Preprint
Review

This version is not peer-reviewed.

Autophagy, P-Body Assembly, Mitochondrial Hyper-Fission, and Cell Death: Unexpected Secondary Roles of the Mediator Kinase Module Following Stress

A peer-reviewed article of this preprint also exists.

Submitted:

10 March 2025

Posted:

11 March 2025

You are already at the latest version

Abstract

Following unfavorable environmental cues, cells reprogram pathways that govern transcription, translation, and protein degradation systems. This reprogramming is essential to restore homeostasis or commit to cell death. This review focuses on the secondary roles of two nuclear transcriptional regulators, cyclin C and Med13, that play key roles in this decision processing. Both proteins are members of the Mediator kinase module (MKM) of the Mediator complex, which, under normal physiological conditions, positively and negatively regulates a subset of stress response genes. However, cyclin C and Med13 translocate to the cytoplasm following cell death and cell survival cue, respectively, interacting with a host of cell death and cell survival proteins. In the cytoplasm, cyclin C is required for stress-induced mitochondrial hyper-fission and promotes regulated cell death pathways. Cytoplasmic Med13 promotes stress-induced assembly of processing bodies (P-bodies) and is required for the autophagic degradation of a subset of P-body assembly factors by cargo hitchhiking autophagy. This review will focus on these secondary, a.k.a. "nigh" jobs" of "cyclin C and Med13, outlining the importance of these secondary functions in maintaining cellular homeostasis following stress.

Keywords: 
;  ;  ;  ;  ;  ;  

1. Introduction

Under normal physiological conditions, cells attain homeostasis through complex regulatory events. This includes balancing the transcription and translation of proteins with their degradation, predominantly by the ubiquitin-proteasome system (UPS) and many macroautophagy (hereafter autophagy) pathways. Understanding the molecular details of how cells balance these events is crucial, as the misregulation of these pathways results in an imbalance in homeostasis. Moreover, as the efficiency of degradation pathways deteriorates with age, such imbalances are linked with age-related pathologies, including cancer and proteinopathies (Figure 1) [1,2,3,4,5,6,7]. Failing degradative pathways allow misfolded proteins to accumulate that, over time, can aggregate into larger structures, which are characteristic of proteinopathies, including Alzheimer's disease and Amyloid Lateral Sclerosis (ALS) [8,9].
Preprints 151920 i001
Cells are constantly subjected to environmental or physiological stress, including osmotic, salt, thermal, and nutritional stress [10]. After exposure, cells face a critical decision: they must either adapt and survive or commit to regulated cell death pathways. Adaptation involves the reprogramming of transcription, translation, and protein degradation pathways. Due to the high degree of conservation among these processes, the budding yeast Saccharomyces cerevisiae is an excellent model for studying the molecular stress response. Insights from this single-celled eukaryote have provided valuable information for understanding mammalian biology. For instance, autophagy was first identified in yeast and later found to be highly conserved across species. Furthermore, yeast has been instrumental in characterizing genetic pathways linked to neurodegenerative diseases such as ALS, Parkinson's, or Alzheimer's [11,12,13,14,15,16].
In addition to guiding studies in higher eukaryotes, studying the stress response in yeast is important in its own right. In the US, the wine industry generates billions of dollars of revenue annually, and craft beer brewing alone has recently emerged as a multibillion-dollar industry [17,18]. S. cerevisiae encounters different stresses during brewing, such as hyperosmotic, ethanol, and thermal stresses [19,20]. In addition, in fed-batch-operated industrial bioreactors, pockets of yeast can face glucose starvation, resulting in negative consequences for production performance [21]. Understanding and manipulating the yeast stress response could spur industrial advancements and increase fermentation yield efficiency [22,23,24].
This review focuses on the Mediator kinase module (MKM – formally Cdk8 kinase module) of the Mediator complex. This highly conserved complex consists of 4 proteins: cyclin C, its cognate kinase Cdk8, and two structural proteins, Med12 and Med13. In mammals, the MKM contains paralogues of its members except cyclin C, namely MED12L, MED13L, and CDK19 [25]. Unlike other better-known members of the cyclin-dependent kinase family, cyclin C-Cdk8 does not mediate cell cycle progression [26,27]. In yeast and mammals alike, the cyclin C-Cdk8 both positively and negatively regulates a subset of stress response genes [28,29,30,31,32,33,34]. In addition, cyclin C and Med13 have secondary cytoplasmic roles following cell death and cell survival cues, respectively (Figure 2). In the cytoplasm, they interact with a host of cell death and cell survival proteins, including the newly defined cargo hitching autophagy pathways [35]. This review outlines the importance of these secondary functions of cyclin C and Med13, maintaining cellular homeostasis following stress.
Preprints 151920 i002

2. Structure of the CKM

The eukaryotic Mediator comprises of a large core 26-subunit Mediator complex (cMED) and a dissociable Mediator Kinase Module (MKM), which functions as a critical coregulator of RNA polymerase II (RNAPII) transcription (Figure 3) [27,36,37,38,39,40]. As such, the mediator complex exists as two distinct entities, depending on whether it is bound to the MKM. Structural and biochemical studies identified the MKM–Mediator association as reversible, leading to the model that the MKM sterically inhibits the Mediator from interacting with RNA Pol II at promoters [27,36,41,42,43,44,45,46,47,48]. Unbound MKM also exists and likely functions to regulate transcription independently of the cMed [42,49,50]. Additional roles for MKM-Mediator in regulating RNA Pol II elongation rate have been described [51,52]. However, it remains unclear if this function is mediated by the MKM-Mediator complex or the MKM acting as an independent entity [51,52,53]. In yeast, the MKM predominantly represses stress response genes, while in mammalian cells, the MKM both negatively and positively regulates a wider array of genes [54,55]. A recent cryo-electron microscopy structure of the yeast MKM also revealed that Med13's bilobal architecture resembles that of argonaute proteins [56]. While this would imply a new role of the MKM in interacting with nucleic acid duplexes, the lack of several critical conser ved catalytic residues found in other argonaute proteins adds complexity to how exactly Med13 regulates transcription [56].
Preprints 151920 i003

3. Transcriptional Reprogramming by the MKM Following Stress

Overview of MKM regulated gene expression.
Precise mechanistic details of how the MKM-Mediator regulates gene expression are still emerging [57]. Typically, cyclin binding activates CDKs, which are stabilized by CDK-activating kinase phosphorylation in the CDK T-loop [58]. Cdk8 lacks this canonical phosphorylation residue, with Med12 possibly stabilizing the cyclin C-CDK8/CDK19 interaction in humans [42,59]. Cyclin C and its cognate kinase are constitutively active in normal unstressed conditions, where they phosphorylate a multitude of factors to both positively and negatively regulate transcription [28,29,33,51,52,53,60,61,62,63,64,65,66,67,68,69]. Key among these targets are many transcription factors (TF) [28,29,30,31,32,33,34]. In addition, the MKM and MKM-Mediator regulate different targets, including histones [42] and RNA polymerase II tail, providing additional control of gene transcription [70,71]. In mammals, phosphorylation of TFs by the MKM-Mediator may be transmitted directly through enhancer regulatory elements to either repress or activate transcription. [53,72]. In summary, in all eukaryotes, the MKM influences important transcriptional outcomes that play a role in maintaining cellular and developmental homeostasis.
B. The MKM predominantly negatively regulates SRGs in yeast.
In yeast, the MKM predominantly negatively regulates a subset of stress response genes, including those that encode for antioxidants, molecular chaperones, autophagy, and development [33,37,49,54,73,74,75,76,77,78,79,80,81,82]. Activating these genes requires disassociating the MKM from cMed and disassembling the MKM. However, whether these actions co-occur remains unknown as the molecular mechanisms controlling these processes are poorly understood. Studies from our group have revealed that the mechanism used to disassemble the MKM depends on the nature of the stress signal and involves the translocation of either cyclin C or Med13 to the cytoplasm (Figure 2). This change in subcellular address contributes to stress-induced transcriptional reprogramming and is also required for cyclin C and Med13 to perform their secondary cytoplasmic functions. In short, cyclin C, but not other MKM members, relocates to the cytoplasm, following cell death cues (oxidative stress -ROS) [83]. Cytoplasmic cyclin C is necessary and sufficient for ROS-induced mitochondrial hyperfission and promotes regulated cell death [84]. This response is conserved [85,86,87,88]. Med13, but not cyclin C, crosses the nuclear pore complex following cell survival cues (nitrogen starvation). In the cytoplasm, Med13 promotes the assembly of a subset of ribonucleoproteins (RNPs) into mRNA processing bodies (P-bodies) [89]. Med13 also directs the autophagic degradation of a subset of P-Body assembly factors, including the conserved decapping protein Edc3, by a newly discovered hybrid autophagy mechanism called cargo hitchhiking autophagy [35,82,90]. The subcellular address of Med13 and Med13L in humans following stress remains unknown.
C. The MKM positively regulates a subset of genes required for translation.
Although the MKM primarily represses stress response genes (SRGs), we recently discovered that it also positively regulates a subset of genes encoding ribosomal proteins (RPs) and translation initiation factors (TIFs) under normal physiological conditions [91]. This mechanism demonstrates the conserved nature of MKM activity, as it also occurs in mouse embryonic fibroblasts (MEFs) and human cell lines [91]. The repression of this subset of translation-associated genes occurs in response to stress and is dependent on the disassembly of the MKM [91]. This process makes sense because ribosome biogenesis consumes over 60% of cellular energy and resources [92]. Reprogramming ribosome biosynthesis helps reduce intracellular energy consumption and represents a conserved mechanism for cellular survival during stress [93]

