bims-proteo Biomed News
on Proteostasis
Issue of 2025–12–28
sixty-nine papers selected by
Eric Chevet, INSERM



  1. Nucleic Acids Res. 2025 Nov 26. pii: gkaf1311. [Epub ahead of print]53(22):
      Transfer RNA (tRNA) modifications tune translation rates and codon optimality, thereby optimizing co-translational protein folding. However, the mechanisms by which tRNA modifications modulate codon optimality and trigger phenotypes remain unclear. Here, we show that ribosomes stall at specific modification-dependent codon pairs in wobble uridine modification (U34) mutants. This triggers ribosome collisions and a coordinated hierarchical response of cellular quality control pathways. High-resolution ribosome profiling reveals an unexpected functional diversity of U34 modifications during decoding. For instance, 5-carbamoylmethyluridine (ncm5U) exhibits distinct effects at the A and P sites. Importantly, ribosomes only slow down at a fraction of codons decoded by hypomodified tRNA, and the decoding speed of most codons remains unaffected. However, the translation speed of a codon largely depends on the identity of A- and P-site codons. Stalling at modification-dependent codon pairs induces ribosome collisions, triggering ribosome-associated quality control (RQC) and preventing protein aggregation by degrading aberrant nascent peptides and messenger RNAs. Inactivation of RQC stimulates the expression of molecular chaperones that remove protein aggregates. Our results demonstrate that loss of tRNA modifications primarily disrupts translation rates of suboptimal codon pairs, showing the coordinated regulation and adaptability of cellular surveillance systems. These systems ensure efficient and accurate protein synthesis and maintain protein homeostasis.
    DOI:  https://doi.org/10.1093/nar/gkaf1311
  2. Nature. 2025 Dec 22.
      The nascent polypeptide-associated complex (NAC) is a conserved ribosome-bound factor with essential yet incompletely understood roles in protein biogenesis1. Here, we show that NAC is a multifaceted regulator that coordinates translation elongation, cotranslational folding, and organelle targeting through distinct interactions with nascent polypeptides both inside and outside the ribosome exit tunnel. Using NAC-selective ribosome profiling in C. elegans, we identify thousands of sequence-specific NAC binding events across the nascent proteome, revealing broad cotranslational engagement with hydrophobic and helical motifs in cytosolic, nuclear, ER, and mitochondrial proteins. Unexpectedly, we discover an intra-tunnel sensing mode, where NAC engages ribosomes with extremely short nascent polypeptides inside the exit tunnel in a sequence-specific manner. Moreover, initial NAC interactions induce an early elongation slowdown that tunes ribosome flux and prevent ribosome collisions, linking NAC's chaperone activity to kinetic control of translation. We propose NAC action protects aggregation-prone intermediates by shielding amphipathic helices, thus promoting cytonuclear folding. NAC also supports mitochondrial membrane protein biogenesis and ER targeting by early recognition of signal sequences and transmembrane domain. Our findings establish NAC as an early-acting, multifaceted orchestrator of cotranslational proteostasis, with distinct mechanisms of action on nascent chains depending on their sequence features and subcellular destinations.
    DOI:  https://doi.org/10.1038/s41586-025-10058-2
  3. FEBS Lett. 2025 Dec 26.
      During gene expression, ribosome stalling frequently occurs and can lead to detrimental effects on cellular homeostasis. Several quality control mechanisms, including ribosome-associated quality control (RQC) and nonfunctional ribosomal RNA decay (NRD), have been identified to resolve these aberrant translation events. While the molecular mechanisms of each pathway have been extensively characterized, the mechanisms underlying the mutual regulation of the expression of pathway factors remain to be elucidated. Here, we employed a series of knockout mouse and human cell lines to investigate the crosstalk between translational quality control factors. Our findings revealed that the E3 ubiquitin ligase LTN1 suppresses expression of the E3 ubiquitin ligase RNF10 in a manner dependent on the RING domain of LTN1. This discovery offers new insights into the coordination of translational surveillance pathways.
    Keywords:  LTN1; RING domain; RNF10; RQC; ribosome ubiquitination
    DOI:  https://doi.org/10.1002/1873-3468.70230
  4. Autophagy Rep. 2026 ;5(1): 2602971
      Reticulophagy regulator 1 (RETREG1)/Family with sequence similarity 134 member B (FAM134B) is a selective endoplasmic reticulum (ER)-phagy receptor that mediates starvation-induced macro-ER-phagy, but whether it participates in other pathways mediating ER turnover has remained unclear. Here, we unveil a previously unrecognized role for RETREG1 in micro-ER-phagy and show how the murine leukemia virus (MLV) accessory protein glycosylated group-specific antigen (glycoGag) exploits this pathway to antagonize the host restriction factor SERINC5 (serine incorporator 5). GlycoGag binds SERINC5 in the endoplasmic reticulum (ER) and selectively recruits RETREG1 to eliminate SERINC5 through an autophagosome-independent process that bypasses ATG3 (autophagy-related), ATG5, ATG7, BECN1 (Beclin-1), LC3 (microtubule-associated protein 1 light chain 3) lipidation, and PIK3C3 (phosphatidylinositol 3-kinase catalytic subunit type 3)/hVPS34 (vacuolar protein sorting 34). RETREG1 knockout abolishes degradation of ER-retained SERINC5, whereas endolysosomal turnover of surface SERINC5 remains partially intact, demonstrating that glycoGag utilizes dual ER-phagy and endolysosomal routes to suppress SERINC5. These findings expand the functional repertoire of RETREG1 in autophagy, identify that retroviruses repurpose micro-ER-phagy to circumvent SERINC5-mediated restriction, and reveal ER-phagy as an understudied battleground in the ongoing arms race between cellular restriction factors and viral accessory proteins.
    Keywords:  Autophagy; ER-phagy; FAM134B; RETREG1; SERINC5; glycogag; micro-ER-phagy; restriction factor; reticulophagy; retrovirus
    DOI:  https://doi.org/10.1080/27694127.2025.2602971
  5. bioRxiv. 2025 Dec 15. pii: 2025.12.12.694052. [Epub ahead of print]
      Hematopoietic stem cells (HSCs) must preserve protein homeostasis (proteostasis) despite dramatic changes in proliferative and biosynthetic demands during development, yet how proteostasis is regulated across these transitions is poorly understood. Here, we show that fetal and neonatal HSCs operate through distinct, stage-specific proteostasis programs that differ fundamentally from those in adulthood. Using quantitative in vivo assays spanning embryonic through adult stages, we uncover an unanticipated decoupling between protein synthesis and protein quality control during development, revealing that fetal and neonatal HSCs employ specialized mechanisms to safeguard proteome integrity under developmental stress. Developing HSCs experience a distinctive proteostasis landscape characterized by elevated protein synthesis, increased unfolded protein burden, and selective engagement of stress-buffering and protein degradation pathways that are largely dispensable in young adult HSCs. Disruption of these pathways compromises early life HSC function and long-term fitness, establishing proteostasis control as a key regulator of stem cell maturation. These findings define previously unrecognized mechanisms by which HSCs manage the proteome during early life and reveal fundamental principles governing stem cell proteostasis across ontogeny.
    DOI:  https://doi.org/10.64898/2025.12.12.694052
  6. bioRxiv. 2025 Dec 20. pii: 2025.12.18.695207. [Epub ahead of print]
      About one-third of the eukaryotic proteome transits the secretory pathway to reach its correct cellular or extracellular destination. At the earliest stage, transport from the endoplasmic reticulum (ER) to the ER-Golgi intermediate compartment (ERGIC) or Golgi apparatus is mediated by coat protein complex II (COPII). COPII coats consist of inner and outer layers formed by Sec23-Sec24 heterodimers and Sec13-Sec31 heterotetramers, respectively, which initially assemble at ER exit sites (ERES) to form transport carriers. Sec23-interacting protein (Sec23IP) links the inner and outer coats through its interactions with both Sec23A and Sec31A, positioning it as a key potential regulator of COPII function. However, the mechanisms controlling Sec23IP activity remain poorly understood. Here, we investigate how physiological stimuli regulate COPII function through the dynamic modification of Sec23IP by O-linked β-N-acetylglucosamine (O-GlcNAc), a reversible, intracellular form of glycosylation. We first validated Sec23IP as a bona fide O-GlcNAcylated protein. Rescue experiments in Sec23IP knockout cells with a nearly unglycosylatable mutant protein demonstrated the essential role of O-GlcNAcylation in the intrinsically disordered domain in protein transport and in recruiting Sec31A to ERES. Moreover, O-GlcNAcylation of Sec23IP increased during protein transport, coinciding with a reduction in its interaction with Sec31A. These results indicate that distinct site-specific O-GlcNAcylation of Sec23IP spatiotemporally modulates its association with Sec31A to fine-tune ERES recruitment and COPII assembly/disassembly. Our work provides new insight into Sec23IP regulation and suggests that O-GlcNAc on other COPII proteins may govern carrier formation, uncoating, and transport.
    DOI:  https://doi.org/10.64898/2025.12.18.695207
  7. J Biol Chem. 2025 Dec 18. pii: S0021-9258(25)02919-9. [Epub ahead of print] 111067
      Inositol-requiring enzyme 1 (IRE1) is one of three known sensor proteins that respond to homeostatic perturbations in the metazoan endoplasmic reticulum. The three sensors collectively initiate an intertwined signaling network called the Unfolded Protein Response (UPR). Although IRE1 plays pivotal roles in human health and development, understanding its specific contributions to the UPR remains a challenge due to signaling crosstalk from the other two stress sensors. To overcome this problem, we engineered a light-activatable version of IRE1 and probed the transcriptomic effects of IRE1 activity in isolation from the other branches of the UPR. We demonstrate that 1) oligomerization alone is sufficient to activate IRE1 in human cells, 2) IRE1's transcriptional response evolves substantially under prolonged activation, and 3) the UPR induces major changes in mRNA splice isoform abundance in an IRE1-independent manner. Our data reveal previously unknown targets of IRE1's transcriptional regulation and direct degradation. Additionally, the tools developed here will be broadly applicable for precise dissection of the UPR in diverse cell types, tissues, and organisms.
    DOI:  https://doi.org/10.1016/j.jbc.2025.111067
  8. Nat Commun. 2025 Dec 25. 16(1): 11382
      E3 ubiquitin ligases are vital enzymes that define the ubiquitin code in cells. Beyond promoting protein degradation to maintain cellular health, they also mediate non-degradative processes like DNA repair, signaling, and immunity. Despite their therapeutic potential, a comprehensive framework for understanding the relationships among diverse E3 ligases is lacking. Here, we classify the "human E3 ligome"-an extensive set of catalytic human E3s-by integrating multi-layered data, including protein sequences, domain architectures, 3D structures, functions, and expression patterns. Our classification is based on a metric-learning paradigm and uses a weakly supervised hierarchical framework to capture authentic relationships across E3 families and subfamilies. It extends the categorization of E3s into RING, HECT, and RBR classes, including non-canonical mechanisms, successfully explains their functional segregation, distinguishes between multi-subunit complexes and standalone enzymes, and maps E3s to substrates and potential drug interactions. Our analysis provides a global view of E3 biology, opening strategies for drugging E3-substrate networks, including drug repurposing and designing specific E3 handles.
    DOI:  https://doi.org/10.1038/s41467-025-67450-9
  9. Nat Commun. 2025 Dec 26.
      The leukodystrophy Pelizaeus-Merzbacher disease (PMD) is caused by myelin protein proteolipid protein gene (PLP1) mutations. PMD is characterized by oligodendrocyte death and CNS hypomyelination; thus, increasing oligodendrocyte survival and enhancing myelination could provide therapeutic benefit. Here, we use the PMD mouse model Jimpy to determine the impact of the integrated stress response (ISR) on the oligodendrocyte response to mutant PLP expression. Male Jimpy animals in which the ISR-triggering eukaryotic initiation factor (eIF) 2α kinase, protein kinase-like endoplasmic reticulum kinase (PERK), is inactivated have an extended lifespan that correlates with increased oligodendrocyte survival and enhanced CNS myelination. Inactivation of downstream components of the ISR pathway, in contrast, does not rescue oligodendrocytes or myelin. Phosphorylated eIF2α inhibits the exchange factor eIF2B, resulting in diminished protein synthesis. Treatment with small molecule eIF2B activators 2BAct and ISRIB increases oligodendrocyte survival, CNS myelination, and doubled the Jimpy lifespan. These results suggest that ISR modulation could provide therapeutic benefit to PMD patients.
