bims-cesirm Biomed News
on Cell Signaling mediated regulation of metabolism
Issue of 2025–11–02
twenty-six papers selected by
Tigist Tamir, University of North Carolina



  1. Cancer Genomics Proteomics. 2025 Nov-Dec;22(6):22(6): 940-952
       BACKGROUND/AIM: The progression of hormone-sensitive prostate cancer (HSPC) to castration-resistant prostate cancer (CRPC) as a result of resistance to androgen deprivation therapy (ADT) remains a major challenge in prostate cancer treatment.
    MATERIALS AND METHODS: To explore the underlying mechanisms, we performed deep comparative proteomic profiling of HSPC and CRPC cell lines. LNCaP and C4-2 cell lines were cultured in isotopically labeled medium, combined, and digested, followed by liquid chromatography-mass spectrometry (LC-MS/MS) and bioinformatic analyses.
    RESULTS: Using SILAC-based proteomic analysis, 3,578 proteins were identified, with 2,474 quantified. In C4-2 cells, 41 proteins were significantly up-regulated, while 201 were down-regulated (fold-change >1.5 or <1.5-1, p<0.05). KEGG pathway analysis linked the increased proteins to fatty acid metabolism and biosynthesis of unsaturated fatty acids. Lipidomic analysis showed a significant rise in fatty acids like DHA, palmitic acid, stearic acid, and arachidic acid, aligning with the proteomic findings.
    CONCLUSION: These results suggest that fatty acids play a key role in HSPC's progression to CRPC, possibly indicating that CRPC cells themselves may generate fatty acids.
    Keywords:  Prostate cancer; castration-resistant prostate cancer; lipid metabolism; proteomics
    DOI:  https://doi.org/10.21873/cgp.20548
  2. Mol Cell. 2025 Oct 28. pii: S1097-2765(25)00819-6. [Epub ahead of print]
      The de novo purine synthesis pathway is fundamental for nucleotide production, yet the role of mitochondrial metabolism in modulating this process remains underexplored. Here, we identify that succinate dehydrogenase (SDH) is essential for maintaining de novo purine synthesis. Genetic or pharmacological inhibition of SDH suppresses purine synthesis, contributing to a decrease in cell proliferation. Mechanistically, SDH inhibition elevates succinate, which in turn promotes the succinylation of serine hydroxymethyltransferase 2 (SHMT2) within the mitochondrial tetrahydrofolate (THF) cycle. This post-translational modification lowers formate output, depriving cells of one-carbon units needed for purine assembly. In turn, cancer cells activate the purine salvage pathway, a metabolic compensatory adaptation that represents a therapeutic vulnerability. Notably, co-inhibition of SDH and purine salvage induces pronounced antiproliferative and antitumoral effects in preclinical models. These findings reveal a signaling role for mitochondrial succinate in tuning nucleotide metabolism and highlight a dual-targeted strategy to exploit metabolic dependencies in cancer.
    Keywords:  TCA cycle; cancer; formate; mitochondrial metabolism; nucleotide metabolism; succinate
    DOI:  https://doi.org/10.1016/j.molcel.2025.10.002
  3. Sci Adv. 2025 Oct 31. 11(44): eadv6637
      Posttranslational modifications of tubulin give microtubule distinct properties to support diverse cellular functions. Trimethylation on lysine-40 of α-tubulin (α-TubK40me3) is involved in cell division and neuronal development. The "writer" (SETD2) and "reader" (PBRM1) of α-TubK40me3 have been identified. However, the "eraser" of α-TubK40me3 and the impact of α-TubK40me3 dynamic balance on cells are still unclear. Here, we report that KDM4A, a member of the histone demethylase family, binds α-tubulin through its catalytic core domain and demethylates α-tubulin. KDM4A knockout significantly enhances α-TubK40me3, inducing microtubule polymerization and mitotic defects. Furthermore, the overpolymerized microtubules and cell mitotic defects caused by KDM4A knockout are rescued by reducing α-TubK40me3 with overexpression of an α-tubulin mutant α-tubulinK40A or depolymerizing microtubules with nocodazole treatment in cells. Together, our study identifies KDM4A as an α-tubulin demethylase, and this demethylation is important for regulating microtubule polymerization and cell mitosis.
    DOI:  https://doi.org/10.1126/sciadv.adv6637
  4. Front Immunol. 2025 ;16 1599223
       Background: Acetyl-CoA carboxylase alpha (ACACA), a crucial rate-limiting enzyme governing de novo biosynthesis of fatty acids, drives oncogenic metabolic reprogramming in diverse malignancies. However, the multiomics investigation and immunological implications of ACACA across cancers remain unclear.
    Methods: We performed a comprehensive pan-cancer analysis of ACACA via transcriptomic, proteomic, and clinical data from The Cancer Genome Atlas (TCGA), Clinical Proteomic Tumor Analysis Consortium (CPTAC), and the Human Protein Atlas (HPA) databases. Then, single-cell RNA sequencing acquired from the Gene Expression Omnibus (GEO) database was employed to map the expression pattern of ACACA in the tumor microenvironment (TME). Subsequently, functional validation experiments were conducted in lung cancer and sarcoma cells.