4. Mechanisms of CKM Disassembly

A. MKM disassembly following cell death cues triggered by ROS
In response to oxidative stress, cyclin C, but not Cdk8, translocates to the cytoplasm, resulting in MKM disassembly [83]. This role is conserved [69,85]. In yeast, the molecular details of MKM disassembly in response to ROS involve an intertwined network of phosphorylation and ubiquitination events. The stress signal is transmitted by the conserved MAPK of the Cell Wall Integrity (CWI) pathway to Slt2 (Mpk1, (ERK ortholog) [94] that directly phosphorylates cyclin C and Med13 [95,96,97,98]. Intriguingly, Kdx1 (Mlp1), the pseudokinase of the CWI pathway, is also required for cyclin C release [97]. Kdx1 also interacts with the transcription factor Ask10 and likely induces cyclin C translocation to the cytoplasm indirectly [97,99]. CWI triggered phosphorylation of Med13 results in its destruction by UPS mediated by the E3 ligase complex SCFGrr1 [98,99]. In addition, cyclin C is directly phosphorylated at a single site (S266). The net outcome is MKM disassembly and cyclin C nuclear release [83,84] In yeast, the UPS destroys cytoplasmic cyclin C following its role in mitochondrial hyper-fission [54,74,84].
Genetic studies showed that Med13 destruction is required for cyclin C nuclear release. In addition, Med13 destruction requires a priming event mediated by Cdk8 in unstressed cells [98]. Thus, recognition of the Med13 degron uses two phosphorylation marks, one to prime the degron, the second for its recognition by ubiquitin ligases [100,101]. This ensures that releasing cyclin C to the cytoplasm is the correct response to the environmental input. Additionally, direct phosphorylation of Med13 by Snf1, a highly conserved adenosine monophosphate-activated protein kinase (AMPK) that is activated in response to a variety of stresses is required for its translocation [102,103].
Taken together, the studies discussed above suggests that Med13 acts as a physical tether, keeping cyclin C in the nucleus. Consistent with this, in the absence of Med13, cyclin C is aberrantly released into the cytoplasm where it induces mitochondrial fission in the absence of stress [99]. Furthermore, deletion of the holoenzyme associating domain (HAD), the domain on cyclin C that interacts with Med13 [71] also results in the precocious release of cyclin C from the MKM into the cytoplasm and mitochondrial fission in the absence of stress [99]. The HAD is highly conserved, being required for the cyclin C-Med13 interaction in mammalian cells [87]. Based upon these studies we designed a peptide mimetic of this domain (S-HAD) which stimulates cyclin C nuclear release in mammalian cells in the absence of stress, highlighting the conservation of both the domain and function [87].
B. MKM disassembly following cell survival cues triggered by nitrogen starvation
In the yeast model system, nitrogen starvation activates both bulk and cargo hitchhiking autophagy pathways. Bulk autophagy is essential for recycling cellular components, producing energy, and ensuring survival [104]. In contrast, cargo hitchhiking autophagy is a selective pathway that degrades specific substrates, including Med13, certain ribosomal proteins, and components involved in PB assembly [35]. Given that the MKM represses many crucial autophagy genes, it is not surprising that effective induction of autophagy requires its disassembly [82,105]. Following nitrogen starvation cyclin C does not translocate to the cytoplasm. Instead, the ubiquitin-proteasome system (UPS) targets and degrades cyclin C in the nucleus via an unidentified E3 ligase [35,82]. This response is vital for survival, as it prevents cyclin C from causing mitochondrial hyper-fission and regulated cell death [105]. Thus mitochondria remain hyperfused, a morphology associated with maximal ATP production [4,106]. MKM disassembly following nitrogen starvation also results in Med13 translocating to the cytoplasm [82,90]. Here it is required for P-Body assembly, and the autophagic degradation of a subset of P-Body assembly factors by cargo hitchhiking autophagy [35,89]
How the stress signal is transmitted to the MKM following nitrogen starvation remains unknown. However, it is known that in yeast the activity of Slt2 is activated in response to the Target of Rapamycin Complex 1 (TORC1) inhibition [107]. However, phosphorylation of cyclin C by Slt2 is not required for its degradation following nitrogen starvation [105]. This suggests that a different protein, possibly Med13, is the target of Slt2 activity following starvation stress. These studies highlight how the MKM responds to cell death and cell survival cues, despite the fact that the CWI is activated in both cases. Interpretation of the input signal is critical as expressing a fusion protein in which cyclin C is tethered to the outer mitochondrial membrane results mitochondrial fission and execution cell death pathways following cell survival cues [105].

5. The Roles of Cytoplasmic Cyclin C

Cytoplasmic cyclin C is required for stress induced hyper-fission in both yeast and mammalian cells, indicating that this response is conserved [108]. Cytoplasmic cyclin C also promotes regulated cell death pathways in both yeast and mammals. However, whilst we discovered that cyclin C interacts with the BAX, a BCL-2 family pro-apoptotic member, in mammalian cells, yeast cells do not harbor a BAX ortholog [109]. Although ROS exposure leads to the same outcome in both yeast and mammals, different mechanisms are employed to direct cells into regulated cell death pathways.
A. Cyclin C and stress-induced mitochondrial hyperfission in yeast.
Mitochondrial fission occurs in two distinct scenarios: firstly, during cell division under normal physiological conditions and mitophagy, and secondly, as a response to stress that results in cell death The key difference between these pathways is the presence of cytoplasmic cyclin C in the latter (see Figure 4). In yeast, following ROS cyclin C translocates to the cytoplasm where it interacts with the yeast mitochondrial fission machinery adaptor protein called Mdv1 (Figure 5A) [84]. Mdv1, and another adaptor protein called Caf4, recruits Dnm1 to sites of mitochondrial fission by interacting with the conserved mitochondrial outer membrane (OMM) protein called Fis1 [110,111]. Dnm1 is a GTPase, that works by constricting and dividing mitochondria in a GTP-hydrolysis dependent manner [112,113]. Here Dnm1 forms a multimeric ring complex around mitochondria which initiates separation of the inner and outer mitochondrial membranes [114,115]. Following GTP hydrolysis the complexes constrict, resulting in mitochondrial fission [116]. Exactly, how the cyclin Dnm1-fission complex reduces mitochondrial hyper-fission is unclear in yeast.
Preprints 151920 i004
This same fission mechanism occurs during regular cell division or during homeostasis to control mitochondrial quality except that cyclin C is not present [117,118]. This has led to a model in which cyclin C localization to the OMM stimulates hyper-fission, a phenotype required for initiating cell death pathways [84,119,120]. In support of this model, the absence of cyclin C, or in cis and trans mutants that keep cyclin C in the nucleus, stress-induced hyper-fission and cell death is significantly reduced [84]. While the exact mechanism by which cyclin C helps induce stress-induced mitochondrial fragmentation is still unclear, cyclin C most likely acts early in the stress response to promote the formation of productive Fis1-Mdv1-Dnm1 complexes, leading to fission [84]. Other observations strongly support this model: firstly, artificially placing cyclin C at the mitochondria via fusion of the OMM-associating domain of Fis1 to cyclin C's C-terminus is enough to induce fission even in unstressed cells [105]. Secondly, cyclin C mediated hyperfission role of cyclin C is not dependent upon its transcriptional duties [83,84]. Lastly, as mentioned above, retaining cyclin C in the nucleus following ROS stress significantly reduces hyperfission.
Preprints 151920 i005
Cyclin C and mitochondrial hyper-fission in mammals.
In mammals, DRP1-dependent mitochondrial fission following oxidative stress is also part of the regulated cell death pathway (RCD, Figure 5A) [108]. Just as in yeast, stress-induced translocation of cyclin C from the nucleus to the cytosol is responsible for the mitochondrial hyper-fission [85]. Outlining the conserved nature of this response, Escherichia coli purified yeast GST-cyclin C can induce mitochondrial fission in mouse embryonic fibroblasts [85]. Cyclin C directly interacts with DRP1 GTPase domain to stimulate it via its second cyclin box domain (CB2) [86]. In this context, the interaction between cyclin C and DRP1 induces structural changes in DRP1, transforming it from oligomers with low GTPase activity to dimers capable of forming high-GTPase activity filaments [86]. The conservation of the cyclin C–Dnm1/DRP1 association could help in future studies examining DRP1 related diseases including neurodegenerative disorders, ischemia-reperfusion injury and optic atrophy [121,122].
Many Drp1 associated diseases are associated with point mutations in the Drp1 gene, which can then be generated in yeast and used to study mitochondrial morphology and protein function [121,123]. Additionally, DRP1 inhibitors are being tested in clinical trials to treat both cancers and cardiovascular diseases [124,125,126,127]. Due to the important role DRP1 plays in cell division in healthy cells, inhibiting it this broadly can often have negative consequences. Since many DRP1 related diseases involve a disease-mediated increase in ROS causing DRP1 dependent mitochondrial hyper-fission, specifically targeting the ROS dependent cyclin C – DRP1 interaction could help overcome negative side effects from general DRP1 inhibition [128,129,130]
Cyclin C and cell death in yeast.
Numerous key features of RCD observed in mammalian cells are also conserved in yeast [131]. However, the mechanisms driving RCD in yeast are distinct from those of metazoan apoptosis and remain poorly understood. For instance, RCD can occur in yeast both in a manner dependent on and independent of its sole caspase-like protein, Yca1 [132,133,134,135]. Interestingly, cells lacking cyclin C display remarkable resistance to stress-induced cell death, a phenomenon observed in both yeast and mammalian systems [85,136]. Although the precise role of cyclin C in this context is not fully understood, simply enhancing cyclin C-mediated mitochondrial fission through overexpression alone is insufficient to trigger programmed cell death (PCD) [84]. This suggests that an additional signal(s) is necessary to initiate RCD, which might come from another protein localized to the mitochondria during the stress response or through a stress response pathway that modifies cyclin C post-translationally. This important distinction sets cyclin C apart from other known inducers of mitochondrial RCD. For example, ectopic targeting of p53 or the BH3 family member Bax to the mitochondria can effectively induce cell death in non-stressed mammalian cells. In contrast, there is conflicting evidence for the role of Ybh3, the sole BH3 protein in yeast, in inducing regulated cell death [137,138,139,140]. In conclusion, while cyclin C plays an integral role in RCD in yeast, the exact mechanisms underlying its function are still not fully elucidated.
Cyclin C and cell death in metazoans.
BAX and other members of the BCL-2 family are important regulators of the intrinsic pathway of apoptosis in mammalian cells [141]. Upon activation, BAX oligomerizes at the OMM resulting in mitochondrial outer membrane permeabilization (MOMP) [142]. In yeast there are no apparent homologs of the Bcl-2 family proteins. Cytoplasmic cyclin C is required for both the activation and mitochondrial localization of BAX in mammalian cells [87]. It directly interacts with active Bax and is dependent on the presence of DRP1 [87]. Later research also confirmed that BAX and DRP1 directly interact [143]. This suggests a model where cyclin C, BAX, and DRP1 all together form a complex essential for apoptosis. Artificially stimulating cyclin C nuclear release in the absence of stress still leads to BAX association, however, BAX does not oligomerize [87]. This supports the model in which release of cyclin C from the nucleus is insufficient to promote cell death pathways.
S. cerevisiae does not encode for any known homologs of BCL-2 family. However, murine BAX expression induces cell death in yeast [144]. This toxicity can be rescued by co-expressing antiapoptotic members of the BCL-2 family, such as BCL-2 or BCL-XL [144]. How BAX kills yeast remains elusive. Deletion of the mitochondrial inner membrane protein Uth1 results in strengthened cell wall that contributes to resistance to BAX-mediated toxicity, [145,146]. Also, overexpression Rgl1, a pro-survival protein that plays a role in iron metabolism, suppresses BAX toxicity [147]. However, the relationship between these two events remains unclear, and has not led to a mechanism on how BAX mediates cell death in yeast. Likewise, despite identifying a yeast homolog of the BAX inhibitor protein 1 (Bxl1), this protein cannot counter the toxic effects of Bax expression in yeast [140,148,149]. Lastly, it is known that the expression of BAX results in cytochrome c release from the mitochondria, confirming that BAX permeabilizes mitochondrial membranes [150]. This is supported by the finding that the F0F1-ATPase proton pump is essential for BAX-induced cell death in yeast and mammalian cells [151,152]. This suggests that the permeability pore operates through a conserved mechanism. The cytochrome c release is partially dependent on ER-mitochondrial association; however, cytochrome c is not required for cell death in yeast [153,154]. Cyclin C's role in BAX-dependent cell death in yeast is unclear, but its mitochondrial association and involvement in RCD make it a protein of interest.

