bims-cediti Biomed News
on Cell death in innate immunity, inflammation, and tissue repair
Issue of 2025–05–11
thirteen papers selected by
Kateryna Shkarina, Universität Bonn



  1. Cell Discov. 2025 May 05. 11(1): 42
      Caspases are critical regulators of cell death, development, innate immunity, host defense, and disease. Upon detection of pathogens, damage-associated molecular patterns, cytokines, or other homeostatic disruptions, innate immune sensors, such as NLRs, activate caspases to initiate distinct regulated cell death pathways, including non-lytic (apoptosis) and innate immune lytic (pyroptosis and PANoptosis) pathways. These cell death pathways are driven by specific caspases and distinguished by their unique molecular mechanisms, supramolecular complexes, and enzymatic properties. Traditionally, caspases are classified as either apoptotic (caspase-2, -3, -6, -7, -8, -9, and -10) or inflammatory (caspase-1, -4, -5, and -11). However, extensive data from the past decades have shown that apoptotic caspases can also drive lytic inflammatory cell death downstream of innate immune sensing and inflammatory responses, such as in the case of caspase-3, -6, -7, and -8. Therefore, more inclusive classification systems based on function, substrate specificity, or the presence of pro-domains have been proposed to better reflect the multifaceted roles of caspases. In this review, we categorize caspases into CARD-, DED-, and short/no pro-domain-containing groups and examine their critical functions in innate immunity and cell death, along with their structural and molecular mechanisms, including active site/exosite properties and substrates. Additionally, we highlight the emerging roles of caspases in cellular homeostasis and therapeutic targeting. Given the clinical relevance of caspases across multiple diseases, improved understanding of these proteins and their structure-function relationships is critical for developing effective treatment strategies.
    DOI:  https://doi.org/10.1038/s41421-025-00791-3
  2. Int J Mol Sci. 2025 Apr 09. pii: 3512. [Epub ahead of print]26(8):
      Traumatic brain injury (TBI) pathology is significantly mediated by an inflammatory response involving inflammasome activation, resulting in the release of interleukin (IL)-1β and pyroptotic cell death through gasdermin-D (GSDMD) cleavage. Inflammasome components are transported through extracellular vesicles (EVs) to mediate systemic inflammation in peripheral organs, including the gut. The purpose of this study was to determine the protective effect of GSDMD knockout (KO) on TBI-induced inflammasome activation, EV signaling, and gut function. GSDMD-KO and C57BL6 (WT) mice were subjected to the controlled cortical impact model of TBI. Cytokine expression was assessed with electrochemiluminescent immunoassay and immunoblotting of the cerebral cortex and gut. EVs were examined for pathology-associated markers using flow cytometry, and gut permeability was determined. GSDMD-KO attenuated IL-1β and IL-6 expression in the cerebral cortex and reduced IL-1β and IL-18 in the gut 3 days post-injury. GSDMD-KO mice had decreased neuronal- and gut-derived EVs compared to WT mice post-TBI. GSDMD-KO EVs also had decreased IL-1β and different surface marker expression post-TBI. GSDMD-KO mice had decreased gut permeability after TBI. These data demonstrate that GSDMD ablation improves post-TBI inflammation and gut pathology, suggesting that GSDMD may serve as a potential therapeutic target for the improvement of TBI-associated pathologies.
    Keywords:  IL-1β; exosomes; extracellular vesicles; gut dysfunction; inflammasome; interleukin; pyroptosis; traumatic brain injury
    DOI:  https://doi.org/10.3390/ijms26083512
  3. Front Immunol. 2025 ;16 1565252
       Introduction: Neutrophil Extracellular Traps (NETs) are vital for innate immunity, playing a key role in controlling pathogen and biofilm proliferation. However, excessive NETosis is implicated in autoimmunity, inflammatory and neoplastic diseases, as well as thrombosis, stroke, and post-COVID-19 complications. Managing NETosis, therefore is a significant area of ongoing research. Herein, we have identified a peptide derived from HMGB1 that we have modified via a point mutation that is referred to as mB Box-97. In our recent study in a murine lung infection model, mB Box-97 was shown to be safe and effective at disrupting biofilms without eliciting an inflammatory response typically associated with HMGB1. Here we show that the lack of an inflammatory response of mB Box-97 is in part due to the inhibition of NETosis of which we investigated the mechanism of action.