    Results: High ACACA expression was associated with poor survival in various cancers, particularly those exhibiting dysregulated lipid metabolism. Immune profiling revealed that elevated ACACA expression was associated with low infiltration of CD8+ T cells and activated natural killer (NK) cells. Single-cell analysis of lung adenocarcinoma revealed that ACACA was expressed predominantly within malignant cells and contributed to an immunosuppressive microenvironment through migration inhibitory factor (MIF) signaling and the extracellular matrix (ECM) remodeling pathway. Furthermore, in vitro studies demonstrated that ACACA inhibition suppresses fatty acid synthesis and tumor growth in lung cancer and sarcoma cells.
    Conclusions: Our study establishes ACACA as a key metabolic regulator that links lipid metabolism to immune evasion and drug resistance, highlighting its potential as a promising therapeutic target across cancers.
    Keywords:  Acetyl-CoA carboxylase alpha (ACACA); drug resistance; pan-cancer analysis; single-cell analysis; tumor microenvironment
    DOI:  https://doi.org/10.3389/fimmu.2025.1599223
  5. BMC Gastroenterol. 2025 Oct 29. 25(1): 772
       BACKGROUND: Metabolic dysfunction-associated steatotic liver disease (MASLD) affects a quarter of the population. Although multiple studies have investigated the mechanisms underlying steatohepatitis and MASLD progression, little attention has been paid to milder cases, particularly those with normal enzyme levels. Our objective was to evaluate the molecular events in patients with MASLD and normal enzyme levels.
    METHODS: We performed a proteomic analysis on serum samples from 83 healthy controls and 50 patients with MASLD. The major findings were then validated in serum samples from 35 controls and 60 patients with MASLD and normal liver enzyme levels (MASLD-NLE), as well as a male mouse model with metabolic dysfunction-associated liver steatosis and normal liver enzymes (mice-NLE) induced by a high-fat diet (HFD).
    RESULTS: Despite having normal enzyme levels, patients with MASLD-NLE exhibited significantly higher levels of ALT, AST, γGGT, hs-CRP, uric acid and creatine compared to controls. Serum proteomics indicated elevated complement-related proteins in patients with MASLD-NLE, evidenced by elevated protein levels of C3 and CFH in both cases using different cutoffs for normal ALT levels. C3 and CFH also showed positive correlations with γGGT, hs-CRP, ALT, and uric acid levels. Additionally, the elevation of C3 and CFH was confirmed in another set of human serum samples, as well as in serum and liver samples of mice-NLE using ELISA.
    CONCLUSIONS: Our study provides novel evidence that the complement system is already activated in patients with MASLD-NLE, potentially offering new avenues for treating MASLD at an early stage.
    Keywords:  Complement system; Liver; MASLD; Normal enzyme level
    DOI:  https://doi.org/10.1186/s12876-025-04361-5
  6. Cancer Genomics Proteomics. 2025 Nov-Dec;22(6):22(6): 929-939
       BACKGROUND/AIM: Lysine lactylation (Kla) is a recently identified post-translational modification derived from lactate that regulates diverse biological processes. Although Kla has been studied in several cancers, its role in prostate cancer (PC) remains unclear. The objective of this study is to profile Kla in PC in order to explore the mechanisms involved in PC progression.
    MATERIALS AND METHODS: We performed global Kla profiling in PC-3M prostate cancer cells using affinity enrichment with anti-Kla antibodies, followed by LC-MS/MS. Bioinformatics analyses were conducted to explore the functional roles of Kla-modified proteins.
    RESULTS: We identified 681 Kla sites across 379 proteins, with modifications predominantly located in nuclear and cytoplasmic proteins. Enrichment analysis indicated Kla involvement in mRNA splicing, chromatin organization, and glycolysis/gluconeogenesis. Several multifunctional proteins, including AHNAK and nucleolin (NCL) harbor multiple Kla sites. Motif analysis indicated conserved amino acid patterns surrounding Kla sites. Notably, PC-3M cells showed reduced expression of sirtuin (SIRT)3, SIRT5, and SIRT6, which may underlie elevated Kla levels.
    CONCLUSION: This study presents the first comprehensive Kla landscape in PCa, suggesting its potential regulatory role in tumor progression. These findings provide a valuable resource for future studies and support Kla as a possible target for therapeutic intervention in prostate cancer.
    Keywords:  Global lactylome; lysine lactylation; prostate cancer cells; protein modification
    DOI:  https://doi.org/10.21873/cgp.20547
  7. Front Oncol. 2025 ;15 1638108
       Background: Cancer cells undergo profound metabolic reprogramming to sustain proliferation, redox homeostasis, and epigenetic remodeling. While the Warburg effect and glutaminolysis have long been recognized as central paradigms, the anabolic and regulatory role of lactate under normoxic conditions remains poorly defined.