    DOI:  https://doi.org/10.1038/s41467-025-68045-0
  10. Essays Biochem. 2025 Dec 22. 69(4): 379-391
      Molecular glue degraders (MGDs) are small molecules that promote interactions between an E3 ligase and a target protein, reconfiguring recognition to trigger proteasome-mediated degradation. Their discovery has so far been largely serendipitous - either recognized in retrospect or uncovered through 'needle-in-a-haystack' screening - but systematic strategies are beginning to emerge. This review frames two complementary routes for discovery. The first views MGDs as modular - typically anchored on either the ligase or the target - which allows the chemical search space to be biased toward such anchoring. Ligase-directed strategies derivatize known ligase binders, as demonstrated for cereblon (CRBN) and now beyond. Conversely, recent target-directed strategies remodel inhibitors into glues through solvent-exposed elaboration, effectively inverting the classical design paradigm. Both approaches tilt discovery toward chemotypes more likely to yield glue activity. Second, biology provides its own guideposts: certain protein pairs appear especially predisposed to stabilization. Endogenous degrons, mutational lesions, and transferable 'glueprints' of surface topology all point to contexts in which small molecules might act as functional surrogates - repairing hypomorphs, mimicking hypermorphs, or creating neomorphs. MGDs, therefore, exemplify how small molecules can reprogram recognition logic by transforming latent compatibilities into selective degradation. Together, these insights help rationalize past discoveries and suggest possible blueprints for more systematic ones ahead.
    Keywords:  drug discovery and design; medicinal chemistry; molecular glue degraders; targeted protein degradation; ubiquitin ligases
    DOI:  https://doi.org/10.1042/EBC20253058
  11. Protein Sci. 2026 Jan;35(1): e70372
      ClpXP and other AAA proteases maintain proteostasis and regulate cellular functions by degrading misfolded, incomplete, or regulatory proteins. ClpX recognizes substrates via unstructured degron sequences, typically located at the N- or C-terminus. Although five classes of degrons are known, only recognition of the ssrA tag, a C-motif-1 degron, is well understood. The ssrA tag initially binds to a conformation of hexameric ClpX in which the axial channel is closed by a pore-2 loop, with subsequent channel opening allowing translocation into and degradation by ClpP. A ClpX variant with a pore-2 loop deletion (ΔNPS) favors the open conformation and exhibits weaker binding to ssrA-tagged substrates. Here, using model substrates representing each of the five known degron classes, we show that ΔNPS ClpXP degrades low micromolar concentrations of N-motif-1, -2, -3, and C-motif-2 substrates more effectively than wild-type ClpXP. Cryo-EM analysis of wild-type ClpXP bound to an N-motif-1 substrate reveals degron engagement within an open axial channel. Our results support a model in which the open- and closed-channel conformations of ClpXP differentially enhance recognition of distinct degron classes: the open channel facilitates degradation of many natural substrates, whereas the closed channel promotes efficient recognition and degradation of ssrA-tagged proteins. We propose that the conformational equilibrium between these two states tunes ClpXP activity to balance broad substrate recognition with specificity, allowing cells to meet dynamic proteolytic demands and minimize off-target degradation.
    Keywords:  molecular machines; proteolytic activation; proteostasis regulation
    DOI:  https://doi.org/10.1002/pro.70372
  12. bioRxiv. 2025 Dec 20. pii: 2025.12.19.695558. [Epub ahead of print]
      The 26S proteasome is the endpoint of the ubiquitin-proteasome system, an essential pathway for maintaining cellular homeostasis through targeted degradation of misfolded, damaged, and obsolete proteins. Substrates labeled with ubiquitin are directed to the 26S proteasome by binding to one or more ubiquitin receptors. However, ubiquitin-dependent degradation occurs even when the canonical receptor sites are mutated, suggesting the presence of additional, unidentified binding sites. Here we created photo-crosslinkable probes for ubiquitin interactions by incorporating the unnatural amino acid p-benzoyl-L-phenylalanine into ubiquitin. We show that these probes can be used to measure apparent affinities for known receptors and to reveal novel ubiquitin-binding sites on the yeast 26S proteasome. Through photo-crosslinking mass-spectrometry experiments we identified a groove on the top of the proteasome, formed by Rpn2, Rpn9, Rpn10, and Rpn12, that serves as an additional ubiquitin-binding interface. Our photo-crosslinkable probes thus serve as versatile tools for the characterization of ubiquitin-protein interactions and the identification of ubiquitin-binding domains.
    DOI:  https://doi.org/10.64898/2025.12.19.695558
  13. Cell Syst. 2025 Dec 22. pii: S2405-4712(25)00289-3. [Epub ahead of print] 101456
      The maintenance of cellular homeostasis requires tight regulation of proteome concentration and composition. To achieve this, protein production and elimination must be robustly coordinated. However, the mechanistic basis of this coordination remains unclear. Here, we address this question using quantitative live-cell imaging, computational modeling, transcriptomics, and proteomics approaches. We found that protein decay rates systematically adapt to global alterations of protein synthesis rates. This adaptation is driven by a core passive mechanism supplemented by facultative changes in mechanistic/mammalian target of rapamycin (mTOR) signaling. Passive adaptation hinges on changes in the production rate of the machinery governing protein decay and allows for partial maintenance of the cellular proteome. Sustained changes in mTOR signaling provide an additional layer of adaptation unique to naive pluripotent stem cells, allowing for near-perfect maintenance of proteome composition. Our work unravels the mechanisms protecting the integrity of mammalian proteomes upon variations in protein synthesis rates. A record of this paper's transparent peer review process is included in the supplemental information.
    Keywords:  Bayesian inference; cell proliferation; dynamic SILAC; passive adaptation; proteasome; protein degradation; protein dilution; protein synthesis; protein turnover; proteomics; superstatistical modeling; tandem fluorescent timer
    DOI:  https://doi.org/10.1016/j.cels.2025.101456
  14. Nat Commun. 2025 Dec 23.
      Cells adapt to proteostatic and metabolic stresses, in part, through stress activated eIF2α kinases that stimulate the translation of ATF4. Stress-induced ATF4 translation is regulated through elements at ATF4 mRNA's 5' leader. In addition to eIF2α kinases, ATF4 induction requires other regulators that remain poorly understood. Here, we report an ATF4 regulatory network consisting of eIF4E-Homologous Protein (4EHP), NELF-E, the 40S ribosome, and eIF3 subunits. Specifically, we found that the mRNA cap-binding protein, 4EHP, was required for ATF4 signaling in the Drosophila larval fat body and in disease models associated with abnormal ATF4 signaling. NELF-E mRNA, encoding a regulator of pol II-mediated transcription, was identified as a top interactor of 4EHP in a TRIBE (Targets of RNA Binding through Editing) screen. Quantitative proteomics analysis revealed that the knockdown of NELF-E or 4EHP commonly reduced several subunits of the 40S ribosome (RpS) and the eIF3 translation initiation factor. Moreover, reduction of NELF-E, 4EHP, RpS12, eIF3l, or eIF3h suppressed the expression of ATF4 and its target genes. These results uncover a previously unrecognized ATF4 regulatory network consisting of 4EHP and NELF-E that impacts proteostasis during normal development and in disease models.
    DOI:  https://doi.org/10.1038/s41467-025-67357-5
  15. Mol Biol Cell. 2025 Dec 24. mbcE25040190
      Activation of the cGAS-STING pathway stimulates innate immune signaling as well as LC3B lipidation and ubiquitylation at Golgi-related vesicles upon STING trafficking. Although ubiquitylation at these subcellular sites has been associated with regulating NF-κB-related innate immune signaling, the mechanisms of Golgi-localized polyubiquitin chain regulation of immune signaling is not well understood. We report here that the ubiquitin- and LC3B-binding proteins, TNIP1 and autophagy receptors p62, NBR1, NDP52, TAX1BP1, and OPTN associate with STING-induced ubiquitin and LC3B-labeled vesicles, and that p62 and NBR1 act redundantly in spatial clustering of the LC3B-labeled vesicles in the perinuclear region. We also find that while TBK1 kinase activity is not required for the recruitment of TNIP1 and the autophagy receptors, it plays a role in sequestration of the LC3B-labeled vesicles. The ubiquitin binding domains, rather than the LC3-interacting regions, of TNIP1 and OPTN are specifically important for their recruitment to Ub/LC3B-associated perinuclear vesicles, and OPTN is also recruited through a TBK1-dependent mechanism. Functionally, we find that TNIP1 plays a role in STING-mediated innate immune signaling, acting as a negative regulator of IRF3-mediated gene expression. Together, these results highlight autophagy-independent mechanisms of autophagy receptors and TNIP1 with unanticipated roles in regulating STING-mediated innate immunity.
    DOI:  https://doi.org/10.1091/mbc.E25-04-0190
  16. Autophagy. 2025 Dec 24.
      Microautophagy involves the direct uptake of cytoplasmic materials by lysosomes, but its regulation, including substrate specificity, has remained largely unclear in mammalian cells. Microlipophagy, a form of lipid droplet microautophagy, has been suggested in mammalian cells, yet the molecular basis that links lysosomes to lipid droplets and supports their uptake has not been elucidated. In our recent study, we showed that the lysosomal membrane protein LAMP2B mediates this process via its cytoplasmic region, which can bind phosphatidic acid, a lipid present on lipid droplets. We also found that this pathway depends on the ESCRT machinery and proceeds independently of macroautophagy. In this commentary, we summarize these findings and describe how LAMP2B affects lipid droplet degradation in cells. We describe that LAMP2B overexpression protects mice from high-fat-diet-induced obesity and related disorders. We also outline a model of microautophagy and microautophagy-like processes in which LAMP2 isoforms use their cytoplasmic regions to recognize distinct cargos.
    Keywords:  Autophagy; LAMP2; LAMP2B; lipid droplet; microautophagy; microlipophagy
    DOI:  https://doi.org/10.1080/15548627.2025.2609920
  17. Autophagy. 2025 Dec 26.
      The conjugation of mammalian Atg8 (ATG8)-family proteins to membrane components is a fundamental process in membrane ATG8ylation (lipidation). While membrane ATG8ylation is well-characterized, protein ATG8ylation, the direct conjugation of ATG8 to cellular proteins, remains enigmatic. In this study, we demonstrate that protein ATG8ylation depends exclusively on ATG4, ATG3, and ATG7. We discovered that the core macroautophagy/autophagy E1 enzyme ATG7 serves a dual role: it is not only the essential E1 enzyme for protein ATG8ylation but also a key substrate. We determined that ATG7 K140 is the modification site and show that protein ATG8ylation of ATG7 forms a mono-LC3B conjugate. We demonstrated that this self-modification creates a negative-feedback loop by hindering the ATG7-ATG3 interaction, thereby attenuating autophagic flux. Our findings redefine ATG7 as a central player and regulator in the protein ATG8ylation cascade, revealing a new mechanism of autophagy regulation.
    Keywords:  ATG7; LC3; post-translational modification; protein ATG8ylation; self-regulation
    DOI:  https://doi.org/10.1080/15548627.2025.2609929
  18. bioRxiv. 2025 Dec 19. pii: 2025.12.18.695023. [Epub ahead of print]
      Cell surface receptors such as NOTCH1 must be tightly regulated to ensure developmental fidelity and prevent pathological activation. Although the proteolytic steps culminating in nuclear NOTCH1 signaling are established, how cells prevent excessive or uncontrolled activation has remained unresolved. Here we identify the autophagy-related protein UVRAG as a negative regulator of NOTCH1. Upon receptor activation, UVRAG, acting independently of autophagy, recruits and activates the E3-ligase ITCH to catalyze K27-linked ubiquitination of membrane-tethered NOTCH1, thereby licensing ESCRT-dependent lysosomal degradation. Disruption of the UVRAG-ITCH-ESCRT axis stabilizes activated NOTCH1 intermediates and amplifies oncogenic signaling. In T-cell leukemia models driven by constitutive NOTCH1 activity, restoring UVRAG expression reinstates receptor turnover, suppresses disease progression, and improves therapeutic response. These findings define a ubiquitin-directed safeguard circuit that enforces NOTCH1 signaling homeostasis and reveals a tunable axis for intervention in NOTCH1-driven cancers.
