bims-imicid Biomed News
on Immunometabolism of infection, cancer and immune-mediated disease
Issue of 2025–08–24
thirteen papers selected by
Dylan Ryan, University of Cambridge



  1. mBio. 2025 Aug 18. e0143625
      Coxiella burnetii, the etiologic agent of Q fever, is a gram-negative intracellular bacterium that infects humans via contaminated aerosols typically while working with livestock. C. burnetii initially targets alveolar macrophages (AMs) to establish a growth niche within a phagolysosome-like compartment termed the Coxiella-containing vacuole (CCV). C. burnetii deploys a type IV secretion system (T4SS) to secrete effector proteins that control host cell functions to benefit the bacterium, orchestrating an immunosuppressive, pro-bacterial environment for replication to high numbers. Although multiple signaling pathways have been characterized in the context of C. burnetii infection, the role of host cell metabolic function in establishing favorable intracellular conditions is undefined. Using a primary human AM model, we show that C. burnetii maintains host oxidative phosphorylation (OXPHOS) at homeostasis in a T4SS-dependent manner. Inhibiting OXPHOS impairs CCV expansion, while preventing glycolysis and fatty acid oxidation does not alter vacuole development. Interestingly, mitochondria are shorter in infected cells, suggesting C. burnetii manipulates mitochondrial function to regulate host metabolism. Finally, endoplasmic reticulum (ER) stress regulates immunosuppressive macrophage activities, and C. burnetii regulates ER stress in a T4SS-dependent manner. Here, we show the involvement of protein kinase R-like endoplasmic reticulum kinase in regulating OXPHOS during infection. Collectively, our results demonstrate that C. burnetii engages human macrophage metabolic processes to establish a replication niche.IMPORTANCECoxiella burnetii causes human Q fever and is a potential bioterrorism threat. In humans, C. burnetii evades host cell killing and establishes a prolonged replication cycle within AMs, which is a critical step toward presentation of acute or chronic disease symptoms. While macrophage metabolism fuels antibacterial activity, we identified key metabolic processes that C. burnetii manipulates to sustain a pro-bacterial growth niche. Currently, few infection models capture C. burnetii interaction with disease-relevant human cells. Here, we used the established primary human AM infection system to characterize bacterial modulation of macrophage metabolism. Our findings advance understanding of C. burnetii-AM interactions and lay the foundation for future therapeutic exploration.
    Keywords:  Coxiella burnetii; PERK; macrophage; metabolism; mitochondria
    DOI:  https://doi.org/10.1128/mbio.01436-25
  2. Nat Commun. 2025 Aug 19. 16(1): 7721
      Viral infection induces robust reprogramming of metabolic pathways in host cells. However, whether host metabolic enzymes detect viral components remains unknown. Our group and others previously identified O-GlcNAc transferase (OGT), an important glucose metabolic enzyme, as a crucial mediator of the antiviral immune responses. Here, by studying a mouse model with a catalytically impaired OGT, we discover a catalytic activity-independent function of OGT in restraining influenza A virus (IAV) infection in addition to its catalytic activity-dependent effect on MAVS-mediated antiviral immunity. Biochemical studies reveal a critical antiviral effect based on OGT interacting with IAV genomic RNA that requires its N-terminal tetracopeptide repeat-4 motif. This interaction causes the translocation of nuclear OGT to cytosolic lipid droplets (LDs) to destabilize LDs-coating perilipin 2, thereby limiting LDs accumulation and in turn virus replication. In sum, our findings reveal OGT as a multifaceted metabolic sensor that integrates MAVS signaling and lipid metabolism to combat viral infection.