    Hypothesis: The Cancer-Induced Lactate Load and Oncologic Remodeling (CILLO) hypothesis proposes that lactate, either imported through MCT1 or produced endogenously, is oxidized to pyruvate by LDHB and subsequently carboxylated to oxaloacetate (OAA) by pyruvate carboxylase. OAA then acts as a metabolic hub driving malate-dependent NADPH production, aspartate synthesis for nucleotide metabolism, activation of the serine/glycine/folate cycle, lipogenesis, and S-adenosylmethionine-mediated epigenetic modifications. In this framework, lactate is no longer a mere by-product of glycolysis but a central integrator of anabolic flux, redox balance, and chromatin dynamics.
    Conclusion: The CILLO hypothesis unifies previously fragmented mechanisms into a coherent paradigm, emphasizing lactate-derived carbon skeletons as active drivers of tumor growth and metabolic plasticity. Key rate-limiting steps-MCT1-mediated uptake, LDHB-dependent oxidation, PC-driven anaplerosis, and PEPCK-M-mediated cataplerosis-emerge as therapeutic nodes for intervention. This model not only advances our understanding of cancer metabolism but also suggests novel strategies for biomarker development, metabolic imaging, and targeted therapies. By reframing lactate as a central determinant of oncologic remodeling, the CILLO hypothesis provides a foundation for translational advances in oncology and personalized medicine.
    Keywords:  CILLO hypothesis; epigenetic regulation; lactate metabolism; metabolic reprogramming; oxaloacetate; pyruvate carboxylase; redox balance
    DOI:  https://doi.org/10.3389/fonc.2025.1638108
  8. Mol Biomed. 2025 Oct 27. 6(1): 87
      Cancer cells rely heavily on de novo pyrimidine synthesis. Inhibiting pyrimidine metabolism directly suppresses tumor growth and fosters immune activation within the tumor microenvironment. Dihydroorotate dehydrogenase (DHODH) is a key enzyme in the de novo pyrimidine synthesis pathway. Inhibiting DHODH can reverse immune suppression and trigger a mild innate immune response. However, the impact of DHODH inhibition on natural killer (NK) cells remains to be explored. In this study, we found that DHODH inhibition promoted NK cell infiltration into tumors efficiently. Mechanistically, DHODH suppression induced mitochondrial oxidative stress, leading to mitochondrial DNA (mtDNA) release into the cytoplasm through voltage-dependent anion channel (VDAC) oligomerization and caspase-3 activation. This subsequently activated the stimulator of interferon gene (STING) pathway, triggered ferroptosis, and induced gasdermin E (GSDME) mediated pyroptosis in cancer cells. These changes collectively facilitated NK cell recruitment. Furthermore, infiltrated NK cells enhanced GSDME-dependent pyroptosis in tumor cells through granzyme release, establishing a positive feedback loop that amplified anti-tumor immunity. Additionally, we developed EA6, a novel DHODH inhibitor that is more effective at promoting NK cell infiltration. In summary, this study reveals that targeting pyrimidine metabolism activates a novel mechanism involving pyroptosis-ferroptosis crosstalk and STING pathway activation to enhance NK cell-mediated immunity. These finding opens new avenues for enhancing the efficacy of targeted nucleotide metabolism in cancer therapy.
    Keywords:  CGAS-STING pathway; DHODH; NK cells; Pyrimidine metabolism; Pyroptosis
    DOI:  https://doi.org/10.1186/s43556-025-00339-7
  9. J Cell Sci. 2025 Oct 29. pii: jcs.264251. [Epub ahead of print]
      Increased aerobic glycolysis and increased cell motility are hallmarks of metastatic cancer. Migrating cancer cells are highly polarized, suggesting glycolytic enzymes could be spatially regulated. Here, we investigated the role of the liver isoform of the "gatekeeper" glycolytic enzyme phosphofructokinase-1 (PFKL) in breast cancer cell migration. Depletion of PFKL significantly decreased migration velocity and directional sensing. We have observed the localization of PFKL to lamellipodia of migrating breast cancer cells where it colocalized with hexokinase-1 and pyruvate kinase M2. We then investigated the functional requirements of PFKL for directional migration. First, we found that expression of catalytically inactive PFKL or indirect pharmacological inhibition of PFKL activity significantly decreased directional migration. Second, we discovered that disrupting PFKL filament formation by expression of a filament incompetent mutant decreased its recruitment to lamellipodia and directional sensing, without altering migration velocity. These findings indicate that both catalytic activity and subcellular localization are required for directional migration in breast cancer cells. These results suggest a novel function of PFKL filaments in cells and provide insight into the function of compartmentalized glycolysis in the cytoplasm.
    Keywords:  Cancer; Cell migration; Chemotaxis; Glycolysis; PFKL; Phosphofructokinase-1
    DOI:  https://doi.org/10.1242/jcs.264251
  10. Cell Rep Methods. 2025 Oct 29. pii: S2667-2375(25)00246-2. [Epub ahead of print] 101210
      Cysteine oxidative modifications are critical signaling events regulating cellular functions, but their low abundance and dynamic nature pose technical challenges. We developed the SICyLIA-TMT workflow, which sequentially labels reduced and reversibly oxidized cysteines with light and heavy iodoacetamide (IAA) within the same sample. The inclusion of tandem mass tags (TMTs) enables simultaneous quantification of oxidative modification dynamics and protein levels across multiple conditions using micrograms of material. To improve the detection of low-abundance oxidized cysteines, a dedicated TMT channel serves as a carrier for heavy IAA-labeled peptides (SICyLIA-cTMT), enhancing quantification and enabling precise stoichiometry calculations. We demonstrate the workflow's applicability to cultured cells and full organs under stress. SICyLIA-cTMT achieves unprecedented depth and accuracy in redox proteome analysis while reducing mass spectrometry time. Combining SICyLIA-TMT with latest mass spectrometry technologies further halves the acquisition time without compromising coverage, improving throughput and enabling comprehensive studies of oxidative signaling.