    DOI:  https://doi.org/10.64898/2025.12.18.695023
  19. Autophagy. 2025 Dec 26.
      Proteotoxic stress, arising from conditions that cause misfolded protein accumulation, is closely linked to the pathogenesis of multiple diseases. Macroautophagy/autophagy activation is considered a compensatory mechanism to maintain protein homeostasis, but the underlying regulatory mechanisms remain incompletely understood. Here, we show that proteotoxic stress induced by proteasome inhibition, puromycin treatment, or polyglutamine-expanded HTT (huntingtin) expression promotes nuclear accumulation of TFEB and TFE3, key regulators of lysosomal biogenesis and autophagy. Mechanistically, TFEB activation under proteotoxic stress occurs independently of canonical MTORC1 inactivation mediated by TSC2 or ATF4. Instead, it involves non-canonical inhibition of MTORC1 via RRAG GTPases. Proteotoxic stress disrupts the RRAGC-TFEB interaction, preventing TFEB recruitment to lysosomes and subsequent MTORC1 phosphorylation. An activated RRAGC mutant rescues impaired lysosomal localization and nuclear accumulation of TFEB, while co-overexpression of FLCN and FNIP2, a GAP for RRAGC, partially restores stress-induced TFEB dephosphorylation. In addition, proteasome inhibition activates non-canonical autophagy. Deletion of ATG16L1 or ATG5, which blocks Atg8-familyh protein lipidation and sequesters the FLCN-FNIP2 complex, partially abolishes proteotoxic stress-induced TFEB dephosphorylation and nuclear accumulation. Together, these findings demonstrate that proteotoxic stress triggers both non-canonical autophagy and TFEB-mediated canonical autophagy, with Atg8-family protein lipidation contributing to TFEB activation. Our results provide novel insights into how proteotoxic stress engages non-canonical MTORC1 inhibition and TFEB activation, thereby enhancing understanding of cellular adaptation to proteotoxic stress.
    Keywords:  Autophagy; MTORC1; RRAG GTPase; TFEB; lysosomal biogenesis; proteosome
    DOI:  https://doi.org/10.1080/15548627.2025.2608973
  20. bioRxiv. 2025 Dec 21. pii: 2025.12.18.695201. [Epub ahead of print]
      Genetic interactions can reveal gene function and identify cancer-relevant synthetic lethals, but systematic mapping in human cells is constrained by inefficient reagents, vast combinatorial search space, and diversity of cell types. Here, we leverage principles from yeast genetic networks to identify human gene modules enriched for genetic interactions. Using our Cas12a-based In4mer combinatorial knockout platform, we screen all pairwise interactions within receptor tyrosine kinase and DNA damage response modules across eight diverse cancer cell lines. We identify hundreds of unreported synthetic lethals, including a dense network within the protein glycosylation machinery, and confirm that interactions in 2D cell culture are maintained in more physiologically relevant models. Our targeted modules show up to 16-fold enrichment of interaction density, providing a scalable strategy for systematic interaction mapping.
    DOI:  https://doi.org/10.64898/2025.12.18.695201
  21. Cell Stress Chaperones. 2025 Dec 19. pii: S1355-8145(25)00087-2. [Epub ahead of print] 100142
      J-domain proteins (JDPs) are known to drive the functional specificity of Hsp70 chaperone machines. Here we report on the latest findings presented at the third international JDP workshop held in 2025 in Gdansk, Poland. Investigators from many different disciplines, including structural biology, genetics, chemical biology, translational research, computational sciences and biophysics took part in the meeting. This article includes short summaries of the seminars presented by many of the speakers, which provided exciting new insights into the chaperone-dependent and chaperone-independent functions of JDPs, some of which go beyond Hsp70-dependent functions. We also provide a revised classification of members of the (human) JDP family that emerged from open discussion at the meeting. This workshop continues to serve as the premier venue for discussions of JDP evolution, structure, function and roles in health, aging and disease.
    Keywords:  Hsp70; J-domain proteins (JDPs); functional classification
    DOI:  https://doi.org/10.1016/j.cstres.2025.100142
  22. bioRxiv. 2025 Dec 12. pii: 2025.12.09.693280. [Epub ahead of print]
      LRRK2 is implicated in lysosomal functions, but the physiological upstream cues that engage endogenous LRRK2 activity are incompletely defined. Here we show that lysosomal swelling serves as a selective and reversible trigger for LRRK2-mediated Rab phosphorylation, without requiring membrane damage. Acute inhibition of PIKfyve, but not the general disruption of phosphoinositide signaling, induces the robust accumulation of phosphorylated Rabs across endolysosomal membranes. Rescue of swelling through pharmacological restoration of lysosomal ionic imbalances from PIKfyve inhibition suppresses LRRK2 activation without restoring lysosomal function. Mechanical lysosomal swelling from indigestible osmolyte uptake causes a dose-dependent increase in LRRK2-mediated Rab phosphorylation on both swollen and non-swollen lysosomes. Together, these findings identify LRRK2 as a sensor of lysosomal volume and mechanical stress, not specifically membrane damage or PIKfyve inhibition. As lysosomal swelling is a shared pathological feature across LRRK2 -linked diseases, these results reframe LRRK2 as part of an endolysosomal surveillance system responsive to lysosomal distension.
    DOI:  https://doi.org/10.64898/2025.12.09.693280
  23. Proteomes. 2025 Nov 25. pii: 63. [Epub ahead of print]13(4):
       BACKGROUND: Multiple myeloma (MM) is essentially an incurable cancer, but treatments with proteasome inhibitors are widely used clinically to extend patient survival. While the mechanisms of proteasome inhibition by Bortezomib are well known, the cellular responses to this proteotoxic stress that leads to sensitivity by MM are not fully elucidated. This study reports on the application of an emerging method to investigate proteostasis by proteomics.
    METHODS: We utilized metabolic labeling with azidohomoalanine (AHA) in a MM cell line in combination with Bortezomib treatment. AHA labeling facilitates the selective isolation and identification of proteins for investigations of protein synthesis or protein degradation.
    RESULTS: The data collected reveals significant changes in gene protein synthesis upon Bortezomib treatment, including protein neddylation. The data also reveals a global increase in protein degradation, which suggests the induction of an autophagy-related process. The resulting data collected reveals significant changes upon Bortezomib treatment in protein synthesis of genes, including protein neddylation, and protein degradation data reveals a global increase in protein degradation, suggesting an induction of an autophagy-related process. Subsequent cellular and proteomic analysis investigated the additional treatment of an autophagy inhibitor, hydroxychloroquine, in combination with Bortezomib treatment by label-free proteomics to further characterize the proteome-wide changes in these two proteotoxic stresses.
    CONCLUSIONS: AHA metabolic labeling proteomics to investigate protein synthesis and degradation enables novel complementary insights into complex cellular responses compared to that of traditional label-free proteomics.
    Keywords:  azidohomoalanine; proteasome; protein degradation; protein synthesis; proteomics; proteostasis
    DOI:  https://doi.org/10.3390/proteomes13040063
  24. bioRxiv. 2025 Dec 08. pii: 2025.12.08.693056. [Epub ahead of print]
      FicD regulates Unfolded Protein Response (UPR) through reversible AMPylation and deAMPylation of BiP, an HSP70 chaperone and master regulator of the UPR. FicD activity is regulated by ER-stress, catalyzing BiP AMPylation under low stress conditions to hold inactive chaperone in reserve. In stressed cells, FicD deAMPylates BiP, acutely increasing its active pool to assist in protein folding. Variants in UPR machinery, including those in the FicD gene, are linked to hereditary diseases. Despite the known role of FicD in UPR, in-vivo regulation of its activity remains elusive, and identifying metabolites that alter FicD activity could prove useful pharmaceutically. We applied an unbiased high-throughput screening platform, known as M ass spectrometry Integrated with equilibrium D ialysis for the discovery of A llostery S ystematically (MIDAS), to identify novel small molecule metabolites that might regulate FicD activity. MIDAS revealed interactions between FicD and two mavelonate pathway intermediates : geranyl-pyrophosphate and farnesyl-pyrophosphate. Biochemical characterization indicates that both potently inhibit FicD-mediated AMPylation and deAMPylation. The crystal structure of FicD bound to farnesyl-pyrophosphate demonstrates a competitive inhibition mechanism, with the pyrophosphate adopting the alpha and beta phosphate positions of ATP and the hydrocarbon chain filling the nucleoside pocket. FicD variants previously appeared as biochemically indistinguishable, yet lead to different human pathologies. We demonstrate farnesyl-pyrophosphate inhibits FicD R374H and FicD R374C variants implicated in causing hereditary spastic paraplegia, but not the FicD R371S variant associated with neonatal diabetes. This study furthers our understanding of FicD inhibitors and distinguishes disease causing variants, providing insight into pharmacological targeting of UPR activity.
    Significance Statement: FicD regulates UPR signaling in metazoans by fine-tuning BiP chaperone capacity. Therefore, targeting FicD activity may be a tractable method of altering UPR signaling for therapeutic benefit. We identify geranyl- and farnesyl-pyrophosphate as specific FicD inhibitors. Notably, these small molecules differentially inhibit disease-causing variants of FicD. A structure of farnesyl-pyrophosphate bound to the FicD active site helps explain the differential inhibition of pathogenic variants and provides insight into interactions that can be differentially exploited for modifying FicD activity. Their composition provides a novel chemical foundation for future drug development efforts targeting FicD activity.
    DOI:  https://doi.org/10.64898/2025.12.08.693056
  25. Mol Cell. 2025 Dec 22. pii: S1097-2765(25)00976-1. [Epub ahead of print]
      The Hsp90 molecular chaperone system is regulated by numerous co-chaperones that modulate its function. In Saccharomyces cerevisiae, most of these cofactors can be deleted without affecting viability. Of the three essential ones, only the function of Sgt1 has remained enigmatic. Our in vivo and in vitro experiments define key structural elements and determine the essential function of Sgt1 in the chaperoning of client proteins. We demonstrate that yeast Sgt1 adopts a unique binding mode, engaging primarily with the middle domain of Hsp90. Through simultaneous interaction with both Hsp90 and client proteins, Sgt1 enhances client maturation efficiency. Specifically, Sgt1 stabilizes Hsp90-client complexes and prevents their dissociation by the co-chaperone Aha1. Our findings reveal a previously unrecognized layer of Hsp90 regulation, highlighting Sgt1 as a critical modulator of chaperone cycle progression.
    Keywords:  Aha1; Hsp90; NMR spectroscopy; Sgt1; co-chaperones; glucocorticoid receptor; molecular chaperones; protein folding
    DOI:  https://doi.org/10.1016/j.molcel.2025.12.002
  26. Biochemistry. 2025 Dec 22.
      UBQLN2 is a member of the UBL-UBA domain protein family that functions as extrinsic substrate receptors for the 26S proteasome. UBQLN2 has been shown to undergo phase separation in vitro. In cells, UBQLN2 forms condensates that may be of importance for tuning protein degradation via the ubiquitin-proteasome system and potentially of relevance for UBQLN2-linked amyotrophic lateral sclerosis (ALS). Here we show that UBQLN2 is ubiquitylated on lysine residues in the N-terminal UBL domain. The C-terminal region of UBQLN2 is lysine-depleted, and we show that introducing lysine residues in this region leads to its E6AP-dependent degradation. The UBL domain critically stabilizes UBQLN2 and protects it from proteasomal degradation. Fusion of ubiquitin to the UBQLN2 N-terminus stabilizes UBQLN2 and increases its propensity for locating in puncta, indicating that ubiquitylation of the UBQLN2 UBL domain regulates abundance and localization.
