Your new experience awaits. Try the new design now and help us make it even better

MINI REVIEW article

Front. Immunol., 12 December 2025

Sec. Viral Immunology

Volume 16 - 2025 | https://doi.org/10.3389/fimmu.2025.1740904

This article is part of the Research TopicImmune Modulation at the Crossroads of Viral Infection and Tumor ProgressionView all articles

Multimodal cell death drives the immunopathogenesis of RSV infection

Tianxiang YangTianxiang Yang1Zhizhong Mi*Zhizhong Mi1*Zhaolong Li,*Zhaolong Li1,2*
  • 1Institute of Virology and AIDS Research, The First Hospital of Jilin University, Changchun, Jilin, China
  • 2Department of Infectious Diseases, Infectious Diseases and Pathogen Biology Center, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China

Respiratory syncytial virus (RSV) is a major cause of severe respiratory tract infections in infants, older adults, and immunocompromised individuals. Despite decades of research, effective therapies are limited, largely due to an incomplete understanding of how infected cells and immune responses interact to shape disease outcomes. Recent evidence indicates that RSV activates multiple regulated cell death (RCD) programs-including apoptosis, necroptosis, pyroptosis, ferroptosis, and autophagy-associated cell death which interact through shared molecular mediators to form a multimodal cell death (MMCD) network. This integrated system regulates the balance between viral clearance and immunopathological injury. Central mediators such as caspase-8, RIPK3, and NLRP3 act as molecular hubs coordinating these death programs and amplifying inflammatory responses. Understanding how MMCD shapes RSV immunopathogenesis provides a unified framework linking cell death to immune dysfunction. This review summarizes recent progress in elucidating the MMCD network, highlights its role in death-inflammation feedback loops, and discusses potential strategies for therapeutic modulation. Conceptualizing RSV disease through the lens of MMCD may guide the development of precision interventions that restore immune homeostasis while preserving antiviral defense.

1 Introduction

RSV, a negative-sense RNA virus of the Pneumovirus genus (Paramyxoviridae family), is the leading cause of acute lower respiratory tract infections (ALRIs) in infants and young children worldwide. It also contributes significantly to pneumonia and respiratory failure among older adults and immunocompromised populations. Epidemiological data estimate that RSV infects more than 33 million children under five years of age each year, leading to over 100,000 deaths-most occurring in low- and middle-income countries (1). In older adults, RSV-associated hospitalization and mortality rates are comparable to or even exceed those of seasonal influenza (2), highlighting its growing cross-age public health significance.

At the mechanistic level, RSV infection is characterized by epithelial barrier disruption, mucus hypersecretion, and massive immune cell infiltration within the airway mucosa (3). Viral entry, mediated by the fusion (F) and attachment (G) glycoproteins, leads to syncytium formation. This process also triggers inflammatory signaling through the TLR4-NF-κB axis (4). These viral protein-host interactions generate inflammatory signaling and cellular stress-including oxidative imbalance, endoplasmic reticulum (ER) stress, and mitochondrial dysfunction that act as upstream triggers for RCD pathways. Emerging evidence suggests that this complex immunopathology is underpinned by a coordinated network of RCD pathways that collectively shape viral clearance, inflammation, and tissue remodeling (5). Historically, RSV-induced cell death was attributed mainly to apoptosis (6). However, advances in single-cell transcriptomics and spatial multi-omics have revealed that RSV simultaneously engages multiple RCD programs-apoptosis, pyroptosis, necroptosis, ferroptosis, and autophagy-related death-within the same tissue microenvironment (7, 8). These pathways are interconnected via shared molecular hubs such as caspase-8, RIPK3 (receptor-interacting serine/threonine-protein kinase 3), and NLRP3 (NOD-like receptor family pyrin domain-containing 3), forming a coordinated MMCD network. Despite accumulating evidence, the mechanisms by which MMCD orchestrates immune dysregulation and tissue injury during RSV infection remain unclear. This mini review summarizes current knowledge of MMCD, its immunopathological implications, and its potential as a target for precision intervention.