    Keywords:  CP: biotechnology; cancer; cysteine oxidation; fibroblasts; mass spectrometry; obesity; oxidative signaling; post-translational modification; redox proteomics; redox stress
    DOI:  https://doi.org/10.1016/j.crmeth.2025.101210
  11. J Exp Bot. 2025 Oct 28. pii: eraf477. [Epub ahead of print]
      Reactive oxygen species (ROS) and calcium (Ca2+) are well-established second messengers in plants that transmit signals from an initial stimulus to initiate downstream cellular processes, playing crucial roles in the development and response to adverse environmental conditions of the plant. Upon stimuli, transient increases in ROS and cytosolic free calcium concentration ([Ca2+]cyt) act as early signals. Later, Ca2+ signals can regulate ROS homeostasis by controlling ROS generation, but reciprocally, ROS can modulate Ca2+ signaling by means of post-translational modifications (PTMs) in specific proteins involved in the transport, sensing, and transmission of the signal. Thioredoxins (TRXs) act as ROS scavengers, being able to revert the oxidized, nitrosated, persulfided, and glutathionylated forms of specific proteins, which acts as a PTM reversible mechanism that contributes to fine-tune protein function. In this review, we summarize the crosstalk between Ca2+ and ROS signaling, focusing on the involvement of TRXs.
    Keywords:  Calcium; post-translational modifications; redox; stress; thioredoxin
    DOI:  https://doi.org/10.1093/jxb/eraf477
  12. Nat Commun. 2025 Oct 29. 16(1): 9547
      Vitamin B2 (VB2) metabolism regulates numerous cellular processes, but its role in hepatocellular carcinoma (HCC) progression remains unclear. Here we show that HCC tumors are characterized by upregulation of a VB2 metabolism signature, and VB2 metabolism promotes HCC progression. Among VB2 metabolic enzymes, flavin adenine dinucleotide synthase (FADS) is the only one that is widely overexpressed in human HCC. Elevated FADS expression correlates with resistance to anti-PD-1 therapy and poor prognosis. In vivo, FADS facilitates HCC cell growth and suppresses T cell-mediated antitumor immunity. Single-cell transcriptomic analysis reveals that FADS-induced changes occur both in the tumor cells and the intra-tumoral CD8+ T cells. Knocking down FADS induces HCC cell death and increases CD8⁺ T cell infiltration. Mechanistically, FADS confers ferroptosis resistance on HCC cells via enzymatic function to produce FAD and non-enzymatic function to stabilize PCBP2. Moreover, FADS impairs CD8+ T cell recruitment by disrupting the cGAS-STING pathway. Hesperidin, a clinically approved FADS inhibitor, shows antitumor efficacy in a mouse model. Our study thus highlights the importance of VB2 metabolism in HCC and provides the proof of principle for targeting FADS as a therapeutic strategy for HCC.
    DOI:  https://doi.org/10.1038/s41467-025-64572-y
  13. Cancer Lett. 2025 Oct 28. pii: S0304-3835(25)00676-7. [Epub ahead of print] 218104
      Obesity increases the risk of colorectal cancer (CRC) development, accelerates disease progression and is associated with decreased disease-free survival. Obesity adversely affects the visceral adipose tissue (VAT) leading to increased secretion of extracellular vesicles (EVs). However, the crosstalk between VAT and CRC tumor cells still remains unclear. EVs are lipid-membraned particles that transfer cargo to and/or induce signaling in other cells. Here, we characterized human VAT-derived non-obese (N-OB) and obese (OB) EVs and investigated the functional interaction between CRC cells and VAT-derived EVs. EVs were isolated from VAT obtained from obese (BMI>30) and non-obese patients (BMI<30). Unbiased proteomics revealed that compared to N-OB EVs, OB EVs were enriched with glycolytic enzymes like triose phosphate isomerase (TPI1). This enrichment was associated with increased TPI1 protein levels in CRC cells and elevated glycolytic activity. OB EV-treated cells also exhibited increased stemness-associated genes, 3D-spheroid formation and Apcmin/+ tumoroid self-renewal capacity. In vivo, mice with an adipocyte-specific knockout of EV cargo sorting protein, Tsg101 (Tsg101ΔAd), have altered EV cargo composition with reduced glycolytic enzyme levels. Functionally, Tsg101ΔAd-EVs were able to protect against high-fat diet (HFD)-induced increase in glycolysis and stem-like ability. Moreover, Apcmin/+:Tsg101ΔAd mice were protected against HFD-induced enhanced tumorigenesis. Collectively, this study identifies adipocyte EVs, and its metabolic cargo, as an important regulator of CRC cell metabolism and function, promoting intestinal tumorigenesis.