    DOI:  https://doi.org/10.1021/acs.biochem.5c00619
  27. J Biol Chem. 2025 Dec 22. pii: S0021-9258(25)02936-9. [Epub ahead of print] 111084
      Sonic hedgehog (Shh) signaling orchestrates diverse developmental processes in metazoans and is implicated in numerous human diseases. While downstream signaling in recipient cells has been extensively characterized, the mechanisms governing secretion of newly synthesized Shh from producer cells remain less well understood. Building on our previous identification of a Surfeit locus protein 4 (SURF4)-to-proteoglycan (PG) relay mechanism that mediates endoplasmic reticulum (ER)-to-Golgi transport of the N-terminal Shh fragment (ShhN), we investigated ShhN export from the trans-Golgi network (TGN). We show that ShhN exits the TGN via a clathrin-dependent secretory pathway. Mechanistic analyses identify the transmembrane protein glutamate receptor 1 (GRIA1) as a key mediator: GRIA1 associates with ShhN in Golgi-derived vesicles, physically interacts with ShhN, colocalizes with ShhN after TGN exit, and is required for efficient TGN export and secretion of ShhN. Notably, the Cardin-Weintraub (CW) motif on ShhN, previously shown to engage SURF4 for ER-to-Golgi trafficking, is also essential for TGN export, and PGs are critical for the GRIA1-ShhN interaction. Furthermore, GRIA1 regulates intracellular trafficking of endogenous full length Shh and modulates Shh pathway activity in Neuro-2a (N2A) cells. Together, these findings identify GRIA1 as an important regulator of Shh TGN export and advance our understanding of the molecular mechanisms that control Shh secretion.
    DOI:  https://doi.org/10.1016/j.jbc.2025.111084
  28. Proc Natl Acad Sci U S A. 2025 Dec 30. 122(52): e2521979122
      The Niemann-Pick type 1 and 2 proteins (NPC1 and NPC2) coordinate cholesterol egress from late endosomes-lysosomes (LE/LY). Proper folding, trafficking, and localization of both NPC proteins are essential for normal LE/LY cholesterol handling. Accordingly, mutations in NPC genes cause Niemann-Pick type C (NPC) disease, a progressive neurodegenerative lysosomal cholesterol storage disorder. The routes by which NPC1 reaches the LE/LY compartment in mammalian cells are not fully elucidated. Therefore, to interrogate NPC1 trafficking, we developed genome-engineered HeLa cells expressing endogenous NPC1mNeon. We demonstrate that endogenous NPC1 localizes to the LE/LY compartment and by using protein proximity-based approaches that NPC1 resides in the same membranes as Vacuolar Protein Sorting-associated protein 41 (VPS41), one of the two unique subunits of the homotypic fusion and vacuole protein sorting complex. Loss of VPS41 increases NPC1 and Lysosomal Associated Membrane Protein 1 (LAMP1) abundance. Paradoxically, this results in marked accumulation of lysosomal cholesterol and induction of sterol regulatory element-binding protein signaling. Mechanistically, using immuno-fluorescence and electron microscopy imaging in combination with a VPS41-dependent ectopic recruitment assay, we demonstrate that this is due to a shift in the localization of NPC1 and LAMP1 from LE/LY to biosynthetic vesicles called LAMP carriers. These vesicles have been recently described to transport lysosomal-destined cargo directly from the trans-Golgi (TGN) network to LE/LY. In conclusion, we identify NPC1 as a cargo for VPS41-dependent LAMP carriers that are instrumental for the delivery of NPC1 to LE/LY and maintaining cellular cholesterol homeostasis.
    Keywords:  NPC1; VPS41; cholesterol metabolism; intracellular cholesterol transport; lysosomes
    DOI:  https://doi.org/10.1073/pnas.2521979122
  29. Biochem Soc Trans. 2025 Dec 24. 53(6): 1527-1541
      Eukaryotic translation initiation factor (eIF) 3 is a multi-subunit protein complex that plays critical roles throughout translation initiation and has been implicated in a variety of human diseases. More recently, eIF3 has been tied to translation elongation and termination, as well as translational regulation. And yet, a mechanistic understanding of how eIF3 and its constituent subunits perform their canonical roles during initiation continues to elude us. Work across the last two decades has delineated broad mechanistic roles for some of these subunits and identified distinct modules of the complex that contribute differentially to the recruitment of messenger RNA (mRNA) to the ribosome during initiation. Structural approaches have further illuminated these putative roles. And yet, key mechanistic questions tied to fundamental technical challenges remain. Even so, new developments are poised to address these challenges and push our understanding of eIF3 function forward in the coming years.
    Keywords:  MRNA; biochemistry; eukaryotic gene expression; molecular mechanisms; ribosomes; translation
    DOI:  https://doi.org/10.1042/BST20253069
  30. bioRxiv. 2025 Dec 16. pii: 2025.12.12.694063. [Epub ahead of print]
       Introduction: Proteostasis dysfunction is a hallmark of frontotemporal dementia (FTD) and Alzheimer's disease (AD), yet the genetic and molecular pathways that disrupt protein homeostasis remain poorly understood.
    Methods: We integrated human genetics, transcriptomics, and functional studies to identify proteostasis network components involved in tauopathy.
    Results: We identified 18 proteostasis network genes harboring 75 rare, damaging variants enriched in FTD and/or AD. These genes, spanning multiple proteostasis pathways, were differentially expressed in MAPT mutant neurons and dysregulated in FTD and AD brains. NINL, which encodes Nlp, emerged as the only gene consistently upregulated across all datasets. NINL overexpression reduced tau seeding and enhanced lysosomal proteolytic activity, whereas two FTD-enriched NINL frameshift variants impaired Nlp expression and abolished these protective effects.
    Discussion: Our findings identify a set of proteostasis genes with genetic and transcriptional links to neurodegeneration and reveal NINL as a novel regulator of tau aggregation, potentially upregulated as an adaptive response to proteotoxic stress.
    DOI:  https://doi.org/10.64898/2025.12.12.694063
  31. Nat Commun. 2025 Dec 21.
      Emerging evidence indicates important interconnections between autophagy and the secretion of small extracellular vesicles. However, our understanding of these secretory vesicles remains incomplete. Here, we identify a subtype of small extracellular vesicles, termed autophagic extracellular vesicles (AEVs), which are distinct from exosomes. Extracellular AEVs characterized by a size less than 100 nm exhibit increased secretion when the autophagy response is induced. Amphisomes, the hybrid organelles, are essential for the secretion of these types of vesicles. Further exploration reveals that autophagic cargos, certain ESCRT Ⅲ components and the Rab13 serve as distinctive markers for distinguishing AEVs from exosomes. Moreover, we find that the biogenesis of AEVs functionally requires components of the ESCRT Ⅲ complex and the GTPase Rab27a. Finally, we confirm that the enteroviral particles or genomes can be encapsulated into AEVs and subsequently infect receptor-negative cells with high efficiency. This model represents a prominent pattern of EV-mediated virus transmission.
    DOI:  https://doi.org/10.1038/s41467-025-67860-9
  32. Mol Plant. 2025 Dec 19. pii: S1674-2052(25)00448-4. [Epub ahead of print]
      Cellular oxidation is essential for many physiological processes but can damage redox-vulnerable proteins. Eukaryotic cells maintain millimolar levels of reduced glutathione (GSH) as a major antioxidative strategy. However, how this highly reduced pool is locally reorganized to permit necessary oxidation without triggering proteolysis remains unclear. Here, we show that protein S-glutathionylation promotes stress granule (SG) assembly, creating reductive microenvironments within an oxidized cytosol to protect redox-sensitive proteins and modulate GSH biosynthesis during salicylic acid (SA)-induced oxidative stress in Arabidopsis thaliana. SA enhanced GSH oxidation and protein glutathionylation both in vitro and in vivo. To visualize glutathionylated proteins under native conditions, we developed CamLog, a transgene-free click-activated metabolic labeling method. CamLog revealed that SA induces glutathionylated protein condensates that strongly overlap with SG markers but not with processing bodies. These condensates share more than 77% of their components with SGs, with translation-related proteins particularly enriched, and pharmacological analyses confirmed their identity as oxidative-stress-induced SGs. Glutathionylation of the SG marker RBP47B regulated its mobility and SA responsiveness, and global inhibition of glutathionylation abolished SG formation. Conversely, SGs sequestered glutathionylated proteins, including translation machinery, forming a reductive niche that prevents oxidation-induced proteolysis. SGs also sequestered GSH1, the rate-limiting enzyme for GSH biosynthesis, suggesting a mechanism to fine-tune GSH metabolism rather than fully counteract oxidation. Together, our findings uncover an organelle-level role of SGs in shaping cytosolic redox heterogeneity and highlight a spatial antioxidative strategy relevant to oxidation-vulnerable systems.
    Keywords:  oxidative stress; protein S-glutathionylation; proteostasis; redox heterogeneity; salicylic acid; stress granules
    DOI:  https://doi.org/10.1016/j.molp.2025.12.018
  33. bioRxiv. 2025 Dec 08. pii: 2025.12.07.692794. [Epub ahead of print]
      The endoplasmic reticulum (ER) membrane protein Rft1 is a scramblase for the anionic glycolipid Man5GlcNAc2-PP-dolichol (M5-DLO), a key intermediate in the pathway of protein N -glycosylation. As a member of the multidrug/oligosaccharidyl-lipid/polysaccharide (MOP) transporter superfamily Rft1 resembles the bacterial MurJ Lipid II flippase which has an analogous substrate, but the mechanism by which it translocates M5-DLO is not known. Here we used AlphaFold3 and Chai1 to develop conformational models of yeast Rft1-M5-DLO complexes. The models suggest an alternating access transport mechanism with inward-open, occluded, and outward-facing states. A central cavity accommodates the hydrophilic M5-DLO head group, while the dolichol lipid tail is guided through a portal formed between transmembrane helices 1 and 8 to a hydrophobic groove outside the cavity. Comparative mutational analysis of cavity residues revealed a significant mechanistic divergence from MurJ which strictly relies on a conserved charged triad to anchor Lipid II via its pyrophosphate neck. Whereas mutations to these residues in MurJ result in total loss of function, Rft1 showed high tolerance to most charge inversion mutations in the central cavity when tested for function using a yeast reporter strain. This tolerance implies greater flexibility in Rft1's electrostatic requirements for engaging M5-DLO. Our findings suggest that Rft1 functions as a specialized, alternating access transporter for M5-DLO and identify key molecular determinants that are essential for function. The alternating access mechanism distinguishes Rft1 from all currently known scramblases which make use of a hydrophilic transmembrane groove to provide a pathway for lipid transit.
    Importance: Cell surface and secreted proteins are decorated with sugar chains. These chains are first assembled on a lipid carrier. Initial stages of assembly occur on the cytoplasmic side of a subcellular structure called the endoplasmic reticulum (ER). To complete assembly, the partially assembled lipid-linked sugar chain must be flipped across the ER. The Rft1 protein facilitates lipid flipping but how it does this is not known. Here we use computational methods and cell-based assays to address this question.
    DOI:  https://doi.org/10.64898/2025.12.07.692794
  34. Nat Commun. 2025 Dec 27.
      Proteolysis targeting chimeric small molecules (PROTACs) offer a strategy for degrading disease-associated proteins or controlling engineered protein tags fused to therapeutic proteins, like chimeric antigen receptors (CARs). New approaches are needed that allow spatiotemporal control of PROTAC activity, restricting degrader activity to targeted cells. Photopharmacology offers a solution by enabling light-mediated spatial control of drug action. Here, we synthesize photocaged and photoswitchable PROTAC molecules and test their regulation of proteins tagged with E. coli dihydrofolate reductase (eDHFR) in tumor and CAR-T cells. Several of the molecules are derived from triazole-linked trimethoprim-PROTACs (TMP-TACtz), that degrade eDHFR fused proteins at picomolar concentrations, show degradation in cells with low cereblon E3 ligase levels, and have little off-target effects. The photocleavable compound, TMP-TAC-PC yields the best light-mediated regulation of CAR T cell cytotoxicity and cytokine secretion. This work introduces photocontrolled, tag-directed degraders for controlling protein expression in tumor cells and CAR T cells.