2 Molecular basis of cell death signaling during RSV infection

The RSV genome is a single-stranded, negative-sense RNA approximately 15.2 kb in length, encoding 11 viral proteins. Among these, the fusion (F) and attachment (G) glycoproteins are central to viral entry and early host signaling. The F protein undergoes proteolytic activation by host enzymes, exposing the fusion peptide that mediates membrane merging and syncytium formation (9). This process simultaneously triggers the TLR4-MAPK-NF-κB axis, promoting the release of proinflammatory cytokines such as IL-6, TNF-α, and CXCL8 (C-X-C motif chemokine ligand 8) (10). The G protein, which contains a CX3C chemokine motif mimicking the host ligand CX3CL1 (C-X3-C motif chemokine ligand 1), binds to CX3CR1 (C-X3-C motif chemokine receptor 1) on epithelial and dendritic cells (11). This interaction facilitates viral attachment while transiently dampening immune cell recruitment and dendritic cell maturation (12), creating an initial phase of immune suppression. G protein-induced calcium dysregulation and oxidative stress further prime infected cells for downstream activation of RCD signaling.

The nonstructural proteins NS1 and NS2 function as potent antagonists of host antiviral defenses (13). The NS1/2 complex inhibits MAVS-mediated RIG-I-IRF3 signaling and promotes STAT2 degradation, thereby suppressing type I interferon responses (14). In addition, NS2 interferes with mitochondrial dynamics by preventing DRP1 (Dynamin-Related Protein 1) phosphorylation, leading to excessive ROS accumulation (15). The resulting oxidative stress activates ER stress-CHOP pathway, downregulates Bcl-2, and increases mitochondrial membrane permeability (16), predisposing cells to apoptosis, necroptosis, or ferroptosis under sustained infection.

Host receptors further contribute to RSV propagation and the integration of death signaling. CX3CR1, IGF1R (Insulin-like growth factor 1 receptor), and P2Y2 (P2Y purinoceptor 2) receptors facilitate viral entry and propagation (17, 18). IGF1R activation transiently delays apoptosis through the PI3K-AKT pathway, while P2Y2-mediated calcium influx promotes viral release. Prolonged ER stress through the PERK-eIF2α-CHOP axis amplifies mitochondrial dysfunction, providing a convergence point for apoptotic, necroptotic, and ferroptotic pathways. Together, these viral and host interactions lay the molecular foundation for the integrated MMCD network that underpins RSV pathogenesis.

3 Forms of regulated cell death in RSV infection

RSV infection activates multiple RCD programs that operate in a coordinated and context-dependent manner (Figure 1), rather than acting independently, apoptosis, pyroptosis, necroptosis, ferroptosis, and autophagy-related death intersect through shared molecular hubs-including caspase-8, RIPK3, NLRP3, and GPX4 (Glutathione Peroxidase 4) that determine whether infected cells undergo containment, inflammatory amplification, or oxidative injury.

Figure 1
Diagram A shows the structure of a virus, labeled with components including large polymerase protein, nucleoprotein, matrix protein, and others, along with a gene map. Diagram B illustrates the mechanism of viral infection and immune feedback, depicting processes like apoptosis, necroptosis, ferroptosis, and autophagic death in a cell, linked to the lung's immune response.

Figure 1. Overview of RSV structure and the integrated MMCD network. (A) Schematic representation of the RSV virion showing surface glycoproteins (F, G, SH), matrix (M), and nucleocapsid components (N-P-L-M2 complex), as well as regulatory proteins NS1/NS2. (B) Viral invasion triggers multiple regulated cell death pathways within infected airway epithelial cells. Caspase-8 acts as a molecular switch linking apoptosis and necroptosis through Caspase-8/9 and RIPK1/3 signaling. Pyroptosis is driven by Caspase-8-NLRP3 activation, while ferroptosis is mediated by GPX4 inhibition and ROS accumulation. Autophagic cell death contributes additional stress signals. These interconnected pathways form an integrated MMCD feedback loop at the core of this circuit lies the Caspase-8-RIPK3-NLRP3 molecular kernel, amplifying oxidative stress and inflammatory signaling. Thus, the resulting immune feedback promotes airway injury and immunopathogenesis in the infected lung.

During early infection, controlled apoptosis and autophagy contribute to viral clearance while limiting inflammation. As infection progresses, inflammatory RCD pathways such as pyroptosis and necroptosis dominate, releasing DAMPs (Damage-associated molecular patterns) that amplify cytokine production and epithelial injury. In later stages, ferroptosis-driven oxidative stress promotes chronic epithelial dysfunction and tissue remodeling. Viral proteins (F, G, NS1/2) and host stress responses cooperatively regulate these transitions through mitochondrial, ER, and lipid metabolic pathways.