    Keywords:  CRC; EV; Tsg101; glycolysis; obesity; stem
    DOI:  https://doi.org/10.1016/j.canlet.2025.218104
  14. Nat Cancer. 2025 Oct 27.
      Systemic therapy is the optimal choice for individuals with unresectable or advanced hepatocellular carcinoma (HCC). However its effectiveness is constrained by resistance. Ferroptosis is a unique form of regulated cell death and plays an essential role in HCC systemic therapy. Here we identified that secernin-1 (SCRN1) was closely associated with ferroptosis resistance and poor prognosis in HCC. Specifically, high expression of SCRN1 enhances the interaction of phosphokinase serine/threonine kinase 38 (STK38) and glutathione peroxidase 4 (GPX4) to promote the phosphorylation of GPX4 at S45. This phosphorylation impairs heat shock protein family A member 8 (HSC70) recognition and degradation of GPX4 by chaperone-mediated autophagy, which further alleviates lipid peroxidation and ferroptosis. Our findings reveal a critical mechanism by which tumor cells antagonize ferroptosis through enhanced GPX4 phosphorylation and provide potential targets and strategies for HCC treatment.
    DOI:  https://doi.org/10.1038/s43018-025-01061-7
  15. Angew Chem Int Ed Engl. 2025 Oct 29. e18593
      A central challenge in top-down proteomics (TDP) is the characterization of large proteoforms (>70 kDa) due to their high spectral complexity in mass spectrometers. Here, we advance individual ion mass spectrometry (I2MS) for intact mass and fragmentation analysis of β- and α-catenins (85-110 kDa), key components of adherens junctions. Using denatured I2MS, we resolved discrete phosphorylation states of catenins isolated from HEK cells subjected to differential actomyosin tension. Up to 10 phosphorylations were detected on β-catenin (β-cat) and 7 on α-catenin (α-cat), with site-specific changes corresponding to actomyosin contractility. Notably, phosphorylation at α-cat S641 was constitutive, while other sites in the P-linker and actin-binding domains, as well as β-cat S675 and S552, were sensitive to actomyosin perturbation. Application of I2MS for fragment ion detection (I2MS2) also enabled 25%-30% sequence coverage for these exceptionally large proteoforms, compared to <1% using conventional methods for top-down mass spectrometry (MS). Our results are consistent with a "catenin phospho‑code" model, wherein combinatorial phosphorylation patterns reflect and potentially modulate the mechanotransductive environment at cell-cell adhesions. This work establishes top-down I2MS as a viable approach for probing complex post-translational modification (PTM) landscapes in high-mass proteins and highlights proteoforms as functional units in cellular regulation.
    Keywords:  Catenins; Mass spectrometry; Phosphorylation; Proteoform; Top‐down proteomics
    DOI:  https://doi.org/10.1002/anie.202518593
  16. J Cell Sci. 2025 Oct 15. pii: jcs264159. [Epub ahead of print]138(20):
      This Opinion piece highlights recent advances in technical approaches to dissect the mechanisms coupling cell cycle and ciliogenesis during G0/quiescence and the importance of transient post-translational modifications (PTMs) as dynamic regulators of these processes. We discuss the latest technologies enabling real-time monitoring of context-dependent phosphoproteomics and emerging concepts in PTM-driven control of ciliary function. Additionally, we outline major unanswered questions and propose future research directions. A better understanding of G0 regulatory pathways might both spur the development of clinical interventions for human cilia-linked pathologies and set the stage for future research linking cell quiescence to cancer and tissue regeneration.
    Keywords:  G0; Kinase; Phosphoproteomics; Primary cilia; Quiescence; STAMP; Stem cells; Structural prediction
    DOI:  https://doi.org/10.1242/jcs.264159
  17. ACS Synth Biol. 2025 Oct 28.
      The booming artificial intelligence (AI) technology provides an opportunity to precisely carry out de novo design of enzymes and create new biocatalysts with significantly enhanced performance. In the past decade, successful de novo enzyme design cases, although they yielded modest improvements that fell short of targets, have shown that this ambitious goal is achievable, especially as AI now enables high-accuracy, from-scratch prediction of enzyme structures. Analyzing the structural features of current de novo enzymes highlights the need for greater design precision to create tailored, high-performance biocatalysts on demand. Herein, the main achievements, latest research progress, and numerous emerging innovation opportunities of de novo enzyme design in the context of AI are summarized and discussed. Building on previous in-depth research on the catalytic mechanisms of enzymes, de novo enzyme design is achieved by modeling the most critical transition state in the catalytic reaction. Currently, the dominant approach is an inside-out design strategy. AI-driven de novo enzyme design methods have great potential to enhance model accuracy and now emerge as promising approaches. It is hopeful to overcome the bottleneck of tailoring industrial enzymes, obtain robust and efficient biocatalysts, and thus meet greener and more economical development.