    DOI:  https://doi.org/10.1038/s41467-025-67527-5
  35. J Proteome Res. 2025 Dec 22.
      The import of most mitochondrial proteins requires that their precursor proteins be bound by the peripheral receptor proteins TOM20, TOM22, and TOM70. Budding yeast TOM20 and TOM70 have been extensively studied regarding their interaction partners and recognized substrates; however, little data is available for metazoan cells. Using APEX2-based proximity labeling, we created association profiles for human TOMM20 and TOMM70 in HeLa cells. We focused particularly on their interactions with RNA-binding proteins (RBPs) because there is evidence of RNA association with the mitochondrial outer membrane (MOM) and of local translation at the mitochondrial surface, however, these processes are poorly understood. Our results demonstrate that several RBPs and translation factors preferentially associate with TOMM20 rather than TOMM70. These include SYNJ2BP, a previously identified membrane-bound RBP that binds and protects mRNA encoding mitochondrial proteins. Inhibiting translation with puromycin increased the association of these RBPs with TOMM20 compared to TOMM70. This suggests that TOMM20, but not TOMM70, may play a role in maintaining cellular homeostasis during translation stress by retaining protective RBPs and translation-related proteins at the MOM.
    Keywords:  APEX2; SYNJ2BP; TOMM20; TOMM70; mitochondrial import; proteomics; proximity labeling
    DOI:  https://doi.org/10.1021/acs.jproteome.5c00905
  36. bioRxiv. 2025 Dec 20. pii: 2025.12.18.695253. [Epub ahead of print]
      Protein glycosylation and phosphorylation constitute two pervasive regulatory layers in mammalian cells, yet the effects that protein glycosylation play in phosphorylation signaling remain poorly understood. Here we show that controlled perturbation of N-linked glycan biosynthesis through glycoengineering fundamentally rewires phosphorylation signaling networks in human cells. Using comprehensive proteomics approaches, we simultaneously profiled the global proteome, glycoproteome, and phosphoproteome in engineered HEK293 cells designed to eliminate core fucosylation while enhancing sialylation and reducing GlcNAc branching complexity. Glycoengineering emerged as the dominant source of molecular variation across all datasets, with over 9,800 intact glycopeptides identified of which 3,400 are significantly altered, establishing a remodeled baseline cellular state. Upon serum stimulation, engineered cells not only exhibited markedly decreased phosphorylation responses compared to wild-type cells, but comprehensively re-wired to prefer signaling away from canonical EGFR/mTOR growth pathways. These findings establish a systematic framework for targeting glycosylation-phosphorylation regulation and nominate glycan-dependent signaling nodes as potential therapeutic vulnerabilities in glycosylation-remodeled disease states.
    DOI:  https://doi.org/10.64898/2025.12.18.695253
  37. bioRxiv. 2025 Dec 21. pii: 2025.12.19.695442. [Epub ahead of print]
      Dysregulation of the E3 ubiquitin ligase TRIM37 is associated with tumor formation and Mulibrey nanism, a recessive developmental syndrome. TRIM37 regulates steady-state levels of centrosome proteins and limits their ectopic assembly, but how it recognizes and ubiquitinates its substrates is poorly understood. We found that TRIM37 directly ubiquitinates the centrosome-forming protein Cep192 at 7 lysines clustered near its C-terminus. TRIM37 binds Cep192 at a C-terminal intrinsically disordered region followed by an ASH domain (IDR+ASH8). Mutation of the 7 lysines or the IDR+ASH8 domain increased Cep192 levels and stability in cells, indicating loss of TRIM37-based regulation. Fusing IDR+ASH8 to an unrelated protein (GFP-EB1) was sufficient to enable its degradation via TRIM37. Biochemical assays revealed that IDR+ASH8 is primarily monomeric and binds TRIM37 via two separate coiled-coil motifs with mid-nanomolar affinity. We propose that the IDR+ASH8 motif is a bipartite degron for TRIM37, enabling it to target centrosome proteins and adjust their levels.
    DOI:  https://doi.org/10.64898/2025.12.19.695442
  38. Brief Bioinform. 2025 Nov 01. pii: bbaf685. [Epub ahead of print]26(6):
      Proteins primarily perform their functions through interactions with other proteins, making the accurate prediction of protein-protein interactions (PPIs) a fundamental problem. Experimental methods for determining PPIs are often slow and expensive, which has driven significant efforts to improve the performance of computational methods in this field. While many methods have been designed, recent thorough investigations proved that the existing methods learn exclusively from sequence similarities and node degrees. When such data leakage is avoided, performances were shown to become random. We introduce C3PI, a novel sequence-based deep learning framework designed for predicting PPIs. C3PI uses as input ProtT5 protein embeddings into a complex architecture that includes two novel components, a puzzler and an entangler, which significantly enhance the model's performance. Through extensive comparisons with state-of-the-art methods across many datasets, C3PI consistently outperforms competing approaches, especially in key metrics such as AUPRC and AUROC. Most importantly, C3PI is the first PPI prediction method to achieve a significant improvement over random on the leakage-free gold standard dataset. C3PI is available as a web server at c3pi.csd.uwo.ca and source code from github.com/lucian-ilie/C3PI.
    Keywords:  ProtT5; machine learning; protein embedding; protein interaction
    DOI:  https://doi.org/10.1093/bib/bbaf685
  39. J Biol Chem. 2025 Dec 24. pii: S0021-9258(25)02956-4. [Epub ahead of print] 111104
      Uncontrolled centriole duplication leads to centrosome amplification and chromosomal instability, but its underlying mechanism is poorly understood. A new centriole is duplicated from a cartwheel-like structure assembled by Plk4-phosphorylated SCL/TAL1-interrupting locus (STIL) and its associated SAS6. Here, we show that depletion of SCF E3 ubiquitin ligase, FBXW7 induces pre-matured duplication of centrioles via excessive stabilization of STIL-SAS6 axis. FBXW7 mediates degradation of STIL-SAS6 axis and Plk4 kinase activity is required for this degradation. Interestingly, phosphorylation of key Plk4-targeting sites in STIL that drives new centriole assembly by facilitating STIL-SAS6 interaction, also stabilizes FBXW7 binding to STIL and promotes degradation of the STIL-SAS6 complex, thus revealing an opposing molecular mechanism to inhibit centriole over-duplication. Genomic analyses of cancer cell line data reveal a negative correlation between FBXW7 expression and aneuploidy, as well as a positive correlation between FBXW7 and STIL expression at the mRNA level. Our results thus contribute to improved understanding of the molecular basis of centrosome amplification and aneuploidy.
    Keywords:  Aneuploidy; Centriole; Centrosome; FBXW7; SAS6; STIL
    DOI:  https://doi.org/10.1016/j.jbc.2025.111104
  40. Proc Natl Acad Sci U S A. 2025 Dec 30. 122(52): e2511925122
      Understanding protein structure and dynamics is crucial for basic biology and drug design. Conventional methods often provide static conformations that inadequately capture protein flexibility. We present PackDock, a framework that integrates deep learning and physics-based modeling to represent protein-ligand interactions. PackDock's core, PackPocket, uses diffusion models to sample diverse binding pocket conformations and predict ligand-induced changes. We validate PackDock through side-chain packing, redocking, and cross-docking experiments, demonstrating its ability to address protein flexibility challenges. In a real-world application, PackDock identified nanomolar affinity compounds with unreported scaffolds for the protein of interest. Additionally, it revealed key amino acid conformational changes, offering insights into protein-ligand interactions. By accurately predicting complex conformations in various scenarios, PackDock enhances our understanding of protein dynamics and provides perspectives for both basic biological research and drug discovery efforts.
    Keywords:  machine learning; molecular docking; protein structure prediction
    DOI:  https://doi.org/10.1073/pnas.2511925122
  41. FEBS J. 2025 Dec 26.
      CML is primarily driven by the oncogenic BCR-ABL fusion kinase; however, tyrosine kinase inhibitor (TKI) resistance remains a significant clinical challenge. A study by Zang et al. identified USP8 as a critical mediator of this resistance. USP8, a deubiquitinase, stabilizes the stress-response regulator EIF2S1 (eIF2α) by removing K48-linked ubiquitin chains. This stabilization sustains PERK-EIF2S1-mediated unfolded protein response (UPR) signaling. The UPR suppresses general protein translation while promoting the expression of adaptive stress-response genes, allowing CML cells to survive TKI-induced stress. Consequently, targeting the USP8-EIF2S1 axis is proposed as a key therapeutic strategy to overcome resistance and enhance patient outcomes.
    Keywords:  TKI resistance drugs; UPR signaling; chronic myeloid leukemia; deubiquitinating enzyme; structure‐based drug design; ubiquitin‐specific protease family
    DOI:  https://doi.org/10.1111/febs.70372
  42. Cell Rep. 2025 Dec 19. pii: S2211-1247(25)01513-X. [Epub ahead of print]45(1): 116741
      Determining if repeat-associated non-AUG (RAN) proteins contribute to the CAG-polyglutamine (polyGln)-encoding spinocerebellar ataxias (CAG-SCAs) is critical for understanding disease mechanisms and for therapy development. Immunohistochemistry shows that sense polyserine (polySer) (AGC frame) and antisense polyleucine (polyLeu) (CUG frame) RAN protein aggregates accumulate throughout the cerebellum and pons, in SCA1, SCA2, SCA3, SCA6, and SCA7 autopsy brains, and in damaged neurons. Cerebellar white matter regions, with prominent polySer and polyLeu but minimal polyGln, show neuroinflammation and demyelination. In SCA3 mice, RAN protein aggregates increase with age. SCA1 Pcp2-ATXN1[82Q] (Pcp2-82Q) mice designed to express ataxin-1 (ATXN1)-polyGln in Purkinje cells show sense and antisense RAN protein aggregates throughout the cerebellum. Disrupting the ATXN182Q:capicua binding, which improves behavior and neuropathology, also reduces RAN protein aggregates. In neural cells, toxic polySer and polyLeu proteins impair autophagy, and reducing RAN protein levels with metformin reduces cytotoxicity. These data identify sense and antisense RAN proteins as a common molecular mechanism shared by the CAG-SCAs.
    Keywords:  CP: molecular biology; CP: neuroscience; RAN translation; SCA; metformin; neurodegeneration; polyGln; polyLeu; polyQ; polySer; repeat-associated non-AUG translation; sense and antisense RAN proteins; spinocerebellar ataxia; white matter loss
    DOI:  https://doi.org/10.1016/j.celrep.2025.116741
  43. Angew Chem Int Ed Engl. 2025 Dec 21. e16809
      The precise regulation of gene expression is fundamental to cellular homeostasis and diversity. Dysregulation of splicing has been implicated in a range of diseases, including cancer and neurodegeneration. Ubiquitin-specific protease 39 (USP39), an essential spliceosome component lacking enzymatic activity, has remained an elusive target for pharmacological intervention. Here, we report the discovery of small-molecule ligands that selectively engage with USP39 through a thiazole scaffold, primarily interacting with its zinc finger domain. Guided by AlphaFold-based structure-activity relationship studies, we designed and optimized proteolysis-targeting chimeras (PROTACs), culminating in the development of USP39_PROTAC_V1, which harnesses the von Hippel-Lindau (VHL) E3 ubiquitin ligase for targeted degradation. Biophysical and biochemical assays demonstrated potent ternary complex formation and nanomolar-range binding affinities. In cellular models, USP39_PROTACs achieved efficient degradation of USP39 at concentrations as low as 1 nM, with minimal off-target effects as confirmed by proteome-wide profiling. Mechanistic studies revealed that degradation was dependent on VHL recruitment and was abrogated by proteasome or neddylation inhibition. Notably, USP39 depletion recapitulated 5'-splice-site-specific splicing patterns previously described, thereby validating both the mechanism of action and the therapeutic relevance of this approach-particularly for modulating splicing-associated disease pathways such as cancer and retinitis pigmentosa.