    Methods: mB Box-97's anti-NETosis activity was assessed using human neutrophils with known NET inducers PMA, LPS, or Ionomycin. Additionally, mB Box-97's binding to Protein Kinase C (PKC), in addition to downstream effects on NADPH oxidase (NOX) activation, Reactive Oxygen Species (ROS) generation and thereby NETosis were assessed.
    Results: mB Box-97 significantly inhibited NETosis regardless of the type of induction pathway. Mechanistically, mB Box-97 inhibits PKC activity likely through direct binding and thereby reduced downstream activities including NOX activation, ROS production and NETosis.
    Conclusions: mB Box-97 is a promising dual acting therapeutic candidate for managing NET-mediated pathologies and resolving biofilm infections. Our results reveal that PKC is a viable target for NETosis inhibition independent of NET inducer and worthy of further study. These findings pave the way for a novel class of therapeutics aimed at controlling excessive NETosis, potentially offering new treatments for a range of inflammatory and immune-related diseases.
    Keywords:  COVID-19; HMGB1; NET inhibition; autoimmunity; inflammation; therapeutic
    DOI:  https://doi.org/10.3389/fimmu.2025.1565252
  4. Cell Immunol. 2025 May 03. pii: S0008-8749(25)00046-2. [Epub ahead of print]411-412 104961
      The innate immune system recognizes pathogen-associated molecular patterns (PAMPs) and damage associated molecular patterns (DAMPs) through pattern recognition receptors (PRRs). Inflammasomes, cytoplasmic protein complexes, are activated in response to PAMPs and DAMPs, leading to the release of inflammatory cytokines such as IL-1β and IL-18. NLRP3 inflammasome is one of the best characterized inflammasomes and recently its activation has been associated with granuloma formation, structures that aggregate immune cells in response to infections, such as those caused by bacteria, fungi and parasites, and autoinflammatory diseases, such as sarcoidosis. Activation of NLRP3 inflammasomes in macrophages induces the release of cytokines that recruit immune cells, such as monocytes and lymphocytes, to the site of infection. Neutrophils, monocytes, T and B lymphocytes are important in the formation and maintenance of granulomas. Although NLRP3 plays a key role in the immune response, cell recruitment and granuloma formation, many aspects of its function in different cell types remain to be elucidated. In this review, we aim to outline the NLRP3 inflammasome not only as a protein complex that aids innate immune cells in combating intracellular pathogens but also as a platform with broader implications in orchestrating immune responses. This underexplored aspect of the NLRP3 inflammasome presents a novel perspective on its involvement in immunity. Thus, we review the current understanding of the role of the NLRP3 inflammasome in immune cell infiltration and its significance in the organization and formation of granulomas in inflammatory diseases.
    DOI:  https://doi.org/10.1016/j.cellimm.2025.104961
  5. Transpl Int. 2025 ;38 14223
      During brain-death, increased numbers of neutrophils are recruited to organs as part of the inflammatory response. In the organ microenvironment, the recruited neutrophils may release neutrophil extracellular traps (NETs) through interaction with various pro-inflammatory stimuli, contributing to brain-death-induced endothelial activation, microthrombus formation and ultimately a decline in organ quality. To investigate whether NETs form in organs from brain-dead donors; kidneys, hearts, livers, and plasma samples were collected from brain-dead or sham-operated rats. The presence of NET-specific components, neutrophils and macrophages were analyzed through immunofluorescent microscopy. Endothelial activation and platelet infiltration were analyzed through immunohistochemistry and qRT-PCR analysis. Plasma free thiol levels were used to evaluate systemic oxidative stress. Increased neutrophils, NETs and NET/neutrophil ratios were observed in kidneys, hearts and livers of brain-dead rats compared to sham-operated rats. Numbers of NETs positively correlated with the extent of endothelial cell activation. Brain-dead animals also had increased kidney and liver macrophages, increased infiltrated platelets in the liver, and elevated systemic oxidative stress, compared to sham-operated animals. Our findings established the presence of NETs in organs from a brain-dead donor model and suggest that NETs, alongside increased inflammation and a redox imbalance, might prime organs for microvascular endothelial dysfunction and increased injury during brain-death.