6. Cytoplasmic Roles of Med13

In the yeast model system, Med13 translocates to the cytoplasm following TOR1 inhibition triggered by either nitrogen starvation or treatment with rapamycin [82,90]. Here it is required for processing body (P-body) assembly and the autophagic degradation of a subset of P-body assembly factors (Figure 5B) [89]. It remains unknown if these roles are conserved, but given the conservation of cyclin Cs secondary job, it is highly likely that Med13 will follow suit.
A. Med13's role in P-body assembly.
Inhibiting mRNA translation is a critical strategy that cells employ to manage protein levels effectively. Under stress, cells strategically downshift the production of proteins vital for growth while ramping up those necessary for adaptation. A pivotal mechanism for blocking mRNA translation is the formation of P-bodies. These highly conserved, membrane-less structures within the cytoplasm are reservoirs that contain non-translating mRNA-protein complexes (mRNPs) [155,156]. Initially, it was believed that mRNAs within P-bodies were simply stored for future translation or subjected to degradation [157,158,159,160]. However, subsequent studies have revealed that P-bodies are not crucial for mRNA degradation [161]. Instead, they are increasingly recognized as crucial temporary storage sites for translationally repressed mRNAs that remain ready for activation when conditions improve [162].
The assembly of P-bodies occurs through a process known as liquid-liquid phase separation (LLPS), creating dynamic droplets that are distinctly separate from the surrounding cytoplasm [163]. Their assembly benefits cells, as P-body formation occurs more rapidly than changes in the transcriptional or translational programs. Moreover, the rapid disassembly of P-bodies after the stress abates, rapidly provides cells with ready-to-go translation components. The proteins that reside within P-bodies are predominantly RNA-binding proteins or factors with low sequence complexity that contain prion-like domains (PLDs). These PLDs possess the remarkable ability to undergo spontaneous conversion into aggregated states and serve as templates for the recruitment of additional proteins.
In yeast, P-bodies are consistently present under normal physiological conditions, but their size and number notably increase in response to various stressors, such as nitrogen and carbon starvation [163]. The assembly of P-bodies is fundamentally dependent on several conserved decapping proteins, including Dcp1/Dcp2, Edc3, Dhh1, along with the Pat1–Lsm1–7 complex and Xrn1 [158]. Edc3 serves as a crucial scaffold for P-body assembly, by leveraging its multivalent interactions [164,165,166,167,168]. Med13 is essential for P-body assembly following nitrogen starvation; the stark reduction of Edc3 foci in med13Δ cells underscores its importance [89]. The mechanistic details of why Med13 is required for P-body formation remain to be completely elucidated; however, it is clear that the PLD domain of Med13 is indispensable for this process [89]. The exact function of these polyQ/N tracts is not well understood, but it has been suggested that the domain can interact with itself or with other polyQ/N regions to promote the aggregation of mRNPs [168].
These studies indicates that Med13 plays a significant role in promoting the liquid-liquid phase separation (LLPS) of P-body proteins. Furthermore, Med13 contains two motifs strongly linked with LLPS: an RNA Recognition Motif (RRM) and a large intrinsically disordered region (IDR). IDRs are recognized as key drivers of LLPS, as they can simultaneously create multiple interactions with other components [169]. Additionally, the amino acids within IDRs are more exposed and accessible to post-translational modifications, which are critical regulators of biomolecular phase separation [170,171,172]. This suggests that Med13 itself may undergo LLPS, a direction future studies should pursue.
B. Med13's role in cargo-hitchhiking autophagy (CHA)
Med13 is also required for the autophagic degradation of both Edc3 and Dhh1 following nitrogen starvation. Since Med13 does not direct the autophagic degradation of Xrn1, this suggests that Med13 serves as an exclusivity factor that differentiates among various P-body components [89]. A new hybrid autophagy mechanism implements the autophagy of these decapping proteins called cargo-hitchhiking autophagy, which uses components from both selective and bulk autophagy mechanisms [35].
i) Snx4 promotes CHA
CHA was previously named Snx4-assisted autophagy, as this conserved sorting nexin is required for the degradation of all identified CHA cargos to date [35,82,173,174,175]. Sorting nexins are a family of conserved phosphoinositide-binding proteins that play fundamental roles in orchestrating cargo sorting through the endosomal network. As such, human SNX4 is implicated in a variety of synaptic processes [176,177]. Significantly, SNX4 has already been linked with the etiology of AD, with SNX4 protein levels being decreased by 70% in brains of severe Alzheimer's disease (AD) cases [178].
Sorting nexins are recruited to endosomal membrane domains by a phox homology (PX) domain that recognizes phosphatidylinositol-3-phosphate (PtdIns3P) [175]. Snx4 also contains two Bin–Amphiphysin– Rvs (BAR) domains that sense membrane curvature and tubulate vesicles [179]. The molecular role of Snx4 (Atg24) in autophagy is also related to membrane bending as it stabilizes and drives the opening of the inner membrane rim of non-selective phagophores in yeast [180]. The net effect is a phagophore membrane with a wide enough opening to accommodate large cargos, including ribosome and proteasome subunits [180,181,182,183].
ii) CHA uses phagophores built by the Atg17 scaffold complex
In CHA, specific cargo, including Med13, are recruited to growing phagophores primarily used to degrade random cytosolic contents by non-selective (a. k. a. bulk) autophagy. These phagophores are triggered by starvation stress. Their formation is dependent on the Atg17 scaffold complex, which consists of the Atg17 scaffold and two regulatory proteins, Atg29 and Atg31 [184]. This process differs from selective autophagy, where phagophores are constructed using the Atg11 scaffold [185]. Selective autophagy pathways play crucial roles in maintaining homeostasis under physiological conditions. They are responsible for removing dysfunctional proteins (such as in mitophagy, ER-phagy, and ribophagy), protein aggregates (aggrephagy), and pathogens like viruses and bacteria (xenophagy) in higher eukaryotes [186]. Likely, Med13-Edc3 is not the only cargo captured in Atg17-built phagophores. Instead, these phagophores probably contain random cytosolic contents alongside other cargo that may hitch a ride, including a subset of ribosomal proteins [174].
The formation of the phagophore assembly site (PAS) for both selective and non-selective autophagy depends on liquid-liquid phase separation (LLPS) [187,188]. During starvation, the Atg1 complex—comprised of Atg1, Atg13, and a trimeric Atg17 scaffold undergoes LLPS [189]. This fluidity is vital for recruiting downstream Atg proteins necessary for autophagosome formation. As mentioned above P-Body assembly occurs via LLPS. How these two biomolecular condensates (BMC) interact remains unknown. One intriguing possibility is that Med13 could play a role, possibly acting as a conduit between these two BMCs.
iii) CHA uses selective autophagy receptor proteins to recognize cargo
Although CHA uses phagophores built for the degradation of random cytoplasmic contents, cargo recognition is dependent upon an autophagic receptor protein [35]. The autophagic receptor for the selective degradation of Med13 is Ksp1. Typical of autophagic receptors, Ksp1 interacts with the phagophore-bound protein Atg8 via its conserved Atg8 interaction motif (AIM) [90,190]. Ksp1 is a casein-like kinase that negatively regulates autophagy in replete media [191,192,193,194,195]. However, its role as a receptor protein is kinase-independent, illustrating its dual and opposing roles in autophagy. Ksp1 also associates with ribosomal and PB proteins [196,197]. Interestingly, Ksp1 does not directly interact with Edc3. Instead, Med13 performs this role, suggesting it may act as a conduit between Edc3 and Ksp1 [90].
iv) The nucleoporin Gle1 is required for CHA of Med13.
Cyclin C is a small 33 kDa protein that can diffuse through the nuclear pore complex (NPC) without the assistance of active transport by exportins. In contrast, Med13 is a larger scaffold protein, approximately 150 kDa in size, which requires active transport to cross the nuclear-cytoplasmic barrier. However, our studies have shown that β karyopherin exportins are not needed for Med13's transit through the NPC. Similarly, Dpb5 and the Mex67-Mtr2 heterodimer, which can transport cargo across the NPC, do not play a role in this process. Instead, Gle1, a conserved component of cytoplasmic NPC fibrils, is essential for Med13's passage across the NPC [82].
GLE1 is a mobile nucleoporin in mammalian cells [198], suggesting that it could play a similar role in the nuclear export of Med13. Interestingly, in yeast Med13 remains in the nucleus following nitrogen starvation in snx4∆ mutants [82]. This observation implies that Snx4 may communicate with the NPC, potentially by interacting with Gle1 during the transfer of Med13 as it exits the NPC. Future experiments will be necessary to investigate this idea further.

7. Diseases Associated with the MKM

Diseases associated with cyclin C-CDK8/CDK19
i) Cyclin C tumor suppressor roles
Deletions of the genomic locus containing the cyclin C encoding gene (6q21) are found in blood cancers and solid tumors, hinting at a tumor suppressor role for cyclin C [88,199,200]. However, the extent to which the nuclear and mitochondrial roles play in tumor suppression is unknown.
The cyclin C gene is heterozygously deleted in a significant majority of T-cell acute lymphoblastic leukemia (T-ALL) cases [199]. In vivo, the cyclin C-CDK8 kinase phosphorylates ICN1, resulting in Notch1 repression, a single-pass transmembrane receptor [201]. Cyclin C heterozygosity or its deletion derepresses ICN1 levels, promoting T-ALL progression [199]. Notch activation is present in many cancers, including several where 6q21 deletions have been observed (acute lymphoblastic leukemia, non-Hodgkin's Lymphoma, prostate cancer, amongst others), likely due to this specific suppressive role involving ICN1 [202,203,204,205,206]. In other cancers, such as human squamous cell carcinoma, Notch1 functions as a tumor suppressor. Investigating cyclin C deletion or activation in those tumors could reveal whether it plays a role in those tumors as an oncogene instead [199,207,208].
Cyclin C also plays a tumor suppressor role in anaplastic thyroid tumors [209]. Deletion of Cyclin C alone in the thyroid only stimulates a modest increase in hyperplastic growth. However, in combination with ablation of the PTEN tumor suppressor [210], the thyroid size increases dramatically, eventually killing the animal [209]. Despite the increase in ROS production throughout thyroid cancer development, it is not the mitochondrial role of cyclin C that is involved, but rather alterations in the Jak-Stat signaling pathway that destabilize tumor suppressors p53 and p21 [87,211]. Given the high degree of conservation between yeast and mammalian cyclin C, yeast cyclin C is an ideal model for understanding its role in carcinogenesis in mammals.
ii) Cdk8 is an oncogene
CDK8 was first reported as a putative oncogene in colorectal cancer in 2012 [212]. In this context, CDK8 mediates the aberrant activation of the Wnt/ β—catenin signaling pathway. Additionally, the phosphorylation of various transcription factors, such as Smads, STAT1, and NFκB, by CDK8 is frequently dysregulated in many other types of cancer [28,64,213]. This suggests that cancers associated with CDK8 are predominantly driven by transcriptional dysregulation [214]. Currently, mutations in both CDK8 and CDK19 have been linked to over 100 malignancies, including cancers of the colon [212,214], breast [215], prostate [216], pancreas [217], melanoma [218] and leukemias [219]. It comes as no surprise then that modulating the transcriptional activity of the MKM has emerged as an attractive therapeutic strategy for treating cancer. Consequently, specific inhibitors targeting CDK8/CDK19 are currently being tested in various clinical trials [213].
Cyclin C and TDP-43 mediated cell death
Transactive Response Binding Protein 43 (TDP-43) is a ubiquitously expressed protein involved in mRNA regulation [220]. It is the primary aggregate protein found in many age-related proteinopathies such as Amyotrophic Lateral Sclerosis (ALS) and Frontal Tubular Dementia (FTD) [221]. TDP-43 toxicity has been associated with autophagy impairment, mitochondrial dysfunction, and the generation of reactive oxidative species [222,223,224,225,226].
Increased levels of ROS are observed in several neurodegenerative diseases, including ALS [227]. Moreover, mitochondrial hyper-fission has also been observed in the post-mortem neurons of patients diagnosed with ALS and other proteinopathies as well as in cell culture [228,229,230,231,232]. Additionally, the hallmark of neurodegeneration is the aberrant premature dying of neurons [233]. This has led to a model in which the expression of mutant TDP-43 results in the formation of aberrant cytoplasmic aggregates of TDP-43.
Using the established yeast model of TDP-43 pathology, it has been shown that cyclin C, but not Med13, promotes TDP-43-mediated cell death [234]. Likewise, the deletion of Dnm1 rescues TDP-43 toxicity, pointing to the potential mitochondrial fission role of cyclin C in TDP-43 toxicity. Furthermore, cyclin C translocates to the cytoplasm following TDP-43 overexpression. The free radical scavenger, N-acetyl-cysteine (NAC), inhibits cyclin C cytoplasmic relocalization. These studies suggest the possibility that cytoplasmic cyclin C may promote mitochondrial hyper-fission and cell death upon expression of mutant TDP-43. This is exciting as cyclin C is a new player in TDP-43 biology and could represent a new target for drug therapies. This is important as currently, there are no effective cures for ALS, and patient survival upon diagnosis is around 3-5 years [235].
B. Diseases associated with Med13 biology.
In mammals, the MKM contains paralogues of its members except cyclin C, namely MED12L, MED13L, and CDK19 [25]. The biological roles of these paralogues remain poorly understood, but they appear to be functionally distinct [236]. Many mutations in human MED13L result in MED13L syndrome, a disease characterized by a range of symptoms that vary in severity. These include intellectual disability, facial dysmorphism, hypotonia, congenital heart disease, as well as speech and motor delay [237,238,239,240]. In MED13L, mutant fibroblasts cyclin C is aberrantly released into the cytoplasm, leading to mitochondrial fragmentation and increased mitochondrial dysfunction [241]. Variants in CDK8, MED13, MED12, and MED12L are also associated with neurodevelopmental disorders [242,243,244,245,246,247], with cyclin C's role here being unknown. In addition, mutations in Med12 are linked with uterine leiomyomas [248]. Interestingly, all the MKM neurodevelopmental disorders are phenotypically similar, indicating a probable overlap in pathogenic mechanisms.