    DOI:  https://doi.org/10.1038/s41467-025-63085-y
  3. J Virol. 2025 Aug 19. e0057925
      Human cytomegalovirus (HCMV) infection reprograms metabolism, including lipid synthesis. While several metabolite-related pathways exhibit altered activity in infected cells, the alteration of lipid-related pathways by HCMV has not been examined beyond fatty acid synthesis and elongation. In this study, we addressed this lack of understanding by focusing on phosphatidylcholine (PC), a class of lipids we previously showed is increased by HCMV infection in human foreskin fibroblasts. Here, we expand upon this finding by demonstrating that HCMV infection increases the abundance of PCs in several different fibroblasts and, similarly, in endothelial and epithelial cells. Additionally, HCMV elevates PC levels regardless of the level of confluency, type of growth medium, and presence of serum. Next, we investigated if HCMV alters the activity in the three PC synthesis pathways. We demonstrate that HCMV infection promotes the activity in the de novo PC synthesis pathway using a 13C-choline isotopic tracer and liquid chromatography high-resolution tandem mass spectrometry. Infection did not alter the activity in the other two pathways. Moreover, we examined the kinetics of PC remodeling by HCMV and found that PC synthesis was promoted and the PC lipidome shifted after 24 h post-infection. Furthermore, we found that PC remodeling occurred when DNA synthesis and subsequent steps of virus replication were inhibited by phosphonoacetic acid. Overall, this work suggests that the early steps of HCMV replication promote the reprogramming of host lipid metabolism to ensure the synthesis of a lipidome necessary to support HCMV infection.IMPORTANCEHuman cytomegalovirus (HCMV) is a common herpesvirus that establishes a lifelong and persistent infection in its human host. HCMV infection in most people does not cause overt disease. However, in immunocompromised individuals, severe CMV-associated disease can lead to permanent disabilities and even death. Additionally, congenital CMV is the leading infectious cause of birth defects. Viruses have evolved to hijack host metabolic pathways to facilitate their replication cycle. In this study, we determine that HCMV promotes the activity in the de novo pathway of phosphatidylcholine (PC) synthesis. We demonstrate that the activity in the other PC synthesis pathways, the phosphatidylethanolamine N-methyltransferase and Lands cycles, is unaltered by HCMV infection. Moreover, we found that HCMV infection alters metabolic activity to increase the PC lipidome before 48 h post-infection. Additionally, our results suggest that immediate-early and early gene expression promotes changes in PC lipids. Together, our findings demonstrate that infection promotes the de novo PC pathway to increase PC lipids during the early stages of virus replication.
    Keywords:  human cytomegalovirus; lipid metabolism; metabolic reprogramming; phosphatidylcholine
    DOI:  https://doi.org/10.1128/jvi.00579-25
  4. Cell Rep Med. 2025 Aug 13. pii: S2666-3791(25)00373-8. [Epub ahead of print] 102300
      Subtle variations of micronutrients in the tumor microenvironment often coincide with tumor progression and immune disorders. Nevertheless, the underlying mechanisms of how micronutrients, such as metal ions, influence tumor-intrinsic properties and tumor-immune crosstalk remain inadequately characterized. Here, our integrative analysis of multi-center single-cell, spatial transcriptome sequencing, and bulk RNA sequencing (RNA-seq) cohorts reveals that nasopharyngeal carcinoma (NPC)-specific SRY-box transcription factor 4 (SOX4) governs microenvironmental and cellular zinc metabolism through its downstream target, SLC39A14 (ZIP14), a membrane zinc uptake transporter. Mechanistically, NPC cells enhance zinc uptake and activate Wnt/β-catenin signaling to initiate tumor growth, creating a zinc-deficient niche hostile to T cells. Zinc deficiency of tumor-infiltrating CD8+ T cells impairs LCK phosphorylation and T cell receptor (TCR) signaling, compromising their effector function. Our study elucidates the idea that the SOX4-ZIP14-zinc metabolism axis has a multifactorial effect in NPC, fostering the malignant phenotypes of NPC and suppressing the T cell response through the deprivation of zinc metabolism.