    Keywords:  brain-death; donor; endothelial activation; neutrophil extracellular traps; neutrophils
    DOI:  https://doi.org/10.3389/ti.2025.14223
  6. Immunity. 2025 Apr 29. pii: S1074-7613(25)00170-0. [Epub ahead of print]
      Emerging evidence indicates that a baseline level of controlled innate immune signaling is required to support proper brain function. However, little is known about the function of most innate immune pathways in homeostatic neurobiology. Here, we report a role for astrocyte-dependent inflammasome signaling in regulating hippocampal plasticity. Inflammasomes are multiprotein complexes that promote caspase-1-mediated interleukin (IL)-1 and IL-18 production in response to pathogens and tissue damage. We observed that inflammasome complex formation was regularly detected under homeostasis in hippocampal astrocytes and that its assembly is dynamically regulated in response to learning and regional activity. Conditional ablation of caspase-1 in astrocytes limited hyperexcitability in an acute seizure model and impacted hippocampal plasticity via modulation of synaptic protein density, neuronal activity, and perineuronal net coverage. Caspase-1 and IL-18 regulated hippocampal IL-33 production and related plasticity. These findings reveal a homeostatic function for astrocyte inflammasome activity in regulating hippocampal physiology in health and disease.
    Keywords:  IL-18; IL-33; astrocytes; caspase-1; hippocampal plasticity; inflammasome; memory; neuroimmunology; seizure
    DOI:  https://doi.org/10.1016/j.immuni.2025.04.007
  7. Front Transplant. 2025 ;4 1575703
      This Part 2 of a bipartite review commences with the delineation of a conceptual model outlining the fundamental role of injury-induced regulated cell death (RCD) in the release of DAMPs that drive innate immune responses involved in early inflammation-related allograft dysfunction and alloimmune-mediated allograft rejection. In relation to this topic, the focus is on the divergent role of donor and recipient dendritic cells (DCs), which become immunogenic in the presence of DAMPs to regulate alloimmunity, but in the absence of DAMPs acquire tolerogenic properties to promote allotolerance. With respect to this scenario, proposals are then made for leveraging RCD and DAMPs as biomarkers during normothermic regional perfusion (NRP) and normothermic machine perfusion (NMP) of transplant organs from DCD donors, a strategy poised to significantly enhance current policies for assessing donor organ quality. The focus is then on the ambitious goal to target RCD and DAMPs therapeutically during NRP and NMP, aiming to profoundly suppress subsequently early allograft inflammation and alloimmunity in the recipient. This strategic approach seeks to prevent the activation of intragraft innate immune cells including DCs during donor organ reperfusion in the recipient, which is driven by ischemia/reperfusion injury-induced DAMPs. In this context, available inhibitors of various types of RCD, as well as scavengers and inhibitors of DAMPs are highlighted for their promising therapeutic potential in NRP and NMP settings, building on their proven efficacy in other experimental disease models. If successful, this kind of therapeutic intervention should also be considered for application to organs from DBD donors. Finally, drawing on current global insights into the critical role of RCD and DAMPs in driving innate inflammatory and (allo)immune responses, targeting their inhibition and/or prevention during normothermic perfusion of transplant organs from DCD donors - and potentially DBD donors - holds the transformative potential to not only alleviate transplant dysfunction and suppress allograft rejection but also foster allograft tolerance.
    Keywords:  DAMP-induced alloimmunity; DBD; DCD; allorecognition; allotolerance; donor and recipient dendritic cells; normothermic machine perfusion; normothermic regional perfusion
    DOI:  https://doi.org/10.3389/frtra.2025.1575703
  8. Front Transplant. 2025 ;4 1571516
      This Part 1 of a bipartite review commences with a succinct exposition of innate alloimmunity in light of the danger/injury hypothesis in Immunology. The model posits that an alloimmune response, along with the presentation of alloantigens, is driven by DAMPs released from various forms of regulated cell death (RCD) induced by any severe injury to the donor or the donor organ, respectively. To provide a strong foundation for this review, which examines RCD and DAMPs as biomarkers and therapeutic targets in normothermic regional perfusion (NRP) and normothermic machine perfusion (NMP) to improve outcomes in organ transplantation, key insights are presented on the nature, classification, and functions of DAMPs, as well as the signaling mechanisms of RCD pathways, including ferroptosis, necroptosis, pyroptosis, and NETosis. Subsequently, a comprehensive discussion is provided on major periods of injuries to the donor or donor organs that are associated with the induction of RCD and DAMPs and precede the onset of the innate alloimmune response in recipients. These periods of injury to donor organs include conditions associated with donation after brain death (DBD) and donation after circulatory death (DCD). Particular emphasis in this discussion is placed on the different origins of RCD-associated DAMPs in DBD and DCD and the different routes they use within the circulatory system to reach potential allografts. The review ends by addressing another particularly critical period of injury to donor organs: their postischemic reperfusion following implantation into the recipient-a decisive factor in determining transplantation outcome. Here, the discussion focuses on mechanisms of ischemia-induced oxidative injury that causes RCD and generates DAMPs, which initiate a robust innate alloimmune response.