8. Outlook

The MKM has evolved in the last 20 years from being a transcriptional regulator to mediating mitochondrial hyper-fission, regulated cell death, PB assembly, and cargo hitchhiking autophagy. These secondary roles depend upon the nuclear release of either cyclin C or Med13, following ROS or starvation stress, respectively. These studies support the emerging concept that proteins can perform two distinct functions, referred to as "day and night jobs" [249,250], which various external or intrinsic stimuli can trigger. As mentioned above, Ksp1 has two opposing roles in autophagy, which are dependent and independent of its kinase function [90,191,192,193,194,195]. Likewise, Cdk8 does not translocate to the cytoplasm following stress in either yeast or mammals [84,85], indicating that the cytoplasmic role of cyclin C is kinase independent.

Funding

K.F. C is supported in part by N. I. H. grant #1R21AG080666-01A1.

Acknowledgments

We especially thank members of the Randy Strich and Katrina Cooper laboratories for the critical reading of this manuscript.

Declaration of interest

The author declares no competing or financial interests.

Abbreviations

Adenosine Monophophate-activated Protein Kinase:AMPK ; Amyloid Lateral Sclerosis:ALS ; ATG8 Interaction Motif:AIM ; Bin–Amphiphysin– Rvs:BAR ; Biomolecular Condensates:BMC ; Cell Wall Integrity pathway:CWI ; Core-Mediator Complex:cMED ; Holoenzyme-associating Domain:HAD ; Inner Mitochondrial Membrane:IMM ; Intrinsically Disordered Region:IDR ; Liquid-liquid Phase Separation:LLPS ; Mediator Kinase Module:MKM ; Outer Mitochondrial Membrane:OMM ; Mitochondrial Outer Membrane permeabilization MOMP: Mouse Embryonic Fibroblasts:MEFs ; N-acetyl-cysteine:NAC ; Non-translating mRNA-protein complexes:mRNPs ; Nuclear Pore Complex:NPC ; Phagophore Assembly Site:PAS ; Phosphatidylinositol-3-phosphate:PI(3)P ; Phox Homology:PX ; Prion-like Domains:PLDs ; Processing Body:P-bodies ; Regulated Cell Death:RCD ; Ribonucleoproteins:RNPs ; Ribosomal Proteins:RPs ; RNA Polymerase II:RNAPII ; RNA Recognition Motif:RRM ; Stress Response Genes:SRGs ; T-Cell Acute Lymphoblastic Leukemia:T-ALL ; Transcription Factors:TFs ; Translation Initiation Factors:TIFs ; Ubiquitin Proteasome System:UPS.