Figure 1 depicts the integrated framework in which RSV components trigger signaling cascades that converge on the MMCD network, producing overlapping waves of regulated cell death (RCD) that collectively shape immune pathogenesis. Table 1 summarizes key regulators of these pathways and their corresponding immunopathological outcomes. Together, these findings highlight that RSV pathogenicity stems not from a single mode of cell death, but from the hierarchical integration of multiple death programs that determine the magnitude of inflammation, the failure of tissue repair, and overall disease severity.

Table 1
www.frontiersin.org

Table 1. Representative regulators and immunopathological consequences of RCD during RSV infection.

4 Integrative network and immunopathological coupling

The MMCD network is governed by a central regulatory triad-the Caspase-8-RIPK3-NLRP3 molecular kernel-which serves as the signaling core that balances apoptosis, necroptosis, and pyroptosis. This molecular kernel integrates stress and death cues, ensuring the dynamic coordination of multimodal cell death responses during RSV infection. Under physiological conditions, caspase-8 restrains necroptosis by cleaving RIPK1 and RIPK3. During RSV infection, NS1/2 proteins inhibit caspase-8 activation and promote RIPK3-MLKL assembly, redirecting apoptotic signals toward necroptotic and pyroptotic outputs (27, 28). This triad-based network has also been implicated in SARS-CoV-2 and influenza pathogenesis, underscoring its conserved immunoregulatory role across viral infections. The ensuing potassium efflux and mitochondrial ROS generation further activate NLRP3, establishing a feed-forward inflammatory loop that amplifies tissue injury.

Beyond intracellular regulation, the MMCD network tightly interacts with innate and adaptive immune circuits. DAMPs and alarmins released from necroptotic and pyroptotic cells stimulate dendritic cells and macrophages via TLR4 and RAGE, fueling IL-1β, IL-6, and TNF-α production (29). These cytokines, in turn, promote Th17 polarization and neutrophil recruitment, which reinforce oxidative stress and perpetuate ferroptotic signaling (30, 31). Spatial transcriptomics has revealed that epithelial, myeloid, and stromal cell subsets engage distinct MMCD modules within inflamed lung tissue (32). This spatial integration underscores MMCD as a systems-level mechanism linking cellular fate to immune orchestration.

Together, the Caspase-8-RIPK3-NLRP3 triad functions as the molecular kernel of MMCD, integrating apoptotic, necroptotic, pyroptotic, and ferroptotic signals into a unified immunopathological continuum. This framework redefines RSV-induced injury as a dynamic equilibrium between protective elimination and destructive amplification.

5 Feedback loops between cell death and immune activation

MMCD not only results from infection but also actively shapes immune responses. Distinct forms of RCD release a spectrum of intracellular contents-DAMPs, alarmins, and oxidized lipids-that engage pattern recognition receptors (PRRs) on immune cells. These outputs are orchestrated by the Caspase-8-RIPK3-NLRP3 molecular kernel, which couples regulated cell death to immune amplification. Molecules such as HMGB1, ATP, and mitochondrial DNA activate macrophages and dendritic cells via TLR4, NLRP3, and RAGE, triggering cytokine cascades dominated by IL-1β, IL-6, TNF-α, and type I interferons (33, 34). These cytokines further sensitize epithelial and immune cells to RCD activation by upregulating death receptors such as Fas and TRAILR.

Conversely, sustained immune activation feeds back to intensify cellular stress pathways. Persistent production of IFN-γ and TNF-α induces ER stress, mitochondrial dysfunction, and oxidative damage, shifting the balance from controlled apoptosis to necroptosis and ferroptosis (35). Activated neutrophils and Th17 cells release reactive oxygen and nitrogen species, exacerbating lipid peroxidation and impairing tissue repair. This bidirectional crosstalk establishes a “death-inflammation” loop, in which MMCD-derived signals perpetuate immune activation, and immune effector molecules in turn reinforce multimodal cell death.

Beyond acute infection, MMCD-driven inflammation influences long-term immune remodeling. Repeated epithelial injury and chronic oxidative stress reshape the airway microenvironment, leading to aberrant macrophage polarization and exhaustion of tissue-resident memory T cells (36, 37). Such maladaptive remodeling may contribute to post-RSV wheezing and asthma-like sequelae observed in susceptible individuals. Understanding these feedback dynamics provides insight into how transient infection evolves into chronic immunopathology. Such maladaptive remodeling may explain post-viral airway hyperresponsiveness and provide a mechanistic bridge between infection and chronic respiratory disorders.