    Keywords:  artificial intelligence; biocatalyst; de novo design; design from scratch; enzyme
    DOI:  https://doi.org/10.1021/acssynbio.5c00452
  18. Nat Commun. 2025 Oct 28. 16(1): 9511
    Tabula Sapiens Consortium
      We develop an open-source package called AnnDictionary to facilitate the parallel, independent analysis of multiple anndata. AnnDictionary is built on top of LangChain and AnnData and supports all common large language model (LLM) providers. AnnDictionary only requires 1 line of code to configure or switch the LLM backend and it contains numerous multithreading optimizations to support the analysis of many anndata and large anndata. We use AnnDictionary to perform the first benchmarking study of all major LLMs at de novo cell-type annotation. LLMs vary greatly in absolute agreement with manual annotation based on model size. Inter-LLM agreement also varies with model size. We find that LLM annotation of most major cell types to be more than 80-90% accurate, and will maintain a leaderboard of LLM cell type annotation. Furthermore, we benchmark these LLMs at functional annotation of gene sets, and find that Claude 3.5 Sonnet recovers close matches of functional gene set annotations in over 80% of test sets.
    DOI:  https://doi.org/10.1038/s41467-025-64511-x
  19. FASEB J. 2025 Oct 31. 39(20): e71170
      Rhizoma Drynariae (RD), derived from the desiccated rhizome of Drynaria fortunei (Kunze) J. Sm., has been researched for its potential therapeutic efficacy in modulating inflammatory responses (IR) and treating osteoporosis (OP). Nevertheless, the molecular mechanisms through which RD attenuates OP by modulating the IR microenvironment have yet to be thoroughly elucidated. Consequently, this study aimed to assess the impact of RD on IR and OP through integrated zebrafish and cellular model assays, focusing on the quantification of neutrophil recruitment, bone mineralization area, and the levels of tumor necrosis factor-α, nitric oxide, interleukin-6, and alkaline phosphatase. The experimental data demonstrate that the < 3 kDa RD fraction (RDE-2) effectively mitigates IR and OP models across both zebrafish and cellular models. Utilizing a novel dose-effect weighted network pharmacology, we identified kaempferol-3-O-rutinoside, procyanidin B2, and prunin as the primary bioactive constituents of RDE-2. Combined with untargeted metabolomics analysis, potential targets for their action were identified. Subsequently, a network comprising active ingredients, core targets, metabolic targets, and metabolites was constructed to elucidate their functional interactions. RT-qPCR analysis revealed a significant upregulation in the mRNA expression levels of AKT1, ESR1, ESR2, MMP9, PI3K, SRC, EP300, NCOA3, Runx2, and CREBBP. The findings suggest that RD has the potential to mitigate OP by modulating the IR microenvironment through the ER/PI3K-EP300 signaling axis. Through the integration of dose-effect weighted network pharmacology and metabolomic analysis, our study advances beyond existing descriptive research on RD and pioneers the elucidation of the ER/PI3K-EP300 axis, thereby offering a novel mechanistic explanation.
    Keywords:  Rhizoma Drynariae; dose‐effect weighted network pharmacology; inflammatory response; metabolomics; osteoporosis
    DOI:  https://doi.org/10.1096/fj.202502419R
  20. Front Endocrinol (Lausanne). 2025 ;16 1614726
      Diabetes mellitus, a condition that significantly elevates the incidence and mortality risks associated with cardiovascular diseases, exacerbates the disease burden in China. Glucagon-like peptide-1 receptor agonists (GLP-1RAs) have garnered considerable attention, as they not only regulate blood glucose but also play a vital role in safeguarding the cardiovascular system. Recent research shows that metabolic reprogramming is a key mechanism for the cardioprotective effects of GLP-1RAs. GLP-1RAs can achieve metabolic reprogramming by regulating fatty acid, glucose, and ketone body metabolism, as well as mitochondrial function. This process optimizes cardiac energy metabolism, alleviates oxidative stress, and reduces the risk of cardiovascular diseases. This review provides a comprehensive summary of the energy metabolism under normal cardiac conditions and the metabolic reprogramming involved in diabetes-related heart disease. The potential applications and challenges of targeted metabolic reprogramming in the cardioprotective effects of GLP-1RAs are further discussed.
    Keywords:  cardiovascular system; diabetes; glucagon-like peptide-1 receptor agonists; heart failure; metabolic reprogramming
    DOI:  https://doi.org/10.3389/fendo.2025.1614726
  21. Front Endocrinol (Lausanne). 2025 ;16 1670637
       Rationale: Corticosteroids play a crucial role in the stress-induced metabolic adjustments, and this stress response is conserved across vertebrates. In teleosts, cortisol is the principal glucocorticoid and regulates metabolic processes predominantly through the activation of the glucocorticoid receptor (GR). In zebrafish (Danio rerio), we recently showed that both the GR and the mineralocorticoid receptor (MR) are essential for stressor perception and metabolic regulation, especially related to glucose production and target-tissue glucose uptake. Here, we tested the hypothesis that GR and MR have distinct roles in modulating the tissue-specific glucose metabolism in response to cortisol stimulation during stress in fish.
    Methods: This was tested using GR knockout (nr3c1-/- ) and either wild-type or MR knockout (nr3c2-/- ) zebrafish treated with cortisol to mimic a chronic stress condition. Stable isotope-labeled glucose (U-13C-glucose) was injected intraperitoneally, and the labeled intermediates were assessed to investigate the fate of the glucose carbon in the serum, liver, and brain. The metabolites in these tissues were analyzed using LC-MS to investigate the 13C incorporation across the metabolic pathway at a systems level.