References

  1. Koyuncu, S., et al., Rewiring of the ubiquitinated proteome determines ageing in C. elegans. Nature, 2021. 596(7871): p. 285-290. [CrossRef]
  2. Kevei, É. and T. Hoppe, Ubiquitin sets the timer: impacts on aging and longevity. Nature Structural & Molecular Biology, 2014. 21(4): p. 290-292. [CrossRef]
  3. Tramutola, A., et al., It Is All about (U)biquitin: Role of Altered Ubiquitin-Proteasome System and UCHL1 in Alzheimer Disease. Oxid Med Cell Longev, 2016. 2016: p. 2756068. [CrossRef]
  4. Abdullah, M.O., et al., Mitochondrial hyperfusion via metabolic sensing of regulatory amino acids. Cell Reports, 2022. 40(7): p. 111198. [CrossRef]
  5. Aman, Y., et al., Autophagy in healthy aging and disease. Nature Aging, 2021. 1(8): p. 634-650. [CrossRef]
  6. Leidal, A.M., B. Levine, and J. Debnath, Autophagy and the cell biology of age-related disease. Nature Cell Biology, 2018. 20(12): p. 1338-1348. [CrossRef]
  7. Kim, Y.J., et al., Ubiquitin–proteasome system as a target for anticancer treatment—an update. Archives of Pharmacal Research, 2023. 46(7): p. 573-597.
  8. Ajmal, M.R., Protein Misfolding and Aggregation in Proteinopathies: Causes, Mechanism and Cellular Response. Diseases, 2023. 11(1). [CrossRef]
  9. Louros, N., J. Schymkowitz, and F. Rousseau, Mechanisms and pathology of protein misfolding and aggregation. Nature Reviews Molecular Cell Biology, 2023. 24(12): p. 912-933. [CrossRef]
  10. Dawes, I.W. and G.G. Perrone, Stress and ageing in yeast. FEMS Yeast Research, 2019. 20(1). [CrossRef]
  11. Cazzanelli, G., et al., The Yeast Saccharomyces cerevisiae as a Model for Understanding RAS Proteins and their Role in Human Tumorigenesis. Cells, 2018. 7(2). [CrossRef]
  12. Cerqueira, F., et al., A Cyclam Salt as an Antifungal Agent: Interference with Candida spp. and Cryptococcus neoformans Mechanisms of Virulence. Antibiotics (Basel), 2024. 13(3).
  13. Stieber, H., et al., The sphingolipid inhibitor myriocin increases Candida auris susceptibility to amphotericin B. Mycoses, 2024. 67(4): p. e13723. [CrossRef]
  14. Schneider, K.L., et al., Elimination of virus-like particles reduces protein aggregation and extends replicative lifespan in Saccharomyces cerevisiae. Proc Natl Acad Sci U S A, 2024. 121(14): p. e2313538121. [CrossRef]
  15. Zhou, L. and R. Xu, Invertebrate genetic models of amyotrophic lateral sclerosis. Front Mol Neurosci, 2024. 17: p. 1328578. [CrossRef]
  16. Rencus-Lazar, S., et al., Yeast Models for the Study of Amyloid-Associated Disorders and Development of Future Therapy. Front Mol Biosci, 2019. 6: p. 15. [CrossRef]
  17. Koller, H. and L.B. Perkins, Brewing and the Chemical Composition of Amine-Containing Compounds in Beer: A Review. Foods, 2022. 11(3). [CrossRef]
  18. Reiter, T., et al., Transcriptomics Provides a Genetic Signature of Vineyard Site and Offers Insight into Vintage-Independent Inoculated Fermentation Outcomes. mSystems, 2021. 6(2). [CrossRef]
  19. Liu, Y., et al., Stress tolerance enhancement via SPT15 base editing in Saccharomyces cerevisiae. Biotechnol Biofuels, 2021. 14(1): p. 155. [CrossRef]
  20. Postaru, M., et al., Cellular Stress Impact on Yeast Activity in Biotechnological Processes-A Short Overview. Microorganisms, 2023. 11(10). [CrossRef]
  21. Minden, S., et al., Performing in spite of starvation: How Saccharomyces cerevisiae maintains robust growth when facing famine zones in industrial bioreactors. Microb Biotechnol, 2023. 16(1): p. 148-168. [CrossRef]
  22. Iorizzo, M., et al., Role of Yeasts in the Brewing Process: Tradition and Innovation. Processes, 2021. 9(5): p. 839. [CrossRef]
  23. Yang, T., et al., Breeding of high-tolerance yeast by adaptive evolution and high-gravity brewing of mutant. J Sci Food Agric, 2024. 104(2): p. 686-697. [CrossRef]
  24. Hou, D., et al., Effect of environmental stresses during fermentation on brewing yeast and exploration on the novel flocculation-associated function of RIM15 gene. Bioresour Technol, 2023. 379: p. 129004. [CrossRef]
  25. Soutourina, J., Transcription regulation by the Mediator complex. Nat Rev Mol Cell Biol, 2018. 19(4): p. 262-274.
  26. Galbraith, M.D., A.J. Donner, and J.M. Espinosa, CDK8: a positive regulator of transcription. Transcription, 2010. 1(1): p. 4-12.
  27. Fant, C.B. and D.J. Taatjes, Regulatory functions of the Mediator kinases CDK8 and CDK19. Transcription, 2019. 10(2): p. 76-90. [CrossRef]
  28. Bancerek, J., et al., CDK8 kinase phosphorylates transcription factor STAT1 to selectively regulate the interferon response. Immunity, 2013. 38(2): p. 250-62. [CrossRef]
  29. Steinparzer, I., et al., Transcriptional Responses to IFN-γ Require Mediator Kinase-Dependent Pause Release and Mechanistically Distinct CDK8 and CDK19 Functions. Molecular Cell, 2019. 76(3): p. 485-499.e8. [CrossRef]
  30. Alarcon, C., et al., Nuclear CDKs drive Smad transcriptional activation and turnover in BMP and TGF-beta pathways. Cell, 2009. 139(4): p. 757-69. [CrossRef]
  31. Morris, E.J., et al., E2F1 represses β-catenin transcription and is antagonized by both pRB and CDK8. Nature, 2008. 455(7212): p. 552-556. [CrossRef]
  32. Zhao, X., et al., Regulation of lipogenesis by cyclin-dependent kinase 8-mediated control of SREBP-1. J Clin Invest, 2012. 122(7): p. 2417-27. [CrossRef]
  33. Hirst, M., et al., GAL4 is regulated by the RNA polymerase II holoenzyme-associated cyclin-dependent protein kinase SRB10/CDK8. Mol Cell, 1999. 3(5): p. 673-8.
  34. Vincent, O., et al., Interaction of the Srb10 kinase with Sip4, a transcriptional activator of gluconeogenic genes in Saccharomyces cerevisiae. Mol Cell Biol, 2001. 21(17): p. 5790-6.
  35. Cooper, K.F., Cargo hitchhiking autophagy - a hybrid autophagy pathway utilized in yeast. Autophagy, 2025: p. 1-13. [CrossRef]
  36. Tsai, K.L., et al., A conserved Mediator-CDK8 kinase module association regulates Mediator-RNA polymerase II interaction. Nat Struct Mol Biol, 2013. 20(5): p. 611-9. [CrossRef]
  37. Allen, B.L. and D.J. Taatjes, The Mediator complex: a central integrator of transcription. Nat Rev Mol Cell Biol, 2015. 16(3): p. 155-66. [CrossRef]
  38. Robinson, P.J., et al., Molecular architecture of the yeast Mediator complex. Elife, 2015. 4. [CrossRef]
  39. Li, T., T.C. Chao, and K.L. Tsai, Structures and compositional dynamics of Mediator in transcription regulation. Curr Opin Struct Biol, 2024. 88: p. 102892. [CrossRef]
  40. Chao, T.C., et al., Structural basis of the human transcriptional Mediator regulated by its dissociable kinase module. Mol Cell, 2024. 84(20): p. 3932-3949.e10. [CrossRef]
  41. Elmlund, H., et al., The cyclin-dependent kinase 8 module sterically blocks Mediator interactions with RNA polymerase II. Proc Natl Acad Sci U S A, 2006. 103(43): p. 15788-93. [CrossRef]
  42. Knuesel, M.T., et al., The human CDK8 subcomplex is a histone kinase that requires Med12 for activity and can function independently of Mediator. Mol Cell Biol, 2009. 29(3): p. 650-61. [CrossRef]
  43. Osman, S., et al., The Cdk8 kinase module regulates interaction of the mediator complex with RNA polymerase II. J Biol Chem, 2021. 296: p. 100734. [CrossRef]
  44. Petrenko, N., et al., Mediator Undergoes a Compositional Change during Transcriptional Activation. Mol Cell, 2016. 64(3): p. 443-454454. [CrossRef]
  45. Davis, M.A., et al., The SCF-Fbw7 ubiquitin ligase degrades MED13 and MED13L and regulates CDK8 module association with Mediator. Genes Dev, 2013. 27(2): p. 151-6. [CrossRef]
  46. Mo, X., et al., Ras induces mediator complex exchange on C/EBP beta. Mol Cell, 2004. 13(2): p. 241-50.
  47. Pavri, R., et al., PARP-1 determines specificity in a retinoid signaling pathway via direct modulation of Mediator. Mol Cell, 2005. 18(1): p. 83-96. [CrossRef]
  48. Lambert, E., et al., From structure to molecular condensates: emerging mechanisms for Mediator function. FEBS J, 2023. 290(2): p. 286-309. [CrossRef]
  49. Holstege, F.C., et al., Dissecting the regulatory circuitry of a eukaryotic genome. Cell, 1998. 95(5): p. 717-28. [CrossRef]
  50. Anandhakumar, J., et al., Evidence for Multiple Mediator Complexes in Yeast Independently Recruited by Activated Heat Shock Factor. Mol Cell Biol, 2016. 36(14): p. 1943-60. [CrossRef]
  51. Donner, A.J., et al., CDK8 is a positive regulator of transcriptional elongation within the serum response network. Nat Struct Mol Biol, 2010. 17(2): p. 194-201. [CrossRef]
  52. Galbraith, M.D., et al., HIF1A employs CDK8-mediator to stimulate RNAPII elongation in response to hypoxia. Cell, 2013. 153(6): p. 1327-39. [CrossRef]
  53. Richter, W.F., et al., The Mediator complex as a master regulator of transcription by RNA polymerase II. Nat Rev Mol Cell Biol, 2022. 23(11): p. 732-749. [CrossRef]
  54. Cooper, K.F., et al., Stress and developmental regulation of the yeast C-type cyclin Ume3p (Srb11p/Ssn8p). EMBO J, 1997. 16(15): p. 4665-75. [CrossRef]
  55. Stieg, D.C., K.F. Cooper, and R. Strich, The extent of cyclin C promoter occupancy directs changes in stress-dependent transcription. J Biol Chem, 2020. 295(48): p. 16280-16291. [CrossRef]
  56. Li, Y.C., et al., Structure and noncanonical Cdk8 activation mechanism within an Argonaute-containing Mediator kinase module. Sci Adv, 2021. 7(3). [CrossRef]
  57. Jeronimo, C. and F. Robert, The Mediator Complex: At the Nexus of RNA Polymerase II Transcription. Trends Cell Biol, 2017. 27(10): p. 765-783. [CrossRef]
  58. Echalier, A., J.A. Endicott, and M.E. Noble, Recent developments in cyclin-dependent kinase biochemical and structural studies. Biochim Biophys Acta, 2010. 1804(3): p. 511-9. [CrossRef]
  59. Schneider, E.V., et al., The structure of CDK8/CycC implicates specificity in the CDK/cyclin family and reveals interaction with a deep pocket binder. J Mol Biol, 2011. 412(2): p. 251-66. [CrossRef]
  60. Gonzalez, D., et al., Suppression of Mediator is regulated by Cdk8-dependent Grr1 turnover of the Med3 coactivator. Proc Natl Acad Sci U S A, 2014. 111(7): p. 2500-5. [CrossRef]
  61. Fryer, C.J., J.B. White, and K.A. Jones, Mastermind recruits CycC:CDK8 to phosphorylate the Notch ICD and coordinate activation with turnover. Mol Cell, 2004. 16(4): p. 509-20. [CrossRef]
  62. Alber, F., et al., The molecular architecture of the nuclear pore complex. Nature, 2007. 450(7170): p. 695-701. [CrossRef]
  63. Adler, A.S., et al., CDK8 maintains tumor dedifferentiation and embryonic stem cell pluripotency. Cancer Res, 2012. 72(8): p. 2129-39.
  64. Chen, M., et al., CDK8/19 Mediator kinases potentiate induction of transcription by NFkappaB. Proc Natl Acad Sci U S A, 2017. 114(38): p. 10208-10213.
  65. Hirst, K., et al., The transcription factor, the Cdk, its cyclin and their regulator: directing the transcriptional response to a nutritional signal. Embo j, 1994. 13(22): p. 5410-20. [CrossRef]
  66. Lenssen, E., et al., The Ccr4-not complex regulates Skn7 through Srb10 kinase. Eukaryot Cell, 2007. 6(12): p. 2251-9. [CrossRef]