    Keywords:  CD8(+) T cells; LCK signaling; SOX4; ZIP14; immune resistance; nasopharyngeal carcinoma; oncogenesis; tumor microenvironment; zinc metabolism
    DOI:  https://doi.org/10.1016/j.xcrm.2025.102300
  5. Exp Dermatol. 2025 Aug;34(8): e70152
      Platelet-rich plasma (PRP) is a safe, autologous plasma component abundant in cytokines and extracellular vesicles, frequently applied to treat inflammatory disorders. Although PRP demonstrates potential for psoriasis therapy, its underlying mechanism remains insufficiently understood. In this study, various PRP constituents were evaluated in an imiquimod (IMQ)-induced mouse model of psoriasis. PRP, platelet-derived extracellular vesicles (PEVs), and platelet-poor plasma (PPP) were isolated from mice and administered subcutaneously. The data showed that PEVs, rather than PPP, served as the principal anti-psoriatic factor. Furthermore, RNA sequencing and flow cytometry revealed that PEVs markedly suppressed M1 polarisation of macrophages, thereby mitigating psoriatic-like inflammation. In vitro, PEVs delivered encapsulated mitochondria to RAW264.7 cells in a concentration-dependent manner. These functional organelles enhanced oxidative phosphorylation and suppressed glycolysis, driving a metabolic shift favouring an anti-inflammatory phenotype and attenuating the inflammatory response. In conclusion, PEVs emerge as the primary PRP component responsible for inflammatory suppression in psoriasis. Notably, mitochondria transfer mediated by PEVs underscores a promising therapeutic avenue and provides novel insight into the role of platelet derivatives in inflammatory diseases.
    Keywords:  macrophage; mitochondria; platelet‐derived extracellular vesicles; platelet‐rich plasma; psoriasis
    DOI:  https://doi.org/10.1111/exd.70152
  6. Mol Immunol. 2025 Aug 15. pii: S0161-5890(25)00199-3. [Epub ahead of print]186 63-69
      This study seeks to examine the effects of long-distance transportation on Simmental calves. While it is established that long-distance transportation can influence animal health, the specific impacts on glucose and lipid metabolism, as well as immune function in cattle, remain inadequately understood. This study analyzed blood samples from twelve 5-month-old Simmental calves that were transported over a distance of 1100 km from Tongliao City, Inner Mongolia, to Zouping City, Shandong Province, with a transportation duration of 15 h. The investigation focused on comparing alterations in serum glucose metabolism, lipid metabolism, and immune parameters before and after the transportation process. The findings indicated that, in contrast to pre-transportation levels, there was an upregulation of enzymes associated with glycolysis and the pentose phosphate pathway post-transportation, resulting in enhanced glucose utilization. Additionally, levels of creatine kinase and lactate dehydrogenase were found to be elevated. The activities of fatty acid synthase and acetyl-CoA carboxylase also increased, while total cholesterol and triglyceride levels decreased, accompanied by a rise in non-esterified fatty acids. Furthermore, high-density lipoprotein cholesterol levels increased, whereas low-density lipoprotein cholesterol levels decreased. Cortisol levels exhibited an upward trend, and there was a significant increase in pro-inflammatory cytokines, including interleukin-1β, interleukin-6, and tumor necrosis factor-α, as well as immunoglobulin G. In conclusion, long-distance transportation appears to induce disorders in glucose and lipid metabolism, systemic inflammation, and immune dysfunction in Simmental calves.
    Keywords:  Glucose metabolism; Immunity; Lipid metabolism; Simmental calves; Transportation stress
    DOI:  https://doi.org/10.1016/j.molimm.2025.08.007
  7. Cell Mol Immunol. 2025 Aug 20.
      T-cell metabolism plays a pivotal role in defining T-cell functional states. Through analysis of a comprehensive pancancer single-cell transcriptional atlas, we identified SARDH, an enzyme involved in one-carbon (1-C) metabolism, as a potential T-cell metabolic checkpoint. SARDH significantly impacts T-cell fate and function, leading to impaired tumor control efficacy. Knocking down SARDH resulted in sarcosine accumulation and reduced consumption of S-adenosylmethionine (SAM), a critical methyl donor for epigenetic modulation, likely due to the shift in glycine-to-sarcosine homeostasis. Deletion of SARDH increased H3K79me2 modification at NF-κB-activating genes, thereby augmenting NF-κB signaling and T-cell function. Additionally, we observed transcriptional dysregulation of 1-C metabolism within tumors across various cancer types, which was often accompanied by increased sarcosine levels. Sarcosine was found to induce SARDH upregulation, suggesting a feedback mechanism for metabolic homeostasis in T cells within tumors. These findings underscore the potential effects and mechanism of targeting 1-C metabolism, particularly SARDH, as an avenue for cancer therapy.