Collectively, MMCD acts as both a driver and target of immune regulation, forming a dynamic feedback network that connects cellular fate with immune homeostasis. Dissecting this loop could uncover novel biomarkers for disease severity and identify intervention points where selective modulation of MMCD may break the cycle of inflammation and tissue injury, setting the MMCD as both an effector and regulator of immune homeostasis, warranting integrative therapeutic exploration.

6 Discussion

The concept of MMCD provides a unified framework to interpret the immunopathogenesis of RSV infection. By integrating apoptosis, pyroptosis, necroptosis, ferroptosis, and autophagy-related processes into a coordinated signaling continuum, MMCD explains how the same virus can simultaneously elicit antiviral defense and excessive inflammation. The hierarchical organization of MMCD-ranging from early containment to chronic oxidative injury-clarifies the dynamic balance between protective clearance and destructive immunopathology. This integrative view expands beyond conventional single-pathway models and highlights the Caspase-8-RIPK3-NLRP3 axis as a pivotal regulatory triad governing immune amplification.

From a translational perspective, the MMCD paradigm opens new opportunities for precision immunotherapy. Translating MMCD insights into therapy demands identification of druggable nodes within this integrated network. Pharmacological inhibition of RIPK1, NLRP3, or ferroptosis regulators (e.g. Necrostatin-1, MCC950, Deferoxamine) has shown promise in preclinical models by mitigating epithelial cell necrosis and restoring immune balance (38, 39). Rather than complete suppression, selective modulation of MMCD pathways may recalibrate host responses without compromising antiviral immunity. Moreover, profiling MMCD-associated signatures through single-cell multi-omics or spatial transcriptomics could enable patient stratification and the identification of biomarkers predicting disease severity or treatment response.

Nevertheless, several challenges remain. First, the heterogeneity of MMCD responses across cell types and disease stages complicates therapeutic targeting. Second, pharmacodynamic optimization and delivery barriers limit clinical translation of existing inhibitors. Third, the dual role of MMCD in both defense and pathology raises concerns that broad inhibition may inadvertently impair antiviral immunity. Future studies should therefore adopt context-specific modulation approaches guided by quantitative modeling and in vivo validation. For instance, early-phase inhibition of the NLRP3 inflammasome might suppress excessive epithelial or macrophage pyroptosis and reduce IL-1β-mediated inflammation-a notion supported by recent studies showing amelioration of RSV-induced immunopathology upon NLRP3 blockade (40). In more severe or late-stage disease characterized by widespread necroptosis and cell lysis, targeting RIPK1/RIPK3 or downstream MLKL may prevent necroptotic amplification, consistent with observations of necroptosis involvement in RSV-infected macrophages (41).Integrating MMCD dynamics into systems immunology frameworks could reveal emergent properties that predict therapeutic windows and adverse outcomes.

Current evidence connecting individual MMCD pathways to RSV pathogenesis is largely correlative, as most mechanistic insights are derived from in vitro experiments or animal models rather than human tissues. Establishing true causality will require longitudinal clinical sampling, single-cell-resolved analyses, and functional perturbation studies in physiologically relevant systems. Moreover, although conceptual temporal models-such as “early apoptotic containment” versus “late-stage inflammatory necroptosis or pyroptosis”- provide a useful framework, MMCD programs in vivo are likely to operate simultaneously and are strongly influenced by cell type, tissue organization, metabolic context, and local immune cues. This inherent spatiotemporal complexity poses challenges for precise in vivo validation. Finally, therapeutic targeting of MMCD remains constrained by issues of specificity. Intervening in one pathway may unintentionally impair beneficial antiviral immunity or provoke compensatory death programs. A critical challenge in the therapeutic targeting of necroptosis lies in achieving sufficient pathway specificity while minimizing off-target effects. Kinase inhibitors, particularly those acting on RIPK1 or RIPK3, may exhibit unintended interactions with other signaling kinases, leading to undesirable side effects. Moreover, inhibiting necroptosis can inadvertently redirect cell death signaling toward alternative programmed pathways, such as apoptosis or pyroptosis, due to compensatory network activation. These complexities underscore the need for developing next-generation inhibitors with improved selectivity, temporal control, and context-dependent modulation to ensure therapeutic efficacy and safety.