    Keywords:  Biological chemistry and chemical biology; Drug discovery; PROTAC; Spliceosome; USP39
    DOI:  https://doi.org/10.1002/anie.202516809
  44. bioRxiv. 2025 Dec 18. pii: 2025.12.18.695067. [Epub ahead of print]
      Lysine 27 (K27)-linked polyubiquitination plays critical yet incompletely defined roles in proteostasis, innate immunity, and disease progression; however, investigations into this process have long been hindered by its extremely low abundance and the lack of conformation-specific enrichment tools. Herein, we describe the development of a long-sought conformation-specific antibody, K27-IgG, which can selectively recognize-among all ubiquitin chain types-the unique architecture of K27-linked polyubiquitin (K27-polyUb) characterized by a distinct buried K27-isopeptide bond, with high affinity (KD = 4.66 nM). This antibody was derived from synthetic antibodies initially generated via phage display, using chemically synthesized K27-linked diubiquitin (K27-diUb) as the antigen. High-resolution co-crystal structures uncovered the unique K27-diUb interface targeted by these sAbs. Subsequent reformatting of these sAbs into a full-length human immunoglobulin G (IgG) scaffold yielded K27-IgG, notably exhibiting markedly enhanced affinity without compromising selectivity. Using K27-IgG as a tool, we achieved sensitive detection and immunoprecipitation (IP) of endogenous K27-polyUb in cells, and delineated the intracellular interaction landscape of K27-polyUb through complementary proteomic approaches. Two key findings emerged: 1) The molecular chaperone DNAJB1 is a specific reader of K27-linked ubiquitin chains (but not other linkages) and that K27-polyUb chains themselves exhibit chaperone-like activity, suggesting a novel mechanism by which K27-polyUb regulates chaperone-mediated proteostasis; 2) The E2 enzyme UBE2Q1 assembles K27-diUb, identifying it as a potential writer for this ubiquitin chain topology. Collectively, this study establishes K27-IgG as a robust tool for deciphering the K27-linked ubiquitin code, thereby opening new avenues for investigating the biological functions of K27-linked polyubiquitination.
    Highlights: First K27-linkage conformation-specific antibody with nanomolar affinity overcomes a major barrier in the field.K27-IgG unlocks functional mapping of the K27 ubiquitin landscape under proteotoxic stress.Molecular chaperone DNAJB1 is a selective "reader" of K27-linked ubiquitin chains.K27 chains possess intrinsic chaperone activity, enabling protein refolding and suppressing aggregation.E2 enzyme UBE2Q1 is a "writer" that directly assembles K27-linked ubiquitin chains.
    Graphical abstract:
    DOI:  https://doi.org/10.64898/2025.12.18.695067
  45. bioRxiv. 2025 Dec 12. pii: 2025.12.09.692983. [Epub ahead of print]
      Stereoselective recognition is a powerful means to differentiate selective versus non-specific activity of small molecules in complex biological systems. Here, we disclose stereochemically defined, sulfonyl-triazole inhibitors of the lipid enzyme diacylglycerol kinase-alpha (DGKα), a key metabolic checkpoint for T cell effector function. Acute treatment with the covalent DGKα inhibitor AHL-7160 recruited endogenous DGKα to the plasma membrane in a stereoselective and isozyme-specific manner. The membrane translocation activity of AHL-7160 correlated with blockade of cellular phosphatidic acid production and potentiation of primary T cell-mediated killing of a glioblastoma cell line. Quantitative chemoproteomics revealed Y669 and K411 as sites of AHL-7160 modification on endogenous DGKα in cells. Extended treatments resulted in proteasome-dependent and proteome-wide selective degradation of DGKα in T cells. Collectively, these findings establish covalent DGKα ligands as potent molecular glues with translational potential in immunotherapy.
    DOI:  https://doi.org/10.64898/2025.12.09.692983
  46. Nat Commun. 2025 Dec 20.
      Predicting changes in protein thermostability caused by amino acid substitutions is essential for understanding human diseases and engineering proteins for practical applications. While recent protein generative models demonstrate impressive zero-shot performance in predicting various protein properties without task-specific training, their strong unsupervised prediction ability remains underexploited to improve protein stability prediction. We present SPURS, a deep learning framework that rewires and integrates two complementary protein generative models-a protein language model and an inverse folding model-and reprograms this unified framework for stability prediction through supervised fine-tuning on mega-scale thermostability data. SPURS delivers accurate, efficient, and scalable stability predictions and generalizes to unseen proteins and mutations. Beyond stability prediction, SPURS enables broad applications in protein informatics, including zero-shot identification of functional residues, improved low-N protein fitness prediction, and systematic dissection of stability-pathogenicity for human diseases. Together, these capabilities establish SPURS as a versatile tool for advancing protein stability prediction and protein engineering at scale.
    DOI:  https://doi.org/10.1038/s41467-025-67609-4
  47. Sci Rep. 2025 Dec 21.
      Triple-negative breast cancer (TNBC) is characterized by the absence of estrogen receptor (ER), progesterone receptor (PR), and HER-2 amplification, rendering it unresponsive to endocrine and HER2-targeted therapies. GRP78 (78 kDa glucose-regulated protein), a key endoplasmic reticulum (ER)-resident chaperone involved in protein folding and stress response, has been observed atypically localized on the cell surface of various cancer and stressed cell types, where it engages in non-canonical signaling and cellular functions. Cell surface GRP78 (csGRP78) is preferentially expressed in malignant cells relative to normal tissue, making it an attractive therapeutic target. In this study, we report that over 70% of MDA-MB-231 TNBC cells express csGRP78. Interestingly, MDA-MB-231 cells predominantly exhibit a distinct unipolar morphology, with csGRP78 prominently co-localizing with the variant isoform of CD44 (CD44v, containing variable exon 3) at the anterior region of the cell. Co-localizations of csGRP78 and CD44v were also observed in MDA-MB-231 tumor xenografts, supporting its relevance in vivo. Importantly, targeting csGRP78 with the monoclonal antibody 76-E6 downregulated CD44v expression, inhibited Src kinase signaling, disrupted cell morphology, and suppressed cell motility. We further mapped the epitope of GRP78 targeted by 76-E6. Together, our findings identify csGRP78 as a functional regulator of cell morphology and migration at least in part via a csGRP78-CD44v axis and underscore its potential as a therapeutic target in TNBC.
    Keywords:  CD44; GRP78; MDA-MB-231; Src; TNBC; Triple-negative breast cancer
    DOI:  https://doi.org/10.1038/s41598-025-33441-5
  48. Cell Rep. 2025 Dec 20. pii: S2211-1247(25)01511-6. [Epub ahead of print]45(1): 116739
      Recent studies have uncovered a number of functional RNA structures in RNA viruses, yet their regulatory roles remain poorly understood. Here, using an unbiased proteomic approach alongside targeted biochemical assays, we investigate a previously uncharacterized functional pseudoknot (pk1) within the hepatitis C virus coding region and show that it stably interacts with host ribosomes, inhibiting translation and potentially acting as a regulator between viral translation and RNA genome replication. Comparative structural analysis identifies pk1-like elements in related RNA viruses, suggesting a conserved regulatory mechanism. This study expands the known functions of pseudoknots in viral coding regions beyond frameshifting, highlights the critical role of RNA structure-mediated regulation within viral open reading frames, and provides insight into the design of viral therapeutics and vaccines.
    Keywords:  CP: microbiology; CP: molecular biology; RNA structure; RNA virus; RNA-binding proteins; anti-sense RNA; genetic code; locked nucleic acids; riboregulation; translation inhibition; translation initiation
    DOI:  https://doi.org/10.1016/j.celrep.2025.116739
  49. Protein Sci. 2026 Jan;35(1): e70376
      Proteins perform diverse functions critical to cellular processes. Transitions between functional states are often regulated by post-translational modifications (PTMs) such as phosphorylation, which dynamically influence protein structure, function, folding, and interactions. Dysregulation of PTMs can therefore contribute to diseases such as cancer and Alzheimer's. However, the structure-function relationship between proteins and their modifications remains poorly understood due to a lack of experimental structural data, the inherent diversity of PTMs, and the dynamic nature of proteins. Recent advances in deep learning, particularly AlphaFold, have transformed protein structure prediction with near-experimental accuracy. However, it remains unclear whether these models can effectively capture PTM-driven conformational changes, such as those induced by phosphorylation. Here, we systematically evaluated AlphaFold models (AF2, AF3-non phospho, and AF3-phospho) to assess their ability to predict phosphorylation-induced structural diversity. By analyzing experimentally derived conformational ensembles, we found that all models predominantly aligned with dominant structural states, often failing to capture phosphorylation-specific conformations. Despite its phosphorylation-aware design, AF3-phospho predictions provided only modest improvement over AF2 and AF3-non phospho predictions. Our findings highlight key challenges in modeling PTM-driven structural landscapes and underscore the need for more adaptable structure prediction frameworks capable of capturing modification-induced conformational variability.
    Keywords:  AlphaFold3; conformational diversity; phosphorylation; post‐translational modifications (PTMs); protein structures
    DOI:  https://doi.org/10.1002/pro.70376
  50. Signal Transduct Target Ther. 2025 Dec 25. 10(1): 419
      High-grade serous ovarian cancer (HGSC) accounts for more than 70% of ovarian cancer-related deaths, yet therapeutic progress remains stagnant. Among the four molecular subtypes reported for HGSC, the C5 subtype is distinguished by high proliferation and immune evasion with an unfavorable MHC-I/PD-L1 ratio. However, the molecular drivers of this immune desert state remain largely undefined. Here, we identify RNA-binding proteins (RBPs) as key regulators of immune evasion in C5-HGSC through integrated single-cell and bulk RNA sequencing. We perform a targeted loss-of-function screen in C5-like cell models and find IGF2BP1 as a central mediator of immune evasion in vitro and in vivo. Mechanistically, IGF2BP1 abrogates interferon-gamma signaling by accelerating IRF1 protein degradation, thereby suppressing MHC-I presentation. We also discover that IGF2BP1 decouples PD-L1 expression from IRF1-dependent transcription and reshapes the immune receptor landscape to limit immune cell infiltration and T cell activation. Therapeutically, the small-molecule BTYNB effectively inhibits IGF2BP1 and synergizes with PD-1 blockade to overcome immune evasion in vivo. Multi-spectral imaging confirms these findings in human HGSC tissues and highlights the role of oncofetal RBPs as molecular drivers of the C5-HGSC subtype. This subtype-wide survey uncovers a previously unrecognized RBP-interferon regulatory axis and establishes RBP inhibition as a therapeutic strategy to enhance immune checkpoint therapy in immunologically cold ovarian tumors.
    DOI:  https://doi.org/10.1038/s41392-025-02515-1
  51. Theranostics. 2026 ;16(5): 2388-2404
      Background: Huntington's disease (HD) is a devastating neurodegenerative disorder caused by CAG repeat expansion in the HTT gene, resulting in a polyglutamine-expanded huntingtin (HTT) protein that forms toxic aggregates. Although heat-shock proteins are known to facilitate the refolding or clearance of misfolded proteins, their precise role in modulating protein aggregation in HD remains unclear. Here, we explore the function of caseinolytic peptidase B (ClpB), a mitochondrial AAA+ ATPase and heat-shock protein, in maintaining proteostasis and synaptic integrity in HD. Methods: We examined how CLPB loss or overexpression in human embryonic kidney 293T (HEK293T) cells impacted the aggregation of wild-type HTT (HTT-Q23) and mutant HTT (HTT-Q79). In parallel, AAV-mediated ClpB knockdown or overexpression was applied to the striatum of HD model mice. and HTT aggregation and inhibitory synaptic alterations were assessed. Aggregate burden was quantified via immunostaining, and inhibitory synapse density was evaluated using VGAT immunohistochemistry and electrophysiological recordings. Results: In HEK293T cells, CLPB knockout led to abnormal aggregation of HTT-Q23 while CLPB overexpression reduced the size of HTT-Q79 aggregates. In the mouse striatum, ClpB knockdown increased HTT-Q23 aggregate numbers and altered HTT-Q79 aggregation morphology, whereas CLPB overexpression restored the density and size of VGAT-positive inhibitory synapses and improved inhibitory synaptic transmission in HD model mice. These effects of CLPB overexpression were associated with a reduced mitochondrial aggregation burden, suggesting that ClpB contributes to mitochondrial protein quality control. Conclusions: These results demonstrate that ClpB regulates both physiological and pathological HTT aggregation and contributes to maintaining inhibitory synaptic integrity. By modulating mitochondrial proteostasis, ClpB acts as a protective factor in HD pathology, highlighting its potential as a therapeutic target for neurodegenerative disorders characterized by protein misfolding.
    Keywords:  ClpB; Huntington's disease; disaggregase; inhibitory synapse; mitochondria; striatum
    DOI:  https://doi.org/10.7150/thno.122651
  52. PLoS Biol. 2024 Apr;22(4): e3002603
      The 3' end of the 18S ribosomal RNA is formed by the endoribonuclease Nob1. How cells ensure the accuracy of the 3' end has remained a mystery. A new study in PLOS Biology revealed that there are multiple checkpoints to ensure that only ribosomes containing the correct 3' end participate in translation.