    Results: Chronic cortisol stimulation enhanced glucose breakdown and its utilization in the TCA cycle for energy production. The GR and MR activation led to distinct and complementary effects on glucose utilization and the generation of TCA intermediates in the brain and liver, suggesting a tissue-specific role for these receptors in energy substrate partitioning during stress in fish.
    Conclusion: Overall, our results underscore the roles of GR and MR activation in elevating circulating energy substrates and facilitating tissue-level oxidative capacity and biomolecule synthesis from glucose metabolism in response to chronic cortisol stimulation in fish.
    Keywords:  brain metabolism; glucocorticoid receptor; intermediary metabolism; metabolomics; mineralocorticoid receptor; stress response
    DOI:  https://doi.org/10.3389/fendo.2025.1670637
  22. Anal Chem. 2025 Oct 31.
      Spatially resolved mass spectrometry (MS)-based multiomics workflows are becoming more utilized for revealing the complex biology that occurs within tissues. However, these approaches commonly require multiple independent tissue sections to analyze the metabolite and protein compositions of these samples. This poses a significant challenge in preserving cell- or region-specific molecular fidelity, as variations between tissue sections can compromise the accurate correlation of molecular data. Here, we developed workflows for comprehensive multiomics profiling from a single tissue section (STS) using different MS modalities. We enhanced the functionality of an electrically insulated substrate by employing metal-assisted approaches that enabled both MS-based untargeted spatial metabolomics and proteomics from STS. This allowed metabolite imaging using matrix-assisted laser desorption/ionization-MS imaging (MALDI-MSI), without compromising it for subsequent proteome profiling with laser capture microdissection (LCM)-based technology. Specifically, implementing copper tape as a backing for polyethylene naphthalate (PEN) slides enabled the detection of >140 metabolites across a poplar root tissue section using MALDI-trapped ion mobility spectrometry time-of-flight (timsTOF)-MS. Afterward, we detected 6571 unique proteins from two distinct root regions by leveraging LCM technology coupled to our microdroplet based sample preparation approach. We also developed an alternative workflow utilizing gold-coated PEN substrates for imaging with MALDI-Fourier-transform ion cyclotron resonance (FTICR)-MS, which permitted the profiling of >170 metabolites and the identification of 6542 unique proteins across a single poplar root tissue section. These results were comparable to using each omics analysis independently. These approaches offer new opportunities for high-resolution molecular profiling of multiple omics levels across biological tissues.
    DOI:  https://doi.org/10.1021/acs.analchem.5c05005
  23. J Immunol. 2025 Oct 31. pii: vkaf250. [Epub ahead of print]
      Cellular immunotherapy has revolutionized the treatment of hematologic malignancies yet has had limited success in the solid tumor microenvironment (TME). While insufficient nutrients can lead to T cell metabolic stress in the TME, the glutamine antagonist DON can paradoxically enhance antitumor immunity. Because DON inhibits both essential and nonessential enzymes whose impairment may contribute to dose-limiting toxicities, mechanisms underlying DON-induced antitumor activity have remained unclear. Here, we aimed to identify specific DON targets that increase T cell antitumor activity and test if more selective inhibition of glutamine metabolism could replicate the effects of DON with reduced toxicity. CRISPR screening in the TME of DON-relevant glutamine metabolizing enzymes identified some targets that were essential in tumor-infiltrating CD8 T cells, but that tumor-infiltrating CD8 T cells lacking the DON target glutamine synthetase (GS) were enriched. Upon adoptive T cell transfers, GS-deficient CD8+ T cells displayed improved survival, a higher proportion TCF-1+ Tox- stem-like cells, and greater antitumor and memory function. GS converts glutamate to glutamine and GS-deficient cells exhibited increased intracellular glutamate and reduced glutathione levels, which correlated with enhanced mitochondrial respiration and resistance to reactive oxygen species. Pharmacological inhibition of GS reduced tumor burden in multiple orthotopic murine tumor models in a manner dependent on adaptive immunity. Our findings establish GS as a key metabolic regulator of CD8+ T cells stress resilience in the TME. By preserving intracellular glutamate, GS inhibition reprograms T cells for improved survival and function, offering a promising therapeutic strategy to enhance immune-based cancer treatments.