Future research should therefore focus on dissecting pathway cross-talk and designing compounds that maintain the delicate balance between inhibition and cellular adaptation. Thus, effective clinical translation will require strategies that enable cell-type-selective or spatially restricted modulation of detrimental MMCD signaling while preserving essential host defense mechanisms.

In summary, viewing RSV immunopathogenesis through the lens of MMCD transforms our understanding of virus-host interactions from a linear cascade into an interconnected, self-regulating network. This conceptual shift emphasizes that immunopathology arises not from isolated molecular events but from the dynamic orchestration of multimodal cell death and immune feedback. Elucidating and therapeutically harnessing this network may ultimately pave the way for precision interventions that disrupt immunopathology while preserving host defense in RSV and other viral inflammatory diseases.

7 Methods

We performed a literature search in PubMed, Web of Science using the keywords “RSV”, “RCD”, “autophagy”, “necroptosis”, “pyroptosis”, “ferroptosis”, “NLRP3”, “RIPK3”,”MMCD”and “immunopathogenesis”. Articles published between 2015–2025 were included. Additional references were identified through manual screening of relevant reviews and cross-referencing of primary research articles.

Author contributions

ZL: Writing – review & editing, Supervision, Funding acquisition. TY: Writing – original draft, Writing – review & editing. ZM: Conceptualization, Writing – original draft.

Funding

The author(s) declared that financial support was received for this work and/or its publication. This work was supported in part by funding from the National Natural Science Foundation of China (82341062 and 82472251), and the Key Laboratory of Molecular Virology, Jilin Province (20102209).

Conflict of interest

The author(s) declared that this work was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Generative AI statement

The author(s) declared that generative AI was not used in the creation of this manuscript.

Any alternative text (alt text) provided alongside figures in this article has been generated by Frontiers with the support of artificial intelligence and reasonable efforts have been made to ensure accuracy, including review by the authors wherever possible. If you identify any issues, please contact us.

Publisher’s note

All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.

References

1. Kelleher K, Subramaniam N, and Drysdale SB. The recent landscape of RSV vaccine research. Ther Adv Vaccines Immunother. (2025) 13:1–19. doi: 10.1177/25151355241310601

PubMed Abstract | Crossref Full Text | Google Scholar

2. Terstappen J, Hak SF, Bhan A, Bogaert D, Bont LJ, Buchholz UJ, et al. The respiratory syncytial virus vaccine and monoclonal antibody landscape: the road to global access. Lancet Infect Dis. (2024) 24:e747–61. doi: 10.1016/S1473-3099(24)00455-9

PubMed Abstract | Crossref Full Text | Google Scholar

3. Yagi K, Ethridge AD, Falkowski NR, Huang YJ, Elesela S, Huffnagle GB, et al. Microbiome modifications by steroids during viral exacerbation of asthma and in healthy mice. Am J Physiol Lung Cell Mol Physiol. (2024) 327:L646–60. doi: 10.1152/ajplung.00040.2024

PubMed Abstract | Crossref Full Text | Google Scholar

4. Xiong Y, Tao K, Li T, Zhou Y, Zhang Z, Ou W, et al. Both chebulagic acid and punicalagin inhibit respiratory syncytial virus entry via multi-targeting glycoprotein and fusion protein. J Virol. (2024) 98:e0153624. doi: 10.1128/jvi.01536-24

PubMed Abstract | Crossref Full Text | Google Scholar

5. Girma A. Biology of human respiratory syncytial virus: Current perspectives in immune response and mechanisms against the virus. Virus Res. (2024) 350:199483. doi: 10.1016/j.virusres.2024.199483

PubMed Abstract | Crossref Full Text | Google Scholar

6. Pengyu Y, Chang M, and Chao L. Programmed cell death in human respiratory syncytial virus infection. Front Cell Infect Microbiol. (2025) 15:1719352. doi: 10.3389/fcimb.2025.1719352

Crossref Full Text | Google Scholar

7. Rajan A, Nagaraj D, Bomidi C, Aloisio GM, Murray AM, Schultz EM, et al. Single cell sequencing analysis of respiratory syncytial virus-infected pediatric and adult human nose organoids reveals age differences, proliferative diversity and identifies novel cellular tropism. J Infect. (2025) 91:106617. doi: 10.1016/j.jinf.2025.106617

PubMed Abstract | Crossref Full Text | Google Scholar

8. Shao Q, Liu T, Hu B, and Chen L. Interplay between autophagy and apoptosis in human viral pathogenesis. Virus Res. (2025) 359:199611. doi: 10.1016/j.virusres.2025.199611

