Cdk1 phosphorylates and activates condensin I [25, 27] ...
  66. [66]
    Phosphorylation of histone H3 on Ser-10 by Aurora B is essential for ...
    Feb 20, 2017 · Phosphorylation of histone H3 on Ser-10 (H3S10ph) is involved in regulating mitotic chromosome condensation and decondensation, ...
  67. [67]
    Condensin complexes regulate mitotic progression and interphase ...
    Feb 22, 2010 · In mammalian somatic cells, depletion of condensins I and II results in an increased frequency of defects during mitotic anaphase, including ...
  68. [68]
    Phosphorylation of Targeting Protein for Xenopus Kinesin-like ...
    We find that Cdk1/2 phosphorylate TPX2 in vitro and in vivo. Using homemade antibodies specific for TPX2 phosphorylated at Thr72, we show that this ...Missing: MPF | Show results with:MPF
  69. [69]
    Self-organization of intracellular gradients during mitosis
    Jan 29, 2010 · Phosphorylation of tubulin-bound OP18/stathmin increases free tubulin, inhibits its ability to induce microtubule catastrophe (promoting ...
  70. [70]
    Changes in Ect2 Localization Couple Actomyosin-Dependent Cell ...
    Aug 13, 2012 · Here we identify the Cdk1 substrate, Ect2, as a central regulator of mitotic rounding, thus uncovering a link between the cell-cycle machinery ...
  71. [71]
    An ECT2–centralspindlin complex regulates the localization and ...
    Aug 15, 2005 · Here, we dissect furrow formation in human cells and show that the RhoGEF ECT2 is required for cortical localization of RhoA and contractile ring assembly.
  72. [72]
    The Aurora B Kinase in Chromosome Bi-Orientation and ... - Frontiers
    Two feedback mechanisms control the process of kinetochore–microtubule attachment during mitosis, and Aurora B contributes decisively to both of them. These ...
  73. [73]
    Prognostic role of cyclin B1 in solid tumors: a meta-analysis - NIH
    Results showed that cyclin B1 overexpression was associated with worse 3-year OS of solid tumors (OR = 2.05, 95% CI = 1.20 to 3.50, P = 0.009) (Figure 2).
  74. [74]
    High cyclin B1 expression is associated with poor survival in breast ...
    This study suggests high cyclin B1 associates with aggressive phenotype and is an independent prognostic factor in breast cancer.
  75. [75]
    Targeting WEE1 kinase as a p53-independent therapeutic strategy ...
    Sep 15, 2023 · Dysregulated p53 most often enables cells to circumvent the G1-S cell cycle checkpoint due to the loss of p53 target gene CDKN1A (encoding ...
  76. [76]
    Wee1 Kinase: A Potential Target to Overcome Tumor Resistance to ...
    Moreover, some data suggest that cells with dysfunctional p53 are more sensitive to Wee1 inhibition combined with conventional chemotherapy than those with ...
  77. [77]
    Wee-1 Kinase Inhibition Overcomes Cisplatin Resistance ...
    Decreased cisplatin sensitivity associated with these high-risk mutations is driven by their inability to undergo cellular senescence, the primary response for ...Clonogenic Survival Assay · Orthotopic Mouse Model Of... · Results
  78. [78]
    Inhibition of CDK1 by RO-3306 Exhibits Anti-Tumorigenic Effects in ...
    Aug 3, 2023 · RO-3306 is a selective, ATP-competitive, and cell-permeable CDK1 inhibitor that shows potent anti-tumor activity in multiple pre-clinical models.Missing: preclinical | Show results with:preclinical
  79. [79]
    Sensitivity to cdk1-inhibition is modulated by p53 status in preclinical ...
    Jun 20, 2015 · Inhibition of cdk1 by RO-3306 caused decreased cell viability after 48 h in all NB cell lines investigated and also in the rhabdomyosarcoma cell ...
  80. [80]
    Blocking CDK1/PDK1/β-Catenin signaling by CDK1 inhibitor ...
    Jun 13, 2018 · CDK1 inhibitor RO3306 in combination with sorafenib treatment significantly decreased tumor growth in PDX tumor models. Furthermore, the ...Missing: trials | Show results with:trials
  81. [81]
    The Role of the APC/C and Its Coactivators Cdh1 and Cdc20 in ...
    Abstract. To sustain genomic stability by correct DNA replication and mitosis, cell cycle progression is tightly controlled by the cyclic activity of ...
  82. [82]
    APC/C prevents a noncanonical order of cyclin/CDK activity ... - PNAS
    From these data, we conclude that APC/C enforces CDK4/6i-induced cell cycle arrest by suppressing CDK1 and CDK2 activity through both cyclin A and cyclin B ...
  83. [83]
    Targeting G1–S-checkpoint-compromised cancers with cyclin A/B ...
    Aug 20, 2025 · Dual inhibitors of cyclin A and cyclin B RxL motifs (cyclin A/Bi) selectively kill SCLC cells and other cancer cells with high E2F activity.
  84. [84]
    A compendium of synthetic lethal gene pairs defined by extensive ...
    Sep 18, 2025 · The aim of our study was to further build upon previous analyses and perform dual-guide CRISPR screens across a large panel of models from ...Missing: MPF p53
  85. [85]
    Dinaciclib, a novel CDK inhibitor, demonstrates encouraging single ...
    Dinaciclib has been well tolerated in initial trials, and clinical efficacy has been observed in patients with chronic lymphocytic leukemia and solid tumors.12, ...
  86. [86]
    Southwest Oncology Group S0826: A phase 2 trial of SCH 727965 ...
    Sep 29, 2024 · Dinaciclib (SCH 727965) is a CDK2 inhibitor with additional activity against CDK1, CDK5, and CDK9, which has demonstrated activity in ...
  87. [87]
    Development of CD33-Targeted Dual Drug-Loaded Nanoparticles ...
    Sep 5, 2024 · Effects of ABT-737 and Purvalanol A treatments on pediatric acute myeloid leukemia cell lines as separate and combined treatments. (A) MV4-11, ...
  88. [88]
    EU/3/14/1255 - orphan designation for treatment of acute myeloid ...
    May 6, 2014 · ... volasertib in patients with AML were ongoing. At the time of submission, volasertib was not authorised anywhere in the EU for AML or ...Missing: approval | Show results with:approval
  89. [89]
    Adjunctive Volasertib for AML Not Eligible for Induction Therapy
    Volasertib has also shown robust antitumor activity in a xenograft model of AML; nude mice with established AML tumors treated with volasertib for 4 weeks ...
  90. [90]
    Research Progress of PROTAC-Degraded CDKs in the Treatment of ...
    Jun 13, 2025 · PROTAC can effectively inhibit the proliferation of breast cancer cells by inducing ubiquitination of CDK1, CDK4/6, CDK9, CDK12/13 and their subsequent ...
  91. [91]
    Targeting CDKs in cancer therapy: advances in PROTACs ... - Nature
    Jun 28, 2025 · The anaphase-promoting complex/cyclosome (APC/C) in late mitosis helps destroy B-type cyclins, preventing CDK1 activity and allowing chromosomal ...
  92. [92]
    Gold nanoparticles improve the embryonic developmental ... - NIH
    Our findings suggest the potential role of AuNPs in early embryonic development for mouse oocytes activated artificially and provide new insights.
  93. [93]
    A Potent CDK2 Inhibitor with Selectivity Against Other CDK Family ...
    Sep 3, 2025 · Discovery of highly selective CDK2 inhibitors may minimize off-target effects, reduce toxicity observed with first generation CDK2 inhibitors, ...