    Keywords:  1-C metabolism; CD8+ exhausted T cells; Mitochondrial metabolism; SARDH; Sarcosine; Tumor microenvironment
    DOI:  https://doi.org/10.1038/s41423-025-01331-5
  8. Clin Transl Med. 2025 Aug;15(8): e70450
       BACKGROUND: Clinical therapeutic approaches to prevent and treat renal injury in patients with acute kidney injury (AKI) and chronic kidney disease (CKD) induced by calcium oxalate (CaOx) are limited. As a pivotal deacetylase, Sirtuin1 (Sirt1) exhibits notably anti-inflammatory effects, but its metabolic mechanism in regulating CaOx nephropathy remains unexplored.
    METHODS: We analysed organic acid metabolism in kidney using the nontargeted metabolome and identified key targets by RNA-seq. Evaluate renal injury and oxidative stress using techniques such as Positron Emission Tomography-Computed Tomography (PET/CT) and transmission electron microscope. The protective mechanisms of Sirt1 against CaOx-induced kidney injury and subsequent crystal deposition were demonstrated using in vitro coculture systems and in vivo Sirt1 conditional knockout mice.
    RESULTS: We found that Sirt1 has a significant protective effect on renal injury and oxidative stress induced by CaOx. Sirt1 expression decreases in CaOx nephropathy mice, and activation of Sirt1 reduces CaOx-induced kidney injury and crystal deposition by increasing the level of itaconate. In addition, it was found that Sirt1 enhances immunoresponsive gene 1 and inhibits Sdha by trimethylating histones, thereby regulating the oxidation levels of itaconate and succinate. Furthermore, we emphasise the valuable role of Sirt1 agonists and exogenous itaconate in alleviating crystal induced kidney injury.
    CONCLUSIONS: Our study revealed a previously unknown function of Sirt1 in CaOx nephropathy. By regulating itaconate level through epigenetic, Sirt1 protects against renal inflammation and oxidative damage induced by CaOx. Our preclinical data suggest that targeted Sirt1 agonism represents a promising therapeutic intervention for progressive crystallopathic nephropathy, potentially disrupting the inflammation-crystallisation vicious cycle.
    HIGHLIGHTS: Sirt1 is significantly reduced in CaOx-induced nephropathy models in vivo and calcium oxalate monohydrate (COM) induced in vitro. Sirt1 in macrophages alleviated CaOx-induced kidney injury and crystal deposition via elevating itaconate levels. Conditional knockout of Sirt1 in vivo significantly exacerbates renal crystal deposition and function damage. We accurately evaluated CaOx-induced renal inflammation status by micro 18F-FDG PET/CT, and observed macrophage phagocytosis and encapsulation of crystals exposed to epithelial cells through scanning electron microscopy. Sirt1 agonists can be used as preventative and therapeutic agents for CaOx nephropathy, and exogenous supplementation of itaconate effectively alleviated renal crystallisation and inflammatory damage.