PubMed Abstract | Crossref Full Text | Google Scholar

9. Neal HE, Barrett CT, Edmonds K, Moncman CL, and Dutch RE. Examination of respiratory syncytial virus fusion protein proteolytic processing and roles of the P27 domain. J Virol. (2024) 98:e0163924. doi: 10.1128/jvi.01639-24

PubMed Abstract | Crossref Full Text | Google Scholar

10. Dudek I, Czerkies M, and Kwiatek A. Differential expression of cytokines and elevated levels of MALAT1 - Long non-coding RNA in response to non-structural proteins of human respiratory syncytial virus. Virology. (2024) 597:110127. doi: 10.1016/j.virol.2024.110127

PubMed Abstract | Crossref Full Text | Google Scholar

11. Rivas-Fuentes S, Salgado-Aguayo A, Santos-Mendoza T, and Sevilla-Reyes E. The role of the CX3CR1-CX3CL1 axis in respiratory syncytial virus infection and the triggered immune response. Int J Mol Sci. (2024) 25:9800. doi: 10.3390/ijms25189800

PubMed Abstract | Crossref Full Text | Google Scholar

12. Georgakopoulou VE and Pitiriga VC. Immunomodulation in respiratory syncytial virus infection: mechanisms, therapeutic targets, and clinical implications. Microorganisms. (2025) 13:1876. doi: 10.3390/microorganisms13081876

PubMed Abstract | Crossref Full Text | Google Scholar

13. Merritt TN, Pei J, and Leung DW. Pathogenicity and virulence of human respiratory syncytial virus: Multifunctional nonstructural proteins NS1 and NS2. Virulence. (2024) 15:2283897. doi: 10.1080/21505594.2023.2283897

PubMed Abstract | Crossref Full Text | Google Scholar

14. Chorvinsky E, Bhattacharya S, Bera BS, Welham A, Ismat K, Lawlor CM, et al. Down syndrome alters type-III IFN and pro-inflammatory airway epithelial responses to RSV infection. Am J Respir Cell Mol Biol. (2025) 73:951-964. doi: 10.1165/rcmb.2025-0068OC

PubMed Abstract | Crossref Full Text | Google Scholar

15. Li C, Wei Y, Li J, Lai J, Zeng Y, Zhu B, et al. Selenomethionine inhibited RSV infection-induced apoptosis and inflammatory response through ROS-mediated signaling pathway. ACS Omega. (2025) 10:40258–70. doi: 10.1021/acsomega.5c05364

PubMed Abstract | Crossref Full Text | Google Scholar

16. Che S, Xie X, Lin J, Liu Y, Xie J, and Liu E. Andrographolide attenuates RSV-induced inflammation by suppressing apoptosis and promoting pyroptosis after respiratory syncytial virus infection in vitro. Comb Chem High Throughput Screen. (2024) 27:1776–87. doi: 10.2174/0113862073256465231024075452

PubMed Abstract | Crossref Full Text | Google Scholar

17. Hayes RS, Oraby AK, Camargo C, Marchant DJ, and Sagan SM. Mapping respiratory syncytial virus fusion protein interactions with the receptor IGF1R and the impact of alanine-scanning mutagenesis on viral infection. J Gen Virol. (2024) 105:1–11. doi: 10.1099/jgv.0.001951

PubMed Abstract | Crossref Full Text | Google Scholar

18. Wang X, Ren W, Wang P, Dong L, Du H, Li N, et al. Investigating the active components and mechanistic effects of Forsythia suspensa Leaf against RSV via the PI3K/Akt-NLRP3 pathway. Heliyon. (2024) 10:e38285. doi: 10.1016/j.heliyon.2024.e38285

PubMed Abstract | Crossref Full Text | Google Scholar

19. Barra J, Liwski CR, Phonchareon P, Portales-Cervantes L, Gaston D, Karakach TK, et al. IL-5 enhances human mast cell survival and interferon responses to viral infection. J Allergy Clin Immunol. (2025) 155:1968–80. doi: 10.1016/j.jaci.2025.02.025

PubMed Abstract | Crossref Full Text | Google Scholar

20. Jiao Y and Yang R. Berberine alleviates respiratory syncytial virus (RSV)-induced pediatric bronchiolitis and fibrosis via suppressing the HMGB1/TLR4/NF-κB pathway. Microbiol Spectr. (2025) 13:e0090025. doi: 10.1128/spectrum.00900-25