    DOI:  https://doi.org/10.1371/journal.pbio.3002603
  53. bioRxiv. 2025 Dec 18. pii: 2025.09.18.676787. [Epub ahead of print]
      The rapid expansion of protein sequence databases has far outpaced experimental structure determination, leaving many unannotated sequences, particularly the more remote homologs with low sequence identity. Because protein folds are more conserved and functionally informative than sequences alone, structural information offers a powerful lens for analysis. Here, we introduce a generative, structure-aware framework that integrates geometric encoding and coevolutionary constraints to map, cluster, and design protein sequences. Our approach employs the 3D interaction (3Di) alphabet to convert local residue geometries into compact, 20-state discrete representations. Using ProstT5, we enable bidirectional translation between amino acid sequences and 3Di representations, facilitating sensitive homology detection and structure-guided sequence generation. We then augment the latent generative landscape methodology by combining 3Di-based alignments with direct coupling analysis (DCA) and variational autoencoders (VAE), imbuing tasks such as clustering, annotation, and design with structural information. This integrative framework enhances the detection of coevolutionary signals and enables rational sampling of structural variants, even without functional labels. We demonstrate the utility of our method across diverse protein families, including globins, kinases, and malate dehydrogenases, achieving improved contact prediction, homology inference, and sequence generation. Together, our approach offers a quantitative, generative view of protein structure space, advancing protein evolution and design studies.
    Significance Statement: Protein sequence databases are growing far faster than our ability to experimentally determine structures, leaving much of protein space poorly annotated, especially for distant homologs. Because protein structure is more conserved and informative than sequence alone, new approaches are needed to exploit structural signals at scale. We present a generative framework that integrates compact structural representations with evolutionary constraints to map, cluster, and design protein sequences. By combining geometric encoding with coevolutionary modeling, our approach enables sensitive homology detection, improved inference of structural contacts, and rational exploration of sequence space without requiring functional labels. This work provides a quantitative bridge between protein sequence and structure, advancing our ability to interpret protein evolution and guide protein design.
    DOI:  https://doi.org/10.1101/2025.09.18.676787
  54. Nat Commun. 2025 Dec 24.
      Generative protein modeling provides advanced tools for designing diverse protein sequences and structures. However, accurately modeling the conformational landscape and designing sequences remain critical challenges: ensuring that the designed sequence reliably folds into the target structure as its most stable conformation, and optimizing the sequence for a given suboptimal fixed input structure. In this study, we present a systematic analysis of jointly optimizing sequence-to-structure and structure-to-sequence mappings. This approach enables us to find optimal solutions for modeling the conformational landscape. We validate our approach with large-scale protein stability measurements, demonstrating that joint optimization is superior for designing stable proteins using a joint model (TrRosetta and TrMRF) and for achieving high accuracy in stability prediction when jointly modeling (half-masked ESMFold pLDDT + ESM2 Pseudo-likelihood). We further investigate features of sequences generated from the joint model and find that they exhibit higher frequencies of hydrophilic interactions, which may help maintain both secondary structure registry and pairing-features not captured by structure-to-sequence modeling alone.
    DOI:  https://doi.org/10.1038/s41467-025-66526-w
  55. J Biol Chem. 2025 Dec 22. pii: S0021-9258(25)02945-X. [Epub ahead of print] 111093
      Dendritic cells (DCs) reside in tissues and are activated by danger or pathogen-associated signals, leading to expression of costimulatory markers and cytokines, downregulation of β2-integrin-mediated adhesion, migration to lymph nodes, and activation of T cells. Conversely, β2-integrins are known to restrict DC activation and migration, but signaling pathways involved in this process remain poorly understood. Here, we show that β2-integrins regulate podosome formation as well as nuclear morphology of bone marrow (BM) derived DCs on both stiff and soft surfaces. Analysis of published gene expression data revealed that loss of beta2-integrin adhesion and nuclear deformation both upregulate DC activation markers and cytokines, including Ccr7, Cd86 and Il12b. Growth arrest and DNA damage-inducible protein (GADD34; Ppp1r15) of the unfolded protein responses was also upregulated in both datasets. Utilizing a cPLA2 inhibitor, we show that the nuclear shape sensor cytosolic phospholipase A2 (cPLA2) controls IL-12 production and CD86 expression of β2-integrin adhesion deficient BMDCs. We further show that the GADD34 pathway is activated in adhesion-deficient BMDCs, as EIF2α phosphorylation is reduced in these cells, whilst cPLA2-inhibition rescues EIF2α phosphorylation. Furthermore, GADD34 inhibition led to decreased IL-12 production and CD86 expression in β2-integrin deficient BMDCs, whilst inhibition of PERK (the canonical EIF2α kinase) had no effect. Together, our results show that loss of β2-integrin adhesion leads to nuclear deformation and activation of a cPLA2/GADD34 pathway in BMDCs, which at least partly controls their activated phenotype. Our results therefore reveal how cell adhesion and nuclear deformation are connected to mechanically control dendritic cell activation.
    Keywords:  cell signaling; dendritic cell; endoplasmic reticulum stress; integrin; nuclear structure
    DOI:  https://doi.org/10.1016/j.jbc.2025.111093
  56. Environ Pollut. 2025 Dec 19. pii: S0269-7491(25)01936-0. [Epub ahead of print] 127562
      Trifloxystrobin (TFX), a potent inhibitor of complex III in the mitochondrial respiratory chain, is a widely used strobilurin fungicide whose neurotoxic mechanisms remain poorly defined. This study investigated the molecular pathways underlying TFX-induced toxicity in human SH-SY5Y neuronal-like neuroblastoma cells, with particular emphasis on oxidative stress, mitochondrial dysfunction, and activation of the Integrated Stress Response (ISR). TFX exposure (24 h) exhibited an IC50 of approximately 100 μM, induced G0/G1 cell cycle arrest, and triggered mitochondria-mediated apoptosis, as evidenced by loss of mitochondrial membrane potential (ΔΨm), Bax activation, cytochrome c release, DNA fragmentation, phosphatidylserine exposure, and caspase-3 activation. These effects were accompanied by increased mitochondrial superoxide levels and decreased ATP production, indicating profound mitochondrial impairment. Pretreatment with N-acetylcysteine (NAC) markedly restored cell viability, reduced ROS accumulation, prevented ΔΨm dissipation, and diminished apoptotic damage. Mechanistically, TFX activated the ISR through the OMA1-DELE1-HRI mitochondrial stress signaling axis, as confirmed by loss-of-function experiments targeting these proteins. Importantly, both NAC and the ISR inhibitor ISRIB (Integrated Stress Response InhiBitor) significantly attenuated ISR activation and the resulting apoptosis, demonstrating that oxidative stress serves as an upstream trigger for ISR engagement and cell death. Collectively, these findings reveal that TFX induces oxidative stress-dependent activation of the OMA1-DELE1-HRI ISR pathway, linking mitochondrial dysfunction to apoptosis in human neuroblastoma cells. To our knowledge, this is the first report identifying ISR activation as a mechanistic component of strobilurin fungicide-induced neurotoxicity.
    Keywords:  ISR; SH-SY5Y cells; Trifloxystrobin; apoptosis; oxidative stress
    DOI:  https://doi.org/10.1016/j.envpol.2025.127562
  57. Nat Commun. 2025 Dec 21.
      Efficient fatty acid (FA) re-esterification is essential for lipid homeostasis in adipocytes, yet the mechanisms coordinating Coenzyme A (CoA) availability at the endoplasmic reticulum (ER)-a major site of lipid synthesis-remain unclear. Here, we identify TMEM120A as an ER-resident CoA-binding protein that regulates intracellular FA metabolism. TMEM120A interacts with the ER-localized acyl-CoA synthetase ACSL1 and ACSL3 to promote long-chain acyl-CoA synthesis and channeling into the ER, thereby facilitating FA re-esterification and lipid cycling during lipolysis. By relieving acyl-CoA-mediated feedback inhibition of lipolysis, TMEM120A enhances lipid turnover while protecting against ER stress and lipotoxicity. Adipocyte-specific deletion of Tmem120a in mice impairs lipolysis-induced energy expenditure and exacerbates inflammation and metabolic dysfunction under high-fat diet conditions. These findings establish TMEM120A as a critical regulator of ER CoA handling and lipid flux, revealing a previously unrecognized mechanism that links intracellular CoA dynamics to systemic energy balance and metabolic health.
    DOI:  https://doi.org/10.1038/s41467-025-67870-7
  58. Neurotox Res. 2025 Dec 27. 44(1): 2
      The objective of the present review is to discuss the involvement of altered mitochondrial quality control in Mn-induced neurotoxicity. Existing data demonstrate that mitochondrial autophagy (mitophagy) and brain mitochondrial unfolded protein response (mtUPR) are activated in response to Mn exposure to counteract the Mn-induced mitochondrial dysfunction. Both mitophagy and mtUPR have significant overlap and mechanistic intersections with the integrated stress response (ISR). Increased Mn exposures impair mitochondrial dynamics, further aggravating Mn-induced mitochondrial dysfunction. Specifically, Mn suppresses PTEN-induced kinase 1 (PINK1)-Parkin-dependent mitophagy through a variety of mechanisms, including nitric oxide synthase 2 (NOS2)-dependent PINK1 S-nitrosylation, inhibition of transcription factor EB (TFEB) signaling, and mammalian target of rapamycin complex 1 (mTORC1) activation. In addition, Mn promotes mitochondrial fission by up-regulating dynamin-1-like protein (Drp1) expression and phosphorylation via the activation of c-Jun N-terminal kinase (JNK) and inhibition of sirtuin 1 (SIRT1)/peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) pathways. Concomitantly, Mn impairs mitochondrial fusion by inhibiting mitofusin (Mfn) 1/2 and dynamin-like 120 kDa protein (Opa1) expression, leading to a reduction in mitochondrial size and disruption of the mitochondrial network. High-dose Mn exposure results in inhibition of peroxisome proliferator-activated receptor gamma coactivator 1α (PGC-1α)/nuclear factor erythroid 2-related factor 2 (NRF2)-dependent mitochondrial biogenesis. The latter may be mediated by inhibition of SIRT1/SIRT3 activity, as well as modulation of PINK1/ zinc finger protein 746 (ZNF746)/PGC-1α axis. Alterations in the mitochondrial quality control system may contribute to Mn-induced neuronal damage and neuroinflammation, indicating that dysregulation of the brain mitochondrial dynamics is an important mechanism by which Mn induces its neurotoxicity.
    DOI:  https://doi.org/10.1007/s12640-025-00776-w
  59. bioRxiv. 2025 Dec 13. pii: 2025.12.10.693600. [Epub ahead of print]
      Landscape Expansion Microscopy (land-ExM) is a light microscopy technique that visualizes both lipid and protein ultrastructural context of cells. Achieving this level of detail requires both superresolution and a high signal-to-noise ratio. Although expansion microscopy (ExM) provides superresolution, obtaining high signal-to-noise images of both proteins and lipids remains challenging. Land-ExM overcomes this limitation by using self-retention trifunctional anchors to significantly enhance protein and lipid signals in expanded samples. This improvement enables the accurate visualization of diverse membrane organelles and phase separations, as well as the three-dimensional visualization of their contact sites. As a demonstration, we revealed triple-organellar contact sites among the stress granule, the nuclear tunnel, and the nucleolus. Overall, land-ExM offers a high-contrast superresolution platform that advances our understanding of how cells spatially coordinate interactions between membrane organelles and phase separations.
    eTOC Summary: Zhuang et al. introduce land-ExM, a super-resolution approach that simultaneously maps protein and lipid ultrastructure in cells with high contrast. This method visualizes 3D interactions between membrane-bound organelles and phase-separated condensates, uncovering organelle contact sites such as stress granules at nuclear tunnels adjacent to nucleoli.