  67. Freitas, K.A., et al., Enhanced T cell effector activity by targeting the Mediator kinase module. Science, 2022. 378(6620): p. eabn5647. [CrossRef]
  68. Chen, M., et al., CDK8 and CDK19: positive regulators of signal-induced transcription and negative regulators of Mediator complex proteins. Nucleic Acids Research, 2023. 51(14): p. 7288-7313. [CrossRef]
  69. Stieg, D.C., et al., Cyclin C Regulated Oxidative Stress Responsive Transcriptome in Mus musculus Embryonic Fibroblasts. G3 (Bethesda), 2019. 9(6): p. 1901-1908.
  70. Hengartner, C.J., et al., Temporal regulation of RNA polymerase II by Srb10 and Kin28 cyclin-dependent kinases. Mol Cell, 1998. 2(1): p. 43-53. [CrossRef]
  71. Cooper, K.F. and R. Strich, Functional analysis of the Ume3p/ Srb11p-RNA polymerase II holoenzyme interaction. Gene Expr, 1999. 8(1): p. 43-57. [CrossRef]
  72. Kim, T.W., et al., MED16 and MED23 of Mediator are coactivators of lipopolysaccharide- and heat-shock-induced transcriptional activators. Proc Natl Acad Sci U S A, 2004. 101(33): p. 12153-8.
  73. Kuchin, S., P. Yeghiayan, and M. Carlson, Cyclin-dependent protein kinase and cyclin homologs SSN3 and SSN8 contribute to transcriptional control in yeast. Proc Natl Acad Sci U S A, 1995. 92(9): p. 4006-10. [CrossRef]
  74. Cooper, K.F., M.J. Mallory, and R. Strich, Oxidative stress-induced destruction of the yeast C-type cyclin Ume3p requires phosphatidylinositol-specific phospholipase C and the 26S proteasome. Mol Cell Biol, 1999. 19(5): p. 3338-48. [CrossRef]
  75. Cooper, K.F. and R. Strich, Saccharomyces cerevisiae C-type cyclin Ume3p/Srb11p is required for efficient induction and execution of meiotic development. Eukaryot Cell, 2002. 1(1): p. 66-74. [CrossRef]
  76. Wahi, M. and A.D. Johnson, Identification of genes required for alpha 2 repression in Saccharomyces cerevisiae. Genetics, 1995. 140: p. 79-90. [CrossRef]
  77. Bjorklund, S. and C.M. Gustafsson, The yeast Mediator complex and its regulation. Trends Biochem Sci, 2005. 30(5): p. 240-4. [CrossRef]
  78. van de Peppel, J., et al., Mediator expression profiling epistasis reveals a signal transduction pathway with antagonistic submodules and highly specific downstream targets. Mol Cell, 2005. 19(4): p. 511-22. [CrossRef]
  79. Surosky, R.T., R. Strich, and R.E. Esposito, The yeast UME5 gene regulates the stability of meiotic mRNAs in response to glucose. Mol Cell Biol, 1994. 14(5): p. 3446-58.
  80. Strich, R., M.R. Slater, and R.E. Esposito, Identification of negative regulatory genes that govern the expression of early meiotic genes in yeast. Proc. Natl. Acad. Sci. USA, 1989. 86: p. 10018-10022. [CrossRef]
  81. Willis, S.D., et al., Cyclin C-Cdk8 Kinase Phosphorylation of Rim15 Prevents the Aberrant Activation of Stress Response Genes. Front Cell Dev Biol, 2022. 10: p. 867257. [CrossRef]
  82. Hanley, S.E., S.D. Willis, and K.F. Cooper, Snx4-assisted vacuolar targeting of transcription factors defines a new autophagy pathway for controlling ATG expression. Autophagy, 2021. 17(11): p. 3547-3565. [CrossRef]
  83. Cooper, K.F., et al., Oxidative-stress-induced nuclear to cytoplasmic relocalization is required for Not4-dependent cyclin C destruction. J Cell Sci, 2012. 125(Pt 4): p. 1015-26. [CrossRef]
  84. Cooper, K.F., et al., Stress-induced nuclear-to-cytoplasmic translocation of cyclin C promotes mitochondrial fission in yeast. Dev Cell, 2014. 28(2): p. 161-73. [CrossRef]
  85. Wang, K., et al., Cyclin C mediates stress-induced mitochondrial fission and apoptosis. Mol Biol Cell, 2015. 26(6): p. 1030-43. [CrossRef]
  86. Ganesan, V., et al., Cyclin C directly stimulates Drp1 GTP affinity to mediate stress-induced mitochondrial hyperfission. Mol Biol Cell, 2019. 30(3): p. 302-311ctly stimulates Drp1 GTP affinity to mediate stress-induced mitochondrial hyperfission. Mol Biol Cell, 2019. 30(3): p. 302-311. [CrossRef]
  87. Jezek, J., et al., Mitochondrial translocation of cyclin C stimulates intrinsic apoptosis through Bax recruitment. EMBO Rep, 2019. 20(9): p. e47425. [CrossRef]
  88. Ježek, J., et al., Cyclin C: The Story of a Non-Cycling Cyclin. Biology (Basel), 2019. 8(1). [CrossRef]
  89. Hanley, S.E., et al., Med13 is required for efficient P-body recruitment and autophagic degradation of Edc3 following nitrogen starvation. Mol Biol Cell, 2024. 35(11): p. ar142. [CrossRef]
  90. Hanley, S.E., et al., Ksp1 is an autophagic receptor protein for the Snx4-assisted autophagy of Ssn2/Med13. Autophagy, 2024. 20(2): p. 397-415. [CrossRef]
  91. Friedson, B., et al., The CDK8 kinase module: A novel player in the transcription of translation initiation and ribosomal genes. Mol Biol Cell, 2025. 36(1): p. ar2.
  92. Shore, D. and B. Albert, Ribosome biogenesis and the cellular energy economy. Curr Biol, 2022. 32(12): p. R611-r61711-r617. [CrossRef]
  93. Hirai, H. and K. Ohta, Comparative Research: Regulatory Mechanisms of Ribosomal Gene Transcription in Saccharomyces cerevisiae and Schizosaccharomyces pombe. Biomolecules, 2023. 13(2). [CrossRef]
  94. Levin, D.E., Regulation of cell wall biogenesis in Saccharomyces cerevisiae: the cell wall integrity signaling pathway. Genetics, 2011. 189(4): p. 1145-75. [CrossRef]
  95. Krasley, E., et al., Regulation of the oxidative stress response through Slt2p-dependent destruction of cyclin C in Saccharomyces cerevisiae. Genetics, 2006. 172(3): p. 1477-86. [CrossRef]
  96. Jin, C., et al., The cell wall sensors Mtl1, Wsc1, and Mid2 are required for stress-induced nuclear to cytoplasmic translocation of cyclin C and programmed cell death in yeast. Oxid Med Cell Longev, 2013. 2013: p. 320823. [CrossRef]
  97. Jin, C., R. Strich, and K.F. Cooper, Slt2p phosphorylation induces cyclin C nuclear-to-cytoplasmic translocation in response to oxidative stress. Mol Biol Cell, 2014. 25(8): p. 1396-407. [CrossRef]
  98. Stieg, D.C., et al., A complex molecular switch directs stress-induced cyclin C nuclear release through SCF(Grr1)-mediated degradation of Med13. Mol Biol Cell, 2018. 29(3): p. 363-375. [CrossRef]
  99. Khakhina, S., K.F. Cooper, and R. Strich, Med13p prevents mitochondrial fission and programmed cell death in yeast through nuclear retention of cyclin C. Mol Biol Cell, 2014. 25(18): p. 2807-16. [CrossRef]
  100. Nash, P., et al., Multisite phosphorylation of a CDK inhibitor sets a threshold for the onset of DNA replication. Nature, 2001. 414(6863): p. 514-21. [CrossRef]
  101. Ang, X.L. and J. Wade Harper, SCF-mediated protein degradation and cell cycle control. Oncogene, 2005. 24(17): p. 2860-70. [CrossRef]
  102. Hong, S.P. and M. Carlson, Regulation of snf1 protein kinase in response to environmental stress. J Biol Chem, 2007. 282(23): p. 16838-45. [CrossRef]
  103. Willis, S.D., et al., Snf1 cooperates with the CWI MAPK pathway to mediate the degradation of Med13 following oxidative stress. Microbial Cell, 2018. 5(8): p. 357-370ith the CWI MAPK pathway to mediate the degradation of Med13 following oxidative stress. Microbial Cell, 2018. 5(8): p. 357-370. [CrossRef]
  104. Feng, Y., et al., Interplay of energy metabolism and autophagy. Autophagy, 2024. 20(1): p. 4-14.
  105. Willis, S.D., et al., Ubiquitin-proteasome-mediated cyclin C degradation promotes cell survival following nitrogen starvation. Mol Biol Cell, 2020. 31(10): p. 1015-1031. [CrossRef]
  106. Rambold, A.S., et al., Tubular network formation protects mitochondria from autophagosomal degradation during nutrient starvation. Proc Natl Acad Sci U S A, 2011. 108(25): p. 10190-5. [CrossRef]
  107. Torres, J., et al., Regulation of the cell integrity pathway by rapamycin-sensitive TOR function in budding yeast. J Biol Chem, 2002. 277(45): p. 43495-504. [CrossRef]
  108. ežek, J., K.F. Cooper, and R. Strich, Reactive Oxygen Species and Mitochondrial Dynamics: The Yin and Yang of Mitochondrial Dysfunction and Cancer Progression. Antioxidants (Basel), 2018. 7(1). [CrossRef]
  109. Tao, W., C. Kurschner, and J.I. Morgan, Modulation of Cell Death in Yeast by the Bcl-2 Family of Proteins*. Journal of Biological Chemistry, 1997. 272(24): p. 15547-15552. [CrossRef]
  110. Bui, H.T., et al., A novel motif in the yeast mitochondrial dynamin Dnm1 is essential for adaptor binding and membrane recruitment. J Cell Biol, 2012. 199(4): p. 613-22. [CrossRef]
  111. Guo, Q., et al., The mitochondrial fission adaptors Caf4 and Mdv1 are not functionally equivalent. PLoS One, 2012. 7(12): p. e53523. [CrossRef]
  112. Kraus, F., et al., Function and regulation of the divisome for mitochondrial fission. Nature, 2021. 590(7844): p. 57-66. [CrossRef]
  113. Bleazard, W., et al., The dynamin-related GTPase Dnm1 regulates mitochondrial fission in yeast. Nat Cell Biol, 1999. 1(5): p. 298-304al fission in yeast. Nat Cell Biol, 1999. 1(5): p. 298-304. [CrossRef]
  114. Zou, W., et al., Application of super-resolution microscopy in mitochondria-dynamic diseases. Adv Drug Deliv Rev, 2023. 200: p. 115043. [CrossRef]
  115. Ingerman, E., et al., Dnm1 forms spirals that are structurally tailored to fit mitochondria. J Cell Biol, 2005. 170(7): p. 1021-7. [CrossRef]
  116. Mears, J.A., et al., Conformational changes in Dnm1 support a contractile mechanism for mitochondrial fission. Nat Struct Mol Biol, 2011. 18(1): p. 20-6. [CrossRef]
  117. Mehta, K., et al., Association of mitochondria with microtubules inhibits mitochondrial fission by precluding assembly of the fission protein Dnm1. J Biol Chem, 2019. 294(10): p. 3385-3396. [CrossRef]
  118. Ashrafi, G. and T.L. Schwarz, The pathways of mitophagy for quality control and clearance of mitochondria. Cell Death Differ, 2013. 20(1): p. 31-42. [CrossRef]
  119. Fannjiang, Y., et al., Mitochondrial fission proteins regulate programmed cell death in yeast. Genes Dev, 2004. 18(22): p. 2785-97. [CrossRef]
  120. Zheng, F., et al., Glucose starvation induces mitochondrial fragmentation depending on the dynamin GTPase Dnm1/Drp1 in fission yeast. J Biol Chem, 2019. 294(47): p. 17725-17734. [CrossRef]
  121. Bauer, B.L., et al., Disease-associated mutations in Drp1 have fundamentally different effects on the mitochondrial fission machinery. Hum Mol Genet, 2023. 32(12): p. 1975-1987.
  122. Oliver, D. and P.H. Reddy, Dynamics of Dynamin-Related Protein 1 in Alzheimer's Disease and Other Neurodegenerative Diseases. Cells, 2019. 8(9). [CrossRef]
  123. Banerjee, R., et al., Mimicking human Drp1 disease-causing mutations in yeast Dnm1 reveals altered mitochondrial dynamics. Mitochondrion, 2021. 59: p. 283-295. [CrossRef]
  124. Woo, S.M., K.J. Min, and T.K. Kwon, Inhibition of Drp1 Sensitizes Cancer Cells to Cisplatin-Induced Apoptosis through Transcriptional Inhibition of c-FLIP Expression. Molecules, 2020. 25(24). [CrossRef]
  125. Deng, Y., et al., Mitochondrial Dynamin-Related Protein Drp1: a New Player in Cardio-oncology. Curr Oncol Rep, 2022. 24(12): p. 1751-1763. [CrossRef]
  126. Xu, R., et al., PCSK9 increases vulnerability of carotid plaque by promoting mitochondrial dysfunction and apoptosis of vascular smooth muscle cells. CNS Neurosci Ther, 2024. 30(2): p. e14640. [CrossRef]