PubMed Abstract | Crossref Full Text | Google Scholar

21. Tang Z, Mao Y, Ruan P, Li J, Qiu X, Meng Y, et al. Drugs targeting CMPK2 inhibit pyroptosis to alleviate severe pneumonia caused by multiple respiratory viruses. J Med Virol. (2024) 96:e29643. doi: 10.1002/jmv.29643

PubMed Abstract | Crossref Full Text | Google Scholar

22. Shen C, Zhang Z, Xie T, Xu J, Yan J, Kang A, et al. Jinxin oral liquid inhibits human respiratory syncytial virus-induced excessive inflammation associated with blockade of the NLRP3/ASC/Caspase-1 pathway. BioMed Pharmacother. (2018) 103:1376–83. doi: 10.1016/j.biopha.2018.04.174

PubMed Abstract | Crossref Full Text | Google Scholar

23. Simpson J, Spann KM, and Phipps S. MLKL regulates rapid cell death-independent HMGB1 release in RSV infected airway epithelial cells. Front Cell Dev Biol. (2022) 10:890389. doi: 10.3389/fcell.2022.890389

PubMed Abstract | Crossref Full Text | Google Scholar

24. Simpson J, Loh Z, Ullah MA, Lynch JP, Werder RB, Collinson N, et al. Respiratory syncytial virus infection promotes necroptosis and HMGB1 release by airway epithelial cells. Am J Respir Crit Care Med. (2020) 201:1358–71. doi: 10.1164/rccm.201906-1149OC

PubMed Abstract | Crossref Full Text | Google Scholar

25. Kombe Kombe AJ, Fotoohabadi L, Nanduri R, Gerasimova Y, Daskou M, Gain C, et al. The role of the nrf2 pathway in airway tissue damage due to viral respiratory infections. Int J Mol Sci. (2024) 25:7042. doi: 10.3390/ijms25137042

PubMed Abstract | Crossref Full Text | Google Scholar

26. Yang X, Liu X, Nie Y, Zhan F, and Zhu B. Oxidative stress and ROS-mediated cellular events in RSV infection: potential protective roles of antioxidants. Virol J. (2023) 20:224. doi: 10.1186/s12985-023-02194-w

PubMed Abstract | Crossref Full Text | Google Scholar

27. Yuan J and Ofengeim D. A guide to cell death pathways. Nat Rev Mol Cell Biol. (2024) 25:379–95. doi: 10.1038/s41580-023-00689-6

PubMed Abstract | Crossref Full Text | Google Scholar

28. Hussain SA, Rohlfing M, Santoro J, Chen P, Muralidharan K, Bochter MS, et al. Neuregulin-1 prevents death from a normally lethal respiratory viral infection. PLoS Pathog. (2025) 21:e1013124. doi: 10.1371/journal.ppat.1013124

PubMed Abstract | Crossref Full Text | Google Scholar

29. Youm YH, Nguyen KY, Grant RW, Goldberg EL, Bodogai M, Kim D, et al. The ketone metabolite β-hydroxybutyrate blocks NLRP3 inflammasome-mediated inflammatory disease. Nat Med. (2015) 21:263–9. doi: 10.1038/nm.3804

PubMed Abstract | Crossref Full Text | Google Scholar

30. Wang C, Huang YM, Zhao J, Bai YM, Yan CQ, Du GH, et al. Fumarprotocetraric acid and geraniin were identified as novel inhibitors of human respiratory syncytial virus infection in vitro. Front Cell Infect Microbiol. (2024) 14:1484245. doi: 10.3389/fcimb.2024.1484245

PubMed Abstract | Crossref Full Text | Google Scholar

31. Chao YY, Puhach A, Frieser D, Arunkumar M, Lehner L, Seeholzer T, et al. Human TH17 cells engage gasdermin E pores to release IL-1α on NLRP3 inflammasome activation. Nat Immunol. (2023) 24:295–308. doi: 10.1038/s41590-022-01386-w

PubMed Abstract | Crossref Full Text | Google Scholar

32. Wang J and Chao J. Alveolar epithelial cell dysfunction in acute respiratory distress syndrome: mechanistic insights and targeted interventions. Biomedicines. (2025) 13:2299. doi: 10.3390/biomedicines13092299