    DOI:  https://doi.org/10.64898/2025.12.10.693600
  60. Nat Commun. 2025 Dec 21.
      Collective cell migration during embryonic development, wound healing, and cancer metastasis requires the emergence of leader cells at the migration front. Despite their physiological relevance, the full mechanisms underlying the emergence of leader cells remain elusive. Here we report that leader cells display a peripheral accumulation of lysosomes in diverse model systems for wound healing, including cultured epithelial monolayer, mouse embryonic skin, and Drosophila embryos. This accumulation involves cellular contractile forces driving lysosomal transport along microtubules towards the leading edge. Indeed, we control leader cell emergence by manipulating lysosomal movement on microtubules. We further find that peripheral lysosomes associate with Rac1 molecules at the leading periphery, regulating local Rac1-activity, triggering actin polymerization and promoting lamellipodium formation. Taken together, we demonstrate that beyond their catabolic role, lysosomes act as an intracellular platform that links mechanical and biochemical signals to control the emergence of leader cells.
    DOI:  https://doi.org/10.1038/s41467-025-67645-0
  61. Cell Death Dis. 2025 Dec 22. 16(1): 897
      Metastatic melanoma (MM) displays remarkable phenotypic plasticity, allowing tumor cells to transition reversibly between proliferative and mesenchymal (MES)-like states. This dynamic switching is strongly associated with therapeutic resistance and poor prognosis. Although transcriptional and epigenetic mechanisms driving these transitions have been extensively studied, the role of post-translational regulation, particularly the ubiquitin-proteasome system, remains poorly understood. Here, we identify the ubiquitin E3 ligase RNF 123 (KPC1) as a key post-translational suppressor of MES reprogramming in MM. Integrative analyses of bulk and single-cell transcriptomic datasets revealed that KPC1 expression is inversely correlated with the expression of core mesenchymal markers such as ZEB1, CDH2, and AXL, and positively associated with epithelial and melanocytic lineage genes, including CDH1 and MITF. Deconvolution of TCGA-SKCM RNA-seq data confirmed that this inverse correlation is specific to malignant melanoma cells and strongest in tumors enriched for mesenchymal gene signatures. Single-cell trajectory and enrichment analyses further demonstrated that decreasing KPC1 expression accompanies MES-like switch. Mechanistically, KPC1 binds and promotes the ubiquitination and proteasomal-mediated degradation of ZEB1, thereby suppressing cadherin switching and cell motility. Loss of KPC1 in melanoma cells prevented ZEB1 proteasomal-mediated degradation, increased expression of mesenchymal markers, and enhanced MM cells migration. Clinically, low KPC1 protein levels were associated with increased expression of ZEB1 and CDH2 and poorer overall survival. Furthermore, combined assessment of KPC1, ZEB1, and CDH2 expression improved patient stratification, suggesting the potential utility of multi-marker signatures for prognostic modeling. These findings establish KPC1 as a central post-translational regulator of melanoma cell state plasticity through targeted degradation of ZEB1. This study highlights a novel mechanism regulating MES-like transition and highlights KPC1 as a potential theragnostic target in MM.
    DOI:  https://doi.org/10.1038/s41419-025-08262-z
  62. Nat Commun. 2025 Dec 22.
      Lysosome-targeting chimeras specifically degrade membrane/extracellular proteins with the specificity and potency highly dependent on the expression pattern and kinetics of lysosome-targeting receptors. Here we describe an endosome-phagophore linking assembly (EPLA), which is constructed by attaching a specific ligand for a protein of interest (POI) to an endosome surface-anchored phagophore-targeting peptide (ES-PTP) that consists of a pH-low insertion peptide and a phagophore-binding element. EPLAs degrade proteins by binding with POIs, being internalized into endosomes, inserting endosome membrane, anchoring the phagophore-targeting moiety onto endosome surface, and then triggering the spatial proximity of endosomes to phagophores. We show the modularity and generality of the EPLA technology by degrading membrane-bound PD-L1, RAGE, GRP94, PSMA, ICAM-1, p32, VCAM-1 and LRP1, and extracellular amyloid-β and tau. Our results establish a technology platform for autophagic degradation of the membrane/extracellular POIs, which may have broad implications for biochemical research and clinical therapeutics.
    DOI:  https://doi.org/10.1038/s41467-025-67805-2
  63. Proc Natl Acad Sci U S A. 2025 Dec 30. 122(52): e2425774122
      We describe peptide mapping through Split Antibiotic Resistance Complementation (SpARC-map), a method to identify the probable interface between two interacting proteins. Our method is based on in vivo affinity selection inside a bacterial host and uses high-throughput DNA sequencing to infer probable protein-protein interaction (PPI) interfaces. SpARC-map uses only routine microbiology techniques, with no reliance on specialized instrumentation, dedicated reagents, or reconstituting protein complexes in vitro. SpARC-map can be tuned to detect PPIs over a broad range of affinities, multiplexed to probe multiple PPIs in parallel, and its nonspecific background can be precisely measured, enabling the sensitive detection of weak PPIs. Using SpARC-map, we recover known PPI interfaces in the p21-PCNA, p53-MDM2, and MYC-MAX complexes. We also use SpARC-map to probe the purinosome, the weakly bound complex of six purine biosynthetic enzymes, where no PPI interfaces are known. There, we identify interfaces that satisfy structural requirements for substrate channeling, as well as protein surfaces that participate in multiple distinct interactions, which we validate using site-specific photocrosslinking in live human cells. Finally, we show that SpARC-map results can impose stringent constraints on machine learning-based structure prediction.
    Keywords:  protein complex; protein interaction interface; protein–protein interaction; purinosome
    DOI:  https://doi.org/10.1073/pnas.2425774122
  64. J Phys Chem Lett. 2025 Dec 26.
      Lipid composition drives membrane protein sorting, interactions, and function, but the precise mechanistic influence of the membrane on the protein free energy landscape remains largely unresolved. In this study, we probe how lipids reshape the folding landscape of the pH low insertion peptide (pHLIP) using a combination of surface-enhanced and ultrafast two-dimensional infrared spectroscopies. The membrane composition has a direct effect on the peptide's structural transitions: anionic phosphatidylserine lipids promote more efficient, rigid insertion, triggering α-helical folding at higher pH and bypassing partially folded intermediates. In contrast, neutral membranes enforce a pathway with more distinct intermediates marked by prolonged surface-bound states. We also demonstrate that this process is bidirectional, where the peptide insertion actively remodels the membrane, disrupts lipid packing, and enhances water penetration. Together, these results indicate that the lipid bilayer functions as a dynamic, responsive energy landscape that not only guides folding but also adapts to it. This framework advances our understanding of how biological membranes modulate cotranslational folding and mitigate misfolding in vivo.
    DOI:  https://doi.org/10.1021/acs.jpclett.5c03418
  65. JACS Au. 2025 Dec 22. 5(12): 5866-5887
      Covalent modification of therapeutic targets has emerged as a powerful platform for creating clinical drugs and chemical probes. Covalent drugs have evolved from serendipitous discoveries to rationally designed therapeutics, driven by advances in electrophile-first screening technologies. This perspective takes stock of alternative technologies currently available in laboratories and industry that collectively enable targeted covalent inhibitor development across historically "undruggable" targets. We highlight five such technologies: activity-based protein profiling (ABPP), provides functional proteomic mapping to identify ligandable residues; covalent tethering, exploits dynamic chemistry to capture transient pockets; covalent DNA-encoded libraries, leverages trillion-member libraries for multiresidue targeting; phage/mRNA display, which facilitates evolution of covalent macrocyclic peptides; and sulfur-(VI) fluoride exchange (SuFEx), engages residues beyond cysteine. Integration of these approaches with chemoproteomics and artificial intelligence accelerates the discovery of covalent inhibitors with enhanced selectivity and reduced off-target risks. This technological convergence establishes a new paradigm for precision covalent therapeutics, offering innovative solutions to overcome drug resistance and target challenging protein interfaces.
    Keywords:  covalent inhibitor; drug design; electrophile screening; technology convergence; undruggable targets
    DOI:  https://doi.org/10.1021/jacsau.5c01134
  66. bioRxiv. 2025 Dec 16. pii: 2025.12.12.694065. [Epub ahead of print]
      Organelle pH is critical to organelle identity and function. Resident proteins that define each organelle modify transiting cargo proteins, with both retention and trafficking between organelles governed by pH-dependent mechanisms. For example, lysosomal enzymes bind mannose-6-phosphate receptors at the higher pH (∼6.5) of the Golgi and dissociate at the lower pH (∼5.5) of late endosomes 1 . Proteins that stray from the endoplasmic reticulum (ER) are captured by KDEL receptors in the acidic Golgi and returned into the neutral ER 2,3 . This pH-tuned trafficking system compartmentalizes organelle function and prevents mis-localization of critical enzymes 4 . Dysregulated organelle pH disrupts their function and leads to various diseases. Because protons move rapidly in water, the pH within a single organelle is currently assumed to be spatially uniform 5 . Here, using a reporter sensitive from pH 5.5 - 10.5 to map a spectrum of organelles at high resolution, we discovered that pH gradients exist within single, large or long organelles such as the ER and mitochondria, and in membrane-less organelles without ion-transporting proteins such as the nucleolus. These new findings upend our understanding of organellar pH, prompting new questions about proton diffusion within the cell, and its potential consequences on organelle function.
    DOI:  https://doi.org/10.64898/2025.12.12.694065
  67. Autoimmunity. 2026 Dec 31. 59(1): 2602715
      Cell stress, including endoplasmic reticulum (ER) stress, heat shock, and hypoxia, plays a pivotal role in cellular homeostasis and immune regulation, particularly in the intestine. ER stress, a key aspect of cell stress, triggers the unfolded protein response (UPR) to restore balance by managing misfolded proteins or inducing apoptosis if unresolved. The activation of these stress responses has emerged as a critical contributor to intestinal inflammation in conditions like inflammatory bowel disease (IBD). Regulatory T cells (Tregs), vital for maintaining mucosal immune tolerance, are strongly influenced by cellular stress, with the UPR shaping their stability, metabolic programming, and function. Microbial dysbiosis and reduced short-chain fatty acids (SCFAs) disrupt these adaptive pathways, further impairing Treg function. In this review, we explore how UPR signaling shapes Treg metabolism and intestinal inflammation. Identifying gaps in UPR-mediated adaptation and stress thresholds, we thus propose microbiome- and ER-stress-based therapeutic strategies as putative strategies for restoring immune balance in IBD.
    Keywords:  CD; ER stress; IBD; Tregs; UC; UPR pathways
    DOI:  https://doi.org/10.1080/08916934.2025.2602715
  68. MicroPubl Biol. 2025 ;2025
      Copper (Cu) is an essential trace element for cellular metabolism, yet its roles in development are not fully defined. We identified murine cysteine-rich intestinal protein 2 (mCrip2) as a novel Cu-binding protein required for myoblast differentiation. RNA-seq of mCrip2 -deficient cells revealed downregulation of ribosome biogenesis and translation genes. Loss of mCrip2 reduced global protein synthesis by 20-30%, partially mimicking cycloheximide treatment. Interestingly, Cu supplementation restored protein synthesis despite persistent differentiation defects. These findings establish mCrip2 as a Cu-responsive regulator linking metal homeostasis to protein synthesis, suggesting a previously unrecognized connection between Cu availability and translational control in mammalian cells.
    DOI:  https://doi.org/10.17912/micropub.biology.001889
  69. Commun Biol. 2025 Dec 22.
      High-energy-demanding tissues, such as skeletal muscle, rely on mitochondrial proteostasis for proper function. Two key quality-control mechanisms -the ubiquitin proteasome system (UPS) and the release of mitochondria-derived vesicles- help maintain mitochondrial proteostasis, but whether these processes interact remains unclear. Here, we show that CRL5Ozz and its substrate, Alix, localize to mitochondria and together regulate the levels and distribution of the mitochondrial solute carrier Slc25A4, which is essential for ATP production. In Ozz-/- or Alix-/- mice, skeletal muscle mitochondria exhibit similar morphological abnormalities, including swelling and dysmorphism, along with partially overlapping metabolomic alterations. We demonstrate that CRL5Ozz ubiquitinates Slc25A4, targeting it for proteasomal degradation, while Alix facilitates Slc25A4 loading into exosomes for lysosomal degradation. Loss of Ozz or Alix in vivo disrupts the steady-state levels of Slc25A4, impairing mitochondrial metabolism and triggering a switch in muscle fiber composition from oxidative, mitochondria-rich slow to glycolytic fast fibers.
    DOI:  https://doi.org/10.1038/s42003-025-09363-3