  127. Guo, Y.W., et al., Ruxolitinib induces apoptosis and pyroptosis of anaplastic thyroid cancer via the transcriptional inhibition of DRP1-mediated mitochondrial fission. Cell Death Dis, 2024. 15(2): p. 125. [CrossRef]
  128. Tsushima, K., et al., Mitochondrial Reactive Oxygen Species in Lipotoxic Hearts Induce Post-Translational Modifications of AKAP121, DRP1, and OPA1 That Promote Mitochondrial Fission. Circ Res, 2018. 122(1): p. 58-73. [CrossRef]
  129. Chang, X., et al., ROS-Drp1-mediated mitochondria fission contributes to hippocampal HT22 cell apoptosis induced by silver nanoparticles. Redox Biol, 2023. 63: p. 102739. [CrossRef]
  130. Cid-Castro, C. and J. Morán, Differential ROS-Mediated Phosphorylation of Drp1 in Mitochondrial Fragmentation Induced by Distinct Cell Death Conditions in Cerebellar Granule Neurons. Oxid Med Cell Longev, 2021. 2021: p. 8832863. [CrossRef]
  131. Carmona-Gutierrez, D., et al., Guidelines and recommendations on yeast cell death nomenclature. Microb Cell, 2018. 5(1): p. 4-31. [CrossRef]
  132. Madeo, F., et al., A caspase-related protease regulates apoptosis in yeast. Mol Cell, 2002. 9(4): p. 911-7. [CrossRef]
  133. Shrestha, A., et al., The metacaspase Yca1 maintains proteostasis through multiple interactions with the ubiquitin system. Cell Discov, 2019. 5: p. 6. [CrossRef]
  134. Lee, R.E., et al., Metacaspase Yca1 is required for clearance of insoluble protein aggregates. Proc Natl Acad Sci U S A, 2010. 107(30): p. 13348-53. [CrossRef]
  135. Lam, D.K. and G. Sherlock, Yca1 metacaspase: diverse functions determine how yeast live and let die. FEMS Yeast Res, 2023. 23. [CrossRef]
  136. Strich, R. and K.F. Cooper, The dual role of cyclin C connects stress regulated gene expression to mitochondrial dynamics. Microb Cell, 2014. 1(10): p. 318-324l role of cyclin C connects stress regulated gene expression to mitochondrial dynamics. Microb Cell, 2014. 1(10): p. 318-324. [CrossRef]
  137. Moll, U.M., et al., Transcription-independent pro-apoptotic functions of p53. Curr Opin Cell Biol, 2005. 17(6): p. 631-6. [CrossRef]
  138. Karbowski, M., et al., Role of Bax and Bak in mitochondrial morphogenesis. Nature, 2006. 443(7112): p. 658-62. [CrossRef]
  139. Wilkinson, D. and M. Ramsdale, Proteases and caspase-like activity in the yeast Saccharomyces cerevisiae. Biochem Soc Trans, 2011. 39(5): p. 1502-8. [CrossRef]
  140. Mentel, M., et al., Yeast Bax Inhibitor (Bxi1p/Ybh3p) Is Not Required for the Action of Bcl-2 Family Proteins on Cell Viability. Int J Mol Sci, 2023. 24(15). [CrossRef]
  141. Qian, S., et al., The role of BCL-2 family proteins in regulating apoptosis and cancer therapy. Front Oncol, 2022. 12: p. 985363. [CrossRef]
  142. Pena-Blanco, A. and A.J. Garcia-Saez, Bax, Bak and beyond - mitochondrial performance in apoptosis. FEBS J, 2018. 285(3): p. 416-431J, 2018. 285(3): p. 416-431. [CrossRef]
  143. Jenner, A., et al., DRP1 interacts directly with BAX to induce its activation and apoptosis. EMBO J, 2022. 41(8): p. e108587.
  144. Sato, T., et al., Interactions among members of the Bcl-2 protein family analyzed with a yeast two-hybrid system. Proc Natl Acad Sci U S A, 1994. 91(20): p. 9238-42. [CrossRef]
  145. Camougrand, N., et al., The product of the UTH1 gene, required for Bax-induced cell death in yeast, is involved in the response to rapamycin. Mol Microbiol, 2003. 47(2): p. 495-506. [CrossRef]
  146. Ritch, J.J., et al., The Saccharomyces SUN gene, UTH1, is involved in cell wall biogenesis. FEMS Yeast Res, 2010. 10(2): p. 168-76es, 2010. 10(2): p. 168-76.
  147. Eid, R., et al., Identification of human ferritin, heavy polypeptide 1 (FTH1) and yeast RGI1 (YER067W) as pro-survival sequences that counteract the effects of Bax and copper in Saccharomyces cerevisiae. Exp Cell Res, 2016. 342(1): p. 52-61. [CrossRef]
  148. Wang, Y., et al., Yeast Bxi1/Ybh3 mediates conserved mitophagy and apoptosis in yeast and mammalian cells: convergence in Bcl-2 family. Biol Chem, 2024. [CrossRef]
  149. Buttner, S., et al., A yeast BH3-only protein mediates the mitochondrial pathway of apoptosis. EMBO J, 2011. 30(14): p. 2779-92.
  150. Manon, S., B. Chaudhuri, and M. Guerin, Release of cytochrome c and decrease of cytochrome c oxidase in Bax-expressing yeast cells, and prevention of these effects by coexpression of Bcl-xL. FEBS Lett, 1997. 415(1): p. 29-32. [CrossRef]
  151. Lhuissier, C., et al., Mitochondrial F0F1-ATP synthase governs the induction of mitochondrial fission. iScience, 2024. 27(5): p. 109808.
  152. Matsuyama, S., et al., The Mitochondrial F0F1-ATPase Proton Pump Is Required for Function of the Proapoptotic Protein Bax in Yeast and Mammalian Cells. Molecular Cell, 1998. 1(3): p. 327-336. [CrossRef]
  153. Légiot, A., et al., Mitochondria-Associated Membranes (MAMs) are involved in Bax mitochondrial localization and cytochrome c release. Microb Cell, 2019. 6(5): p. 257-266. [CrossRef]
  154. Toth, A., et al., Membrane-tethering of cytochrome c accelerates regulated cell death in yeast. Cell Death Dis, 2020. 11(9): p. 722. [CrossRef]
  155. Anderson, P. and N. Kedersha, RNA granules: post-transcriptional and epigenetic modulators of gene expression. Nat Rev Mol Cell Biol, 2009. 10(6): p. 430-6 RNA granules: post-transcriptional and epigenetic modulators of gene expression. Nat Rev Mol Cell Biol, 2009. 10(6): p. 430-6. [CrossRef]
  156. Brangwynne, C.P., Phase transitions and size scaling of membrane-less organelles. J Cell Biol, 2013. 203(6): p. 875-81. [CrossRef]
  157. Sheth, U. and R. Parker, Decapping and decay of messenger RNA occur in cytoplasmic processing bodies. Science, 2003. 300(5620): p. 805-8. [CrossRef]
  158. Luo, Y., Z. Na, and S.A. Slavoff, P-Bodies: Composition, Properties, and Functions. Biochemistry, 2018. 57(17): p. 2424-2431.
  159. Wang, C., et al., Context-dependent deposition and regulation of mRNAs in P-bodies. eLife, 2018. 7: p. e29815 e29815. [CrossRef]
  160. Standart, N. and D. Weil, P-Bodies: Cytosolic Droplets for Coordinated mRNA Storage. Trends Genet, 2018. 34(8): p. 612-626. [CrossRef]
  161. Horvathova, I., et al., The Dynamics of mRNA Turnover Revealed by Single-Molecule Imaging in Single Cells. Mol Cell, 2017. 68(3): p. 615-625 e9. [CrossRef]
  162. Hubstenberger, A., et al., P-Body Purification Reveals the Condensation of Repressed mRNA Regulons. Mol Cell, 2017. 68(1): p. 144-157.e54-157.e5. [CrossRef]
  163. Kroschwald, S., et al., Promiscuous interactions and protein disaggregases determine the material state of stress-inducible RNP granules. eLife, 2015. 4: p. e06807. [CrossRef]
  164. Fromm, S.A., et al., The structural basis of Edc3- and Scd6-mediated activation of the Dcp1:Dcp2 mRNA decapping complex. Embo j, 2012. 31(2): p. 279-90. [CrossRef]
  165. Fromm, S.A., et al., In vitro reconstitution of a cellular phase-transition process that involves the mRNA decapping machinery. Angew Chem Int Ed Engl, 2014. 53(28): p. 7354-9. [CrossRef]
  166. Sharif, H., et al., Structural analysis of the yeast Dhh1–Pat1 complex reveals how Dhh1 engages Pat1, Edc3 and RNA in mutually exclusive interactions. Nucleic Acids Research, 2013. 41(17): p. 8377-8390. [CrossRef]
  167. Schütz, S., E.R. Nöldeke, and R. Sprangers, A synergistic network of interactions promotes the formation of in vitro processing bodies and protects mRNA against decapping. Nucleic Acids Research, 2017. 45(11): p. 6911-6922.
  168. Decker, C.J., D. Teixeira, and R. Parker, Edc3p and a glutamine/asparagine-rich domain of Lsm4p function in processing body assembly in Saccharomyces cerevisiae. J Cell Biol, 2007. 179(3): p. 437-49. [CrossRef]
  169. Uversky, V.N., et al., Intrinsically disordered proteins as crucial constituents of cellular aqueous two phase systems and coacervates. FEBS Lett, 2015. 589(1): p. 15-22. [CrossRef]
  170. Galea, C.A., et al., Regulation of cell division by intrinsically unstructured proteins: intrinsic flexibility, modularity, and signaling conduits. Biochemistry, 2008. 47(29): p. 7598-609. [CrossRef]
  171. Wang, J.T., et al., Regulation of RNA granule dynamics by phosphorylation of serine-rich, intrinsically disordered proteins in C. elegans. Elife, 2014. 3: p. e04591. [CrossRef]
  172. Saito, M., et al., Acetylation of intrinsically disordered regions regulates phase separation. Nat Chem Biol, 2019. 15(1): p. 51-61.
  173. Isoda, T., et al., Atg45 is an autophagy receptor for glycogen, a non-preferred cargo of bulk autophagy in yeast. iScience, 2024. 27(6): p. 109810. [CrossRef]
  174. akeda, E., et al., Receptor-mediated cargo hitchhiking on bulk autophagy. Embo j, 2024. 43(15): p. 3116-3140. [CrossRef]
  175. Hanley, S.E. and K.F. Cooper, Sorting Nexins in Protein Homeostasis. Cells, 2020. 10(1). [CrossRef]
  176. Vazquez-Sanchez, S., et al., The endosomal protein sorting nexin 4 is a synaptic protein. Scientific Reports, 2020. 10(1): p. 18239. [CrossRef]
  177. Poppinga, J., et al., Endosomal sorting protein SNX4 limits synaptic vesicle docking and release. 2024, eLife Sciences Publications, Ltd. [CrossRef]
  178. Kim, N.Y., et al., Sorting nexin-4 regulates β-amyloid production by modulating β-site-activating cleavage enzyme-1. Alzheimers Res Ther, 2017. 9(1): p. 4.
  179. Cullen, P.J. and F. Steinberg, To degrade or not to degrade: mechanisms and significance of endocytic recycling. Nat Rev Mol Cell Biol, 2018. 19(11): p. 679-696. [CrossRef]
  180. Kotani, T., et al., A mechanism that ensures non-selective cytoplasm degradation by autophagy. Nat Commun, 2023. 14(1): p. 5815. [CrossRef]
  181. Nemec, A.A., et al., Autophagic clearance of proteasomes in yeast requires the conserved sorting nexin Snx4. J Biol Chem, 2017. 292(52): p. 21466-21480. [CrossRef]
  182. Makino, S., et al., Selectivity of mRNA degradation by autophagy in yeast. Nat Commun, 2021. 12(1): p. 2316. [CrossRef]
  183. Shpilka, T., et al., Fatty acid synthase is preferentially degraded by autophagy upon nitrogen starvation in yeast. Proc Natl Acad Sci U S A, 2015. 112(5): p. 1434-9. [CrossRef]
  184. Liu, X., et al., The Atg17-Atg31-Atg29 Complex Coordinates with Atg11 to Recruit the Vam7 SNARE and Mediate Autophagosome-Vacuole Fusion. Curr Biol, 2016. 26(2): p. 150-160. [CrossRef]
  185. Suzuki, H. and N.N. Noda, Biophysical characterization of Atg11, a scaffold protein essential for selective autophagy in yeast. FEBS Open Bio, 2018. 8(1): p. 110-116. [CrossRef]
  186. Lamark, T. and T. Johansen, Mechanisms of Selective Autophagy. Annu Rev Cell Dev Biol, 2021. 37: p. 143-169. [CrossRef]
  187. Fujioka, Y., et al., Phase separation organizes the site of autophagosome formation. Nature, 2020. 578(7794): p. 301-305.
  188. Noda, N.N., Z. Wang, and H. Zhang, Liquid–liquid phase separation in autophagy. Journal of Cell Biology, 2020. 219(8). [CrossRef]
  189. Yamamoto, H., et al., The Intrinsically Disordered Protein Atg13 Mediates Supramolecular Assembly of Autophagy Initiation Complexes. Dev Cell, 2016. 38(1): p. 86-99.