    Keywords:  Sirt1; calcium oxalate nephropathy; itaconate; macrophage
    DOI:  https://doi.org/10.1002/ctm2.70450
  9. Eur J Immunol. 2025 Aug;55(8): e70039
      Dendritic cells (DCs) rely on Toll-like receptor 9 (TLR9) to detect unmethylated CpG motifs in microbial DNA, triggering essential immune responses. While the downstream signaling pathways of TLR9 activation are well characterized, their impact on S-palmitoylation is unknown. S-palmitoylation, involving the reversible attachment of palmitic acid to cysteine residues, plays a crucial role in regulating protein function and is catalyzed by the ZDHHC family of palmitoyl-acyltransferases (PATs). In this study, we investigated the S-palmitoylated proteome of bone marrow-derived GM-CSF DCs (GM-DCs) at resting and following TLR9 activation with CpGB. Using the click-chemistry-compatible analog 17-octadecynoic acid (17-ODYA) and mass spectrometry (MS)-based proteomics, we characterized dynamic remodeling of S-palmitoylation in response to TLR9 activation. This included enrichment of targets involved in immune and metabolic pathways. Transcriptomic analysis of mice and human DCs revealed TLR9-driven modulation of PAT-encoding genes. Subsequently, we explored the contribution of Zdhhc9 expression to the regulation of S-palmitoylation in DCs. Using gene knockout approaches, we identified candidate protein targets potentially linked to ZDHHC9 activity. Interestingly, modulation of Zdhhc9 expression alone did not influence DC maturation, suggesting that other PATs might compensate for its activity. Together, our findings reveal a novel layer of regulation in TLR9 signaling mediated by S-palmitoylation.
    Keywords:  S‐palmitoylation; TLR9 signaling; dendritic cells; innate immunity
    DOI:  https://doi.org/10.1002/eji.70039
  10. AIDS Res Hum Retroviruses. 2025 Aug 14.
      Little is known about the relationships between circulating short-chain fatty acids (SCFAs) and genital microbiota, inflammation, and the risk for HIV infection in women. As circulating SCFAs are potentially modifiable, for example, through dietary fiber or probiotics, we investigated association of circulating SCFA levels with these outcomes. We carried out a nested matched case-control study within a randomized trial of an antiretroviral microbicide to prevent HIV infection to study the association between circulating SCFAs and HIV acquisition (primary outcome for case definition), vaginal microbiota, and genital inflammation. Levels of the SCFAs butyrate, acetate, and propionate were quantified in plasma using mass spectrometry. Vaginal microbiota was assessed using metaproteomics and characterized as Lactobacillus dominant (LD) or low Lactobacillus (LL). Genital inflammation was measured using multiplex immunoassays. Logistic regression models were used to study the association of SCFAs with each outcome. Study population (N = 99) characteristics were similar between cases (33 who acquired HIV) and controls (66 who did not acquire HIV). We did not observe any associations between any of the circulating SCFAs with HIV acquisition or with LL vaginal microbiota status. However, there was an inverse association between circulating SCFAs and several pro-inflammatory genital cytokines, including interleukin-6 (IL-6), IL-1α, and IL-8. In our study of women with high risk of HIV infection, higher levels of circulating SCFAs were associated with lower levels of various genital inflammatory markers, but not with HIV acquisition or a LL microbiota profile. Future larger studies, including genital SCFA assessment, are needed to confirm these findings.
    Keywords:  acetate; butyrate; cervicovaginal; molecular bacterial vaginosis; propionate
    DOI:  https://doi.org/10.1177/08892229251366174
  11. bioRxiv. 2025 Aug 14. pii: 2025.08.13.670040. [Epub ahead of print]
      Anopheles darlingi is the principal malaria vector in the Amazon basin, where Plasmodium vivax accounts for the majority of cases. Despite its epidemiological importance, the molecular and microbial determinants of A. darlingi susceptibility to P. vivax remain poorly understood. Here, we investigated vector-parasite-microbiota interactions using experimental infections with field-derived P. vivax gametocytaemic blood, which produced two distinct infection phenotypes: low and high oocyst burdens. Transcriptomic profiling of mosquito midguts across key parasite developmental timepoints revealed that low-infection mosquitoes mounted an early and sustained response characterised by activation of detoxification pathways, redox regulation, aromatic amino acid catabolism, and purine depletion, likely coordinated through neurophysiological cues, which collectively create a metabolically restrictive environment for parasite development. These physiological changes were accompanied by reduced bacterial diversity and enrichment of Enterobacteriales and Pseudomonadales, taxa previously linked to anti- Plasmodium activity. Conversely, high-infection mosquitoes exhibited limited metabolic reprogramming, expansion of Flavobacteriales, and transcriptional signatures consistent with permissive physiological states, potentially associated with reproductive trade-offs. Importantly, low infection outcomes consistently arose from bloodmeals with the lowest gametocyte densities, suggesting that host- and parasite-derived components of the bloodmeal act as early conditioning factors that prime the mosquito midgut for either resistance or susceptibility. These findings reframe A. darlingi vector competence to P. vivax not as a fixed immune trait but as a dynamic outcome of early redox, metabolic, and microbial interactions. They also highlight ecological and physiological targets for transmission-blocking strategies and reinforce the importance of studying vector-parasite interactions in regionally relevant systems.