    Keywords:  T cell; antitumor immunity; glutamine; glutamine synthetase; immunometabolism
    DOI:  https://doi.org/10.1093/jimmun/vkaf250
  24. J Proteome Res. 2025 Oct 28.
      RSK plays a central role in oncogenic signaling, yet its broader regulatory impact in esophageal squamous cell carcinoma (ESCC) remains unclear. We performed integrated phosphoproteomics and kinase-substrate network analysis to elucidate global signaling alterations following RSK inhibition in ESCC cells. In Eca-109 cells, treatment with a moderate concentration of the selective RSK inhibitor SL0101 effectively suppressed proliferation without inducing apoptosis. To elucidate potential compensatory survival mechanisms, we performed integrated proteomic and phosphoproteomic analyses, which revealed that RSK inhibition provokes broad cellular adaptations. Importantly, the magnitude of these effects varied across pathways. Ribosome biogenesis and mitochondrial organization exhibited substantial perturbation primarily at the proteomic level, reflecting system-level dysregulation. Phosphoproteomic analysis revealed that large-magnitude alterations in nuclear transport and mRNA processing, and more subtle, fine-tuned modulation of mitotic fidelity. Kinase activity inference further identified MAPK14, CDK2, SRPK3, AKT1, PLK1, and PIM2 as principal regulators that may regulate compensatory signaling in response to RSK suppression. Taken together, our study reveals that RSK inhibition reprograms kinase networks to enforce stress adaptation, maintaining oncogenic homeostasis despite perturbations. These findings highlight a potential therapeutic window for early intervention strategies, suggesting that combination strategies may enhance the efficacy of RSK-directed therapies in ESCC.
    Keywords:  ESCC; RSK inhibitor; SL0101; phosphoproteomics; proteomics
    DOI:  https://doi.org/10.1021/acs.jproteome.5c00690
  25. Cell Commun Signal. 2025 Oct 30. 23(1): 468
      Ammonia has long been regarded as the end-toxic product of hepatic metabolism. Under normal physiological conditions, ammonia is metabolized through the urea cycle; however, its metabolic imbalance is closely related to various diseases, including hepatic encephalopathy, liver fibrosis, and cancer. Ammonia-induced cell death, specifically the selective death of immune cells, has emerged in recent years as a new form of cell death in the field of tumor biology, offering a new perspective on the regulation of tumor cell fate. This review creatively focuses on the role of ammonia in tumorigenesis, development, and treatment resistance. We systematically reviewed the sources and dynamic balance of ammonia in the tumor microenvironment and found that it plays a key role in tumor metabolic reprogramming by regulating glutamine metabolism, mitochondrial function, and lysosomal stability in tumor cells. Ammonia can also induce the selective death of immune cells, reshape the immune cell map in the tumor microenvironment, and regulate the anti-tumor immune response. Mechanistically, we analyzed the multi-level network of ammonia metabolism regulation, including the role of glutamine synthetase, the mTOR signaling pathway, and epigenetic modification in ammonia death. In addition, this review emphasizes the importance of ammonia as a potential target for cancer therapy and proposes multimodal strategies combining metabolic regulation and immunotherapy to achieve precision in cancer treatment. Finally, the comprehensive map of ammonia in the tumor ecosystem was constructed, highlighting its potential clinical value as a new anti-cancer target.
    Keywords:  Ammonia; Apoptosis; Autophagy; Immunotherapy; Metabolic reprogramming; Tumor microenvironment
    DOI:  https://doi.org/10.1186/s12964-025-02504-5
  26. Sci Rep. 2025 Oct 29. 15(1): 37890
      Lead (Pb) induces physiological, morphological, and metabolic effects in plants. The exogenous application of ethylenediaminetetraacetic acid (EDTA) and citric acid (CA) is a common method to enhance the phytoextraction of Pb from plants. However, the impact of these treatments on harmel's ability to tolerate and accumulate Pb remains unclear. This study aimed to investigate the effects of CA (0, 2.5, and 5 mM) and EDTA (0, 2.5, and 5 mM) on physiological growth responses, antioxidant enzymes, as well as lead translocation factor (TF) and uptake of harmel under Pb stress (0, 5, and 25 mg L-1). The results indicated that Pb treatment significantly reduced growth parameters in harmel plants. The application of 5 mM EDTA adversely affected plant growth, while the 2.5 and 5 mM CA treatments enhanced it. The application of 2.5 mM CA and EDTA significantly decreased lipid peroxidation and improved biomass production in Pb-stressed plants. The diversity of enzymatic activity in the 2.5 mM EDTA treatments and both CA concentrations was greater than in the control and the 5 mM EDTA treatment. CA and EDTA primarily increased the levels of ascorbate, dehydroascorbate, glutathione, and protein in the leaves, thereby enhancing the Pb tolerance of harmel plants. Furthermore, CA and EDTA significantly elevated TF values, with the highest amount observed in Pb 5 + EDTA 5, compared to plants treated with Pb alone. Correlation analysis, visualized through a heat map, revealed strong positive relationships among all growth indicators and a consistently strong negative correlation between root Pb concentration and plant growth, underscoring root Pb accumulation as the primary constraint on development. Conversely, shoot Pb concentration showed weaker associations, and TF exhibited no consistent correlation with growth suppression. Principal Component Analysis (PCA) effectively distinguished Pb-stressed from treated plants and confirmed the ameliorative effects of CA and EDTA. The PCA further highlighted distinct physiological response patterns induced by the two chelating agents, suggesting different underlying mechanisms of action. Overall, CA and EDTA improved Pb tolerance in harmel by modulating antioxidant defense systems and influencing Pb distribution, offering promising tools for enhancing phytoremediation potential under Pb-contaminated conditions.
    Keywords:  Antioxidant system; Ascorbate; Glutathione; Phytoremediation; Translocation factor
    DOI:  https://doi.org/10.1038/s41598-025-21662-7