  190. Rogov, V.V., et al., Atg8 family proteins, LIR/AIM motifs and other interaction modes. Autophagy Rep, 2023. 2(1). [CrossRef]
  191. Laxman, S. and B.P. Tu, Multiple TORC1-associated proteins regulate nitrogen starvation-dependent cellular differentiation in Saccharomyces cerevisiae. PLoS One, 2011. 6(10): p. e26081. [CrossRef]
  192. Umekawa, M. and D.J. Klionsky, Ksp1 kinase regulates autophagy via the target of rapamycin complex 1 (TORC1) pathway. J Biol Chem, 2012. 287(20): p. 16300-10. [CrossRef]
  193. Huber, A., et al., Characterization of the rapamycin-sensitive phosphoproteome reveals that Sch9 is a central coordinator of protein synthesis. Genes Dev, 2009. 23(16): p. 1929-43. [CrossRef]
  194. Oliveira, A.P., et al., Dynamic phosphoproteomics reveals TORC1-dependent regulation of yeast nucleotide and amino acid biosynthesis. Sci Signal, 2015. 8(374): p. rs4. [CrossRef]
  195. Soulard, A., et al., The rapamycin-sensitive phosphoproteome reveals that TOR controls protein kinase A toward some but not all substrates. Mol Biol Cell, 2010. 21(19): p. 3475-86. [CrossRef]
  196. Buchan, J.R., D. Muhlrad, and R. Parker, P bodies promote stress granule assembly in Saccharomyces cerevisiae. J Cell Biol, 2008. 183(3): p. 441-55.
  197. Mitchell, S.F., et al., Global analysis of yeast mRNPs. Nat Struct Mol Biol, 2013. 20(1): p. 127-33. [CrossRef]
  198. Mason, A.C. and S.R. Wente, Functions of Gle1 are governed by two distinct modes of self-association. J Biol Chem, 2020. 295(49): p. 16813-16825. [CrossRef]
  199. Li, N., et al., Cyclin C is a haploinsufficient tumour suppressor. Nat Cell Biol, 2014. 16(11): p. 1080-91. [CrossRef]
  200. Clark, A.D., M. Oldenbroek, and T.G. Boyer, Mediator kinase module and human tumorigenesis. Crit Rev Biochem Mol Biol, 2015. 50(5): p. 393-426. [CrossRef]
  201. Trakala, M. and M. Malumbres, Cyclin C surprises in tumour suppression. Nat Cell Biol, 2014. 16(11): p. 1031-3. [CrossRef]
  202. Gharaibeh, L., et al., Notch1 in Cancer Therapy: Possible Clinical Implications and Challenges. Mol Pharmacol, 2020. 98(5): p. 559-576. [CrossRef]
  203. Koch, U. and F. Radtke, Notch signaling in solid tumors. Curr Top Dev Biol, 2010. 92: p. 411-55. [CrossRef]
  204. Hyytinen, E.R., et al., Defining the region(s) of deletion at 6q16-q22 in human prostate cancer. Genes Chromosomes Cancer, 2002. 34(3): p. 306-12. [CrossRef]
  205. Jackson, A., et al., Deletion of 6q16-q21 in human lymphoid malignancies: a mapping and deletion analysis. Cancer Res, 2000. 60(11): p. 2775-9.
  206. Sinclair, P.B., et al., A fluorescence in situ hybridization map of 6q deletions in acute lymphocytic leukemia: identification and analysis of a candidate tumor suppressor gene. Cancer Res, 2004. 64(12): p. 4089-98.
  207. Parmigiani, E., V. Taylor, and C. Giachino, Oncogenic and Tumor-Suppressive Functions of NOTCH Signaling in Glioma. Cells, 2020. 9(10). 9(10). [CrossRef]
  208. Choschzick, M., et al., NOTCH1 and PIK3CA mutation are related to HPV-associated vulvar squamous cell carcinoma. Pathol Res Pract, 2023. 251: p. 154877. [CrossRef]
  209. Jezek, J., et al., Synergistic repression of thyroid hyperplasia by cyclin C and Pten. J Cell Sci, 2019. [CrossRef]
  210. Masson, G.R. and R.L. Williams, Structural Mechanisms of PTEN Regulation. Cold Spring Harb Perspect Med, 2020. 10(3). [CrossRef]
  211. Gong, Z., et al., Targeting Nrf2 to treat thyroid cancer. Biomed Pharmacother, 2024. 173: p. 116324. [CrossRef]
  212. Firestein, R., et al., CDK8 is a colorectal cancer oncogene that regulates beta-catenin activity. Nature, 2008. 455(7212): p. 547-51. [CrossRef]
  213. Ho, T.-Y., et al., The study of a novel CDK8 inhibitor E966-0530–45418 that inhibits prostate cancer metastasis in vitro and in vivo. Biomedicine & Pharmacotherapy, 2023. 162: p. 114667. [CrossRef]
  214. Roninson, I.B., et al., Identifying Cancers Impacted by CDK8/19. Cells, 2019. 8(8). [CrossRef]
  215. Porter, D.C., et al., Cyclin-dependent kinase 8 mediates chemotherapy-induced tumor-promoting paracrine activities. Proc Natl Acad Sci U S A, 2012. 109(34): p. 13799-804. [CrossRef]
  216. rägelmann, J., et al., Pan-Cancer Analysis of the Mediator Complex Transcriptome Identifies CDK19 and CDK8 as Therapeutic Targets in Advanced Prostate Cancer. Clin Cancer Res, 2017. 23(7): p. 1829-1840. [CrossRef]
  217. Menzl, I., A. Witalisz-Siepracka, and V. Sexl, CDK8-Novel Therapeutic Opportunities. Pharmaceuticals (Basel), 2019. 12(2). [CrossRef]
  218. Kapoor, A., et al., The histone variant macroH2A suppresses melanoma progression through regulation of CDK8. Nature, 2010. 468(7327): p. 1105-9. [CrossRef]
  219. Pelish, H.E., et al., Mediator kinase inhibition further activates super-enhancer-associated genes in AML. Nature, 2015. 526(7572): p. 273-276. [CrossRef]
  220. Ayala, Y.M., et al., Structural determinants of the cellular localization and shuttling of TDP-43. J Cell Sci, 2008. 121(Pt 22): p. 3778-85. [CrossRef]
  221. Prasad, A., et al., Molecular Mechanisms of TDP-43 Misfolding and Pathology in Amyotrophic Lateral Sclerosis. Front Mol Neurosci, 2019. 12: p. 25. [CrossRef]
  222. Liu, Y.J., et al., Mitochondrial fission and fusion: A dynamic role in aging and potential target for age-related disease. Mech Ageing Dev, 2020. 186: p. 111212. [CrossRef]
  223. Schwenk, B.M., et al., TDP-43 loss of function inhibits endosomal trafficking and alters trophic signaling in neurons. EMBO J, 2016. 35(21): p. 2350-2370. [CrossRef]
  224. McDonald, K.K., et al., TAR DNA-binding protein 43 (TDP-43) regulates stress granule dynamics via differential regulation of G3BP and TIA-1. Hum Mol Genet, 2011. 20(7): p. 1400-10. [CrossRef]
  225. Osaka, M., D. Ito, and N. Suzuki, Disturbance of proteasomal and autophagic protein degradation pathways by amyotrophic lateral sclerosis-linked mutations in ubiquilin 2. Biochem Biophys Res Commun, 2016. 472(2): p. 324-31. [CrossRef]
  226. Bose, J.K., C.C. Huang, and C.K. Shen, Regulation of autophagy by neuropathological protein TDP-43. J Biol Chem, 2011. 286(52): p. 44441-8. [CrossRef]
  227. Olufunmilayo, E.O., M.B. Gerke-Duncan, and R.M.D. Holsinger, Oxidative Stress and Antioxidants in Neurodegenerative Disorders. Antioxidants (Basel), 2023. 12(2). [CrossRef]
  228. Yu, C.H., et al., TDP-43 Triggers Mitochondrial DNA Release via mPTP to Activate cGAS/STING in ALS. Cell, 2020. 183(3): p. 636-649 e18. [CrossRef]
  229. Wang, W., et al., The inhibition of TDP-43 mitochondrial localization blocks its neuronal toxicity. Nat Med, 2016. 22(8): p. 869-78. [CrossRef]
  230. Joshi, A.U., et al., Inhibition of Drp1/Fis1 interaction slows progression of amyotrophic lateral sclerosis. EMBO Mol Med, 2018. 10(3). [CrossRef]
  231. Sharifi-Rad, M., et al., Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front Physiol, 2020. 11: p. 694. [CrossRef]
  232. Jiang, L. and S.T. Ngo, Altered TDP-43 Structure and Function: Key Insights into Aberrant RNA, Mitochondrial, and Cellular and Systemic Metabolism in Amyotrophic Lateral Sclerosis. Metabolites, 2022. 12(8). [CrossRef]
  233. Goel, P., et al., Neuronal cell death mechanisms in Alzheimer's disease: An insight. Front Mol Neurosci, 2022. 15: p. 937133. [CrossRef]
  234. Bharathi, V., A. Girdhar, and B.K. Patel, Role of CNC1 gene in TDP-43 aggregation-induced oxidative stress-mediated cell death in S. cerevisiae model of ALS. Biochim Biophys Acta Mol Cell Res, 2021. 1868(6): p. 118993. [CrossRef]
  235. Riva, N., et al., Update on recent advances in amyotrophic lateral sclerosis. J Neurol, 2024. 271(7): p. 4693-4723. [CrossRef]
  236. Daniels, D.L., et al., Mutual Exclusivity of MED12/MED12L, MED13/13L, and CDK8/19Paralogs Revealed within the CDK-Mediator Kinase Module. Journal of Proteomics & Bioinformatics, 2013. 2013: p. 1-7. [CrossRef]
  237. Bessenyei, B., et al., MED13L-related intellectual disability due to paternal germinal mosaicism. Cold Spring Harb Mol Case Stud, 2022. 8(1)ud, 2022. 8(1). [CrossRef]
  238. Hamada, N., I. Iwamoto, and K.I. Nagata, MED13L and its disease-associated variants influence the dendritic development of cerebral cortical neurons in the mammalian brain. J Neurochem, 2023. 165(3): p. 334-347. [CrossRef]
  239. Siavrienė, E., et al., Molecular and Functional Characterisation of a Novel Intragenic 12q24.21 Deletion Resulting in MED13L Haploinsufficiency Syndrome. Medicina (Kaunas), 2023. 59(7). [CrossRef]
  240. Heilmann, R., et al., The MED13L Foundation strategic research plan: a roadmap to the future. Ther Adv Rare Dis, 2024. 5: p. 26330040241290252. [CrossRef]
  241. Chang, K.T., et al., Aberrant cyclin C nuclear release induces mitochondrial fragmentation and dysfunction in MED13L syndrome fibroblasts. iScience, 2022. 25(2): p. 103823. [CrossRef]
  242. Snijders Blok, L., et al., De novo mutations in MED13, a component of the Mediator complex, are associated with a novel neurodevelopmental disorder. Hum Genet, 2018ons in MED13, a component of the Mediator complex, are associated with a novel neurodevelopmental disorder. Hum Genet, 2018. [CrossRef]
  243. Polla, D.L., et al., De novo variants in MED12 cause X-linked syndromic neurodevelopmental disorders in 18 females. Genetics in Medicine, 2021. 23(4): p. 645-652. [CrossRef]
  244. Plassche, S.V. and A.P. Brouwer, MED12-Related (Neuro)Developmental Disorders: A Question of Causality. Genes (Basel), 2021. 12(5)).
  245. Luyties, O. and D.J. Taatjes, The Mediator kinase module: an interface between cell signaling and transcription. Trends Biochem Sci, 2022. 47(4): p. 314-327. [CrossRef]
  246. Calpena, E., et al., De Novo Missense Substitutions in the Gene Encoding CDK8, a Regulator of the Mediator Complex, Cause a Syndromic Developmental Disorder. Am J Hum Genet, 2019. 104(4): p. 709-720. [CrossRef]
  247. Uehara, T., et al., Pathogenesis of CDK8-associated disorder: two patients with novel CDK8 variants and in vitro and in vivo functional analyses of the variants. Sci Rep, 2020. 10(1): p. 17575. [CrossRef]
  248. Lee, M., et al., Analysis of MED12 Mutation in Multiple Uterine Leiomyomas in South Korean patients. Int J Med Sci, 2018. 15(2): p. 124-128. [CrossRef]
  249. Shamas-Din, A., et al., Mechanisms of action of Bcl-2 family proteins. Cold Spring Harb Perspect Biol, 2013. 5(4): p. a008714. [CrossRef]
  250. Gross, A. and S.G. Katz, Non-apoptotic functions of BCL-2 family proteins. Cell Death Differ, 2017. 24(8): p. 1348-1358. [CrossRef]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.
Copyright: This open access article is published under a Creative Commons CC BY 4.0 license, which permit the free download, distribution, and reuse, provided that the author and preprint are cited in any reuse.
Prerpints.org logo

Preprints.org is a free preprint server supported by MDPI in Basel, Switzerland.

Subscribe

Disclaimer

Terms of Use

Privacy Policy

Privacy Settings

© 2025 MDPI (Basel, Switzerland) unless otherwise stated