    Keywords:  DAMPs; donation after brain death; donation after circulatory death; injuries to donor organs; innate alloimmunity; normothermic machine perfusion; normothermic regional perfusion; regulated cell death
    DOI:  https://doi.org/10.3389/frtra.2025.1571516
  9. Cell Rep. 2025 May 06. pii: S2211-1247(25)00429-2. [Epub ahead of print]44(5): 115658
      Histones have an important role in eukaryotic innate immunity, wherein histones co-localize with antimicrobial peptides (AMPs). The mechanism of histone cooperation with AMPs and the extent to which histones form pores both remain a mystery. Here, we show that histones form large pores in bacterial membranes that lack lipopolysaccharide (LPS) and that their antimicrobial effect is significantly stronger than that of the clinical AMP polymyxin B. We find that histones and AMPs together produce potent antimicrobial synergy through the formation of 26 nm pores, whereby the pore-forming activity of AMPs on LPS-containing membranes enables histones to enter the periplasmic space and subsequently attack unprotected membranes to create pores. We provide a mechanistic explanation for the long-standing observations of histone antimicrobial activity and demonstrate how antimicrobial synergy arises. The ubiquity of histones and AMPs in innate immunity has significant implications for organismal defense and can be leveraged for novel antibiotic strategies.
    Keywords:  CP: Microbiology; antibiotics; antimicrobial peptides; antimicrobial synergy; bacterial membrane pores; colistin; histones; innate immunity; lipopolysaccharide; polymyxin; pore formation
    DOI:  https://doi.org/10.1016/j.celrep.2025.115658
  10. Biomed Opt Express. 2025 Apr 01. 16(4): 1602-1615
      Mitochondria plays an important role in cell death and undergoes dramatic changes in states of disequilibrium. As mitochondria respond sensitively to cell stress, their dynamics should be studied without affecting cell state. However, current methods rely on labeling cells with fluorescence and introduce additional stress to the cell due to photobleaching and phototoxicity. Here, we propose to use label-free organelle-specific phase contrast microscopy (OS-PCM) to achieve prolonged, specific observation and quantitative analysis of mitochondria dynamics during cell death with minimum perturbation to cells. Using apoptosis and ferroptosis as two examples of cell death, we show quantitatively that large mitochondria tend to increase in size through a combination of swelling and fusion in response to apoptosis, while they decrease in size through fission during ferroptosis. These results provide a new and deeper understanding of mitochondrial dynamics during cell death and demonstrate that OS-PCM is a powerful tool for the gentle, facile, and quantitative study of delicate organelles under stress.
    DOI:  https://doi.org/10.1364/BOE.557745
  11. EMBO J. 2025 May 09.
      Annexin A5 (AnxA5) is a Ca2+-dependent phospholipid-binding protein associated with the regulation of intracellular Ca2+ homeostasis. However, the precise role of AnxA5 in controlling mitochondrial Ca2+ signaling remains elusive. Here, we introduce a novel function of AnxA5 in regulating mitochondrial Ca2+ signaling. Our investigation revealed that AnxA5 localizes at and in the mitochondria and orchestrates intermembrane space Ca2+ signaling upon high Ca2+ elevations induced by ER Ca2+ release. Proximity ligation assays and co-immunoprecipitation revealed a close association but no direct contact of AnxA5 with the voltage-dependent anion channel (VDAC1) in the outer mitochondrial membrane (OMM). In single-cell mitochondrial Ca2+ measurements and electrophysiological recordings, AnxA5 was found to enhance Ca2+ flux through the OMM by promoting the Ca2+-permeable state of VDAC1. By modulating intermembrane space Ca2+ signaling, AnxA5 shapes mitochondrial ultrastructure and influences the dynamicity of the mitochondrial Ca2+ uniporter. Furthermore, by controlling VDAC1's oligomeric state, AnxA5 is protective against cisplatin and selenite-induced apoptotic cell death. Our study uncovers AnxA5 as an integral regulator of VDAC1 in physiological and pathological conditions.