PubMed Abstract | Crossref Full Text | Google Scholar

33. Müller H, Straßmann JK, Baier AS, von Bülow V, Stettler F, Hagen MJ, et al. Liver fibrosis is enhanced by a higher egg burden in younger mice infected with S. mansoni. Cells. (2024) 13:1643. doi: 10.3390/cells13191643

PubMed Abstract | Crossref Full Text | Google Scholar

34. Cruz-Pulido YE, LoMascolo NJ, May D, Hatahet J, Thomas CE, Chu AKW, et al. Polyamines mediate cellular energetics and lipid metabolism through mitochondrial respiration to facilitate virus replication. PLoS Pathog. (2024) 20:e1012711. doi: 10.1371/journal.ppat.1012711

PubMed Abstract | Crossref Full Text | Google Scholar

35. Vieira Antão A, Oltmanns F, Schmidt A, Viherlehto V, Irrgang P, Rameix-Welti MA, et al. Filling two needs with one deed: a combinatory mucosal vaccine against influenza A virus and respiratory syncytial virus. Front Immunol. (2024) 15:1376395. doi: 10.3389/fimmu.2024.1376395

PubMed Abstract | Crossref Full Text | Google Scholar

36. Komiya Y, Kamiya M, Oba S, Kawata D, Iwai H, Shintaku H, et al. Necroptosis in alveolar epithelial cells drives lung inflammation and injury caused by SARS-CoV-2 infection. Biochim Biophys Acta Mol Basis Dis. (2024) 1870:167472. doi: 10.1016/j.bbadis.2024.167472

PubMed Abstract | Crossref Full Text | Google Scholar

37. Zhang M, Lan H, Jiang M, Yang M, Chen H, Peng S, et al. NLRP3 inflammasome mediates pyroptosis of alveolar macrophages to induce radiation lung injury. J Hazard Mater. (2025) 484:136740. doi: 10.1016/j.jhazmat.2024.136740

PubMed Abstract | Crossref Full Text | Google Scholar

38. Shi X, Liu K, Tian Y, Bi X, Zhang J, Ma F, et al. Huaier suppresses lung cancer by simultaneously and independently inhibiting the antioxidant pathway SLC7A11/GPX4 while enhancing ferritinophagy. Cell Death Discov. (2025) 11:309. doi: 10.1038/s41420-025-02598-3

PubMed Abstract | Crossref Full Text | Google Scholar

39. Zhou F, Wang M, Wang Z, Li W, and Lu X. Screening of novel tumor-associated antigens for lung adenocarcinoma mRNA vaccine development based on pyroptosis phenotype genes. BMC Cancer. (2024) 24:28. doi: 10.1186/s12885-023-11757-7

PubMed Abstract | Crossref Full Text | Google Scholar

40. Malinczak CA, Schuler CF, Duran AJ, Rasky AJ, Mire MM, Núñez G, et al. NLRP3-inflammasome inhibition during respiratory virus infection abrogates lung immunopathology and long-term airway disease development. Viruses. (2021) 13:692. doi: 10.3390/v13040692

PubMed Abstract | Crossref Full Text | Google Scholar

41. Bedient L, Pokharel SM, Chiok KR, Mohanty I, Beach SS, Miura TA, et al. Lytic cell death mechanisms in human respiratory syncytial virus-infected macrophages: roles of pyroptosis and necroptosis. Viruses. (2020) 12:932. doi: 10.3390/v12090932

PubMed Abstract | Crossref Full Text | Google Scholar

Keywords: caspase-8, immune amplification, multimodal cell death, NLRP3 inflammasome, regulated cell death, RSV, therapeutic targeting

Citation: Yang T, Mi Z and Li Z (2025) Multimodal cell death drives the immunopathogenesis of RSV infection. Front. Immunol. 16:1740904. doi: 10.3389/fimmu.2025.1740904

Received: 06 November 2025; Accepted: 01 December 2025; Revised: 01 December 2025;
Published: 12 December 2025.

Edited by:

Sheefa Mirza, University of Witwatersrand, South Africa

Reviewed by:

Abayeneh Girma, Mekdela Amba University, Ethiopia

Copyright © 2025 Yang, Mi and Li. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

*Correspondence: Zhaolong Li, bGl6aGFvbG9uZ0BqbHUuZWR1LmNu; Zhizhong Mi, TWl6aGl6aG9uZ0BqbHUuZWR1LmNu

Disclaimer: All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article or claim that may be made by its manufacturer is not guaranteed or endorsed by the publisher.