    DOI:  https://doi.org/10.1101/2025.08.13.670040
  12. Adv Sci (Weinh). 2025 Aug 18. e06307
      Tumor driven immune suppression poses a significant impediment to the success of immunotherapy in ovarian cancer. Among the various mechanisms contributing to immune suppression, intracellular communication facilitated by tumor-derived extracellular vesicles (EVs) within the tumor microenvironment emerges as a pivotal factor influencing tumor growth. Here, it is demonstrated that extracellular vesicle-packaged eIF4E from tumor cells alters protein translation in macrophages, contributing to antitumor immune response. Mechanistically, tumor derived EV-packaged eIF4E significantly enhances the expression of 3-hydroxy-3-methyl-glutaryl-coenzyme A reductase (HMGCR), driving the synthesis and secretion of cholesterol. This, in turn, activates macrophages and causes immunosuppression through the X-box binding protein 1 and Programmed death-ligand 1 (XBP1/PD-L1) axis. Strikingly, both genetic and pharmacological depletion of HMGCR in macrophages effectively restores their antitumor activity. Clinically, elevated HMGCR expression in tumor-associated macrophages is associated with poor survival outcomes in ovarian cancer patients. The pivotal role of eIF4E is underscored here as a key signaling mediator, facilitating the communication between tumor and immune cells via EVs to promote immune suppression and suggesting HMGCR as a potential therapeutic target for tumor immunotherapy.
    Keywords:  PD‐L1; eIF4E; extracellular vesicles; metabolism; ovarian cancer; tumor associated macrophages
    DOI:  https://doi.org/10.1002/advs.202506307
  13. Nat Cardiovasc Res. 2025 Aug 19.
      Septic cardiomyopathy, one manifestation of multiple organ dysfunction syndrome, is a challenging complication in sepsis, and cytopathic hypoxia has been proposed to have a key role in the pathophysiology of multiple organ dysfunction syndrome. However, the underlying mechanisms remain unknown. Here, we show that upregulation of hypoxia-inducible factor-1α (HIF-1α) in cardiomyocytes following lipopolysaccharide (LPS) treatment suppresses mitochondrial respiration via inducible nitric oxide synthase-dependent nitric oxide, leading to cytopathic hypoxia. Cardiac-specific heterozygous deletion of HIF-1α ameliorates mitochondrial and contractile dysfunction in a mouse model of septic cardiomyopathy. Mechanistically, nuclear factor-κB (NF-κB)-mediated upregulation of cyclooxygenase 2 (COX2) and secretory phospholipases A2 (sPLA2) enhances HIF-1α expression following LPS exposure, whereas their inhibition prevents LPS-induced HIF-1α upregulation, cytopathic hypoxia and contractile dysfunction. In addition, phospholipid metabolites (prostaglandins and lysophospholipids/free fatty acids, respectively) stabilize HIF-1α via protein kinase A activation. These findings highlight a crucial role of excessive HIF-1α, driven by LPS-enhanced phospholipid metabolism, in septic cardiomyopathy through induction of cytopathic hypoxia.
    DOI:  https://doi.org/10.1038/s44161-025-00687-1