    Keywords:  Annexin-A5; Apoptotic Cell Death; Intermembrane Space Ca2⁺ Signaling; VDAC1 Ca2+ Permeability
    DOI:  https://doi.org/10.1038/s44318-025-00454-9
  12. FEBS J. 2025 May;292(9): 2287-2305
      Membrane blebbing is a hallmark of apoptotic cell death. However, the molecular mechanism that regulates this event has not been fully elucidated. To understand this underlying mechanism, we developed visualization systems suitable for spatiotemporal analysis. By monitoring the plasma membrane labeled with a fluorescent protein and reconstructing the image data as three-dimensional (3D) volumes based on the rendering technique, we observed that dying cells exhibit cycles of bleb formation at the same region of the cell surface. In addition, a Förster Resonance Energy Transfer (FRET)-based biosensor incorporating a regulatory myosin light chain (RMLC) displayed phosphorylation at the base of the retracting bleb, and dephosphorylation before re-expansion, implying the involvement of not only a kinase but also a phosphatase in the regulation of RMLC. To extend these observations, we focused on a scaffold protein, myosin phosphatase Rho interacting protein (MPRIP), which interacts with RhoA and myosin phosphatase targeting subunit 1 (MYPT1), involved in activation of Rho-associated coiled-coil kinase-I (ROCK-I) or protein phosphatase 1 (PP1), respectively. We found that MPRIP is cleaved both in dying cells and in an in vitro cleavage assay in a caspase-dependent manner. A cleaved C-terminal peptide fragment maintains the interaction with MYPT1. Cytological analysis showed that this fragment forms a complex with MYPT1 and myosin after translocating to the cytoplasm. These results suggest that this complex formation promotes the dephosphorylation of RMLC. Collectively, our study indicates that repetitive bleb formation, which is unique to apoptosis, is regulated by both phosphorylation and dephosphorylation of RMLC through MPRIP in a coordinated manner.
    Keywords:  MPRIP; MYPT1; apoptosis; bleb formation; myosin light chain
    DOI:  https://doi.org/10.1111/febs.17422
  13. Cell Rep. 2025 May 06. pii: S2211-1247(25)00427-9. [Epub ahead of print]44(5): 115656
      Neuroinflammation is a well-established risk factor for various neurological disorders and cognitive decline. However, the precise molecular mechanisms linking inflammation with neuropsychiatric symptoms remain unclear. Here, using NLRP3 (NOD-like receptor family, pyrin domain-containing protein 3) conditional knockin (cKI) mice harboring a D301N point mutation originating in patients with autoinflammatory diseases, we found that activation of the NLRP3 inflammasome by administration of lipopolysaccharide induced anxiety-like and repetitive behaviors frequently found in patients with neuropsychiatric disorders, as well as increasing NMDAR (N-methyl-D-aspartate receptor)-mediated excitatory synaptic functions in the medial prefrontal cortex of mice. In addition, interleukin 1β (IL-1β), a downstream cytokine of the NLRP3 inflammasome, enhanced NMDAR activation and increased surface levels of the selective NMDAR subunit GluN2A in cultured cortical neurons. Strikingly, treatment with an NMDAR antagonist or IL-1 receptor antagonist completely normalized the specific behavioral deficits in Nlrp3D301N-cKI mice. Collectively, our results demonstrate that NLRP3-mediated neuroinflammation elicits repetitive behavior through impaired NMDAR functions.
    Keywords:  CP: Immunology; CP: Neuroscience; NLPR3; NMDAR; excitatory synapse; neuroinflammation; repetitive behavior
    DOI:  https://doi.org/10.1016/j.celrep.2025.115656