Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (2,538)

Search Parameters:
Keywords = interferon responses

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
19 pages, 2498 KB  
Article
Multi-Modal Biomarker Profiling of Tumor Microenvironment and Genomic Alterations to Enhance Immunotherapy Stratification in Melanoma
by Meshack Bida, Thabiso Victor Miya, Tebogo Marutha, Rodney Hull, Mohammed Alaouna and Zodwa Dlamini
Curr. Issues Mol. Biol. 2025, 47(10), 821; https://doi.org/10.3390/cimb47100821 - 3 Oct 2025
Abstract
Tumor mutational burden (TMB) and tumor-infiltrating lymphocytes (TILs) are key biomarkers for predicting immunotherapy responses in cutaneous melanoma. The discordance between brisk TIL morphology and absent cytokine signals complicates immune profiling. We examined the interactions between TMB, TIL patterns, cytokine expression, and genomic [...] Read more.
Tumor mutational burden (TMB) and tumor-infiltrating lymphocytes (TILs) are key biomarkers for predicting immunotherapy responses in cutaneous melanoma. The discordance between brisk TIL morphology and absent cytokine signals complicates immune profiling. We examined the interactions between TMB, TIL patterns, cytokine expression, and genomic alterations to uncover immune escape mechanisms and refine prognostic tools. A structure-based BRAF druggability analysis was performed to anchor the genomic findings in a therapeutic context. Primary cutaneous melanoma cases (N = 205) were classified as brisk (n = 65), non-brisk (n = 60), or absent TILs (n = 80) according to the American association for cancer research (AACR) guidelines. Inter-observer concordance was measured using intraclass correlation. Tumor necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ) levels were graded using immunohistochemistry. Eleven brisk TIL cases lacking TNF-α expression were analyzed using the (Illumina TruSight Oncology 500, Illumina-San Diego, CA, USA). Dabrafenib docking to the BRAF ATP site was performed with Glide SP/XP and rescored with Prime MM-GBSA. Brisk TILs lacking cytokine signals suggested post-translational silencing of TNF-α/IFN-γ. Among the 11 profiled cases, eight exhibited high TMB and copy number alterations, with enrichment of nine metastasis/immune regulation genes. Inter-observer concordance was high (absent TILs, 95%; brisk TILs, 90.7%). BRAF docking yielded a canonical type-I pose and strong ATP pocket engagement (ΔG_bind −84.93 kcal·mol−1). Single biomarkers are insufficient for diagnosis. A multiparametric framework combining histology, cytokine immunohistochemistry (IHC), and genomic profiling enhances stratification and reveals immune escape pathways, with BRAF modeling providing a mechanistic anchor for the targeted therapy. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

24 pages, 3834 KB  
Article
Temporal Dynamics of Cytokine, Leukocyte, and Whole Blood Transcriptome Profiles of Pigs Infected with African Swine Fever Virus
by Daniel W. Madden, Bianca Libanori Artiaga, Jessie D. Trujillo, Patricia Assato, Chester D. McDowell, Isaac Fitz, Taeyong Kwon, Konner Cool, Yonghai Li, Natasha N. Gaudreault, Igor Morozov and Juergen A. Richt
Pathogens 2025, 14(10), 992; https://doi.org/10.3390/pathogens14100992 - 1 Oct 2025
Abstract
African swine fever virus (ASFV) is an important transboundary animal pathogen with significant impacts on the global swine industry. Overwhelming proinflammatory responses are a major virulence mechanism for ASFV, but the dynamics of these changes during clinical disease are not completely understood. We [...] Read more.
African swine fever virus (ASFV) is an important transboundary animal pathogen with significant impacts on the global swine industry. Overwhelming proinflammatory responses are a major virulence mechanism for ASFV, but the dynamics of these changes during clinical disease are not completely understood. We constructed a detailed portrait of the innate immune responses during acute African swine fever (ASF) at the cellular, transcriptomic, and cytokine levels. Samples serially obtained from infected piglets show that progression of acute ASF is characterized by rapid increases in plasma type I interferons, TNF-α, IL-12p40, and IL-10, which coincide with the manifestation of clinical disease and viral DNAemia. Lymphocytes and natural killer (NK) cells progressively declined, with fluctuations in B cell, CD8+ T cell, and CD4+/CD8+ T cell populations. Blood monocytes and macrophages were highly variable throughout infection, with an abrupt spike in CD203+ mature macrophages immediately prior to death. Transcriptomic analysis of blood showed downregulation of cellular translation as early as 1 day post-challenge (DPC) and significant upregulation of antiviral immune processes at 5 DPC and 7 DPC, which overlapped with the onset of clinical disease. Together, these results present a detailed delineation of fatal ASF which involves an initial infection and damage of susceptible myeloid cells prior to symptomatic disease characterized by pro-inflammatory immune responses, lymphoid depletion, and clinical deterioration. Full article
(This article belongs to the Special Issue Emergence and Control of African Swine Fever: Second Edition)
Show Figures

Figure 1

19 pages, 7006 KB  
Article
Dynamic Reprogramming of Immune-Related Signaling During Progression to Enzalutamide Resistance in Prostate Cancer
by Pengfei Xu, Huan Qu, Joy C. Yang, Fan Wei, Junwei Zhao, Menghuan Tang, Leyi Wang, Christopher Nip, Henson Li, Allen C. Gao, Kit Lam, Marc Dall'Era, Yuanpei Li and Chengfei Liu
Cancers 2025, 17(19), 3187; https://doi.org/10.3390/cancers17193187 - 30 Sep 2025
Abstract
Background: Treatment with androgen receptor (AR) signaling inhibitors, such as enzalutamide, can induce neural lineage plasticity in prostate cancer, potentially progressing to t-NEPC. However, the molecular mechanisms underlying this enzalutamide-driven plasticity, particularly the contribution of immune signaling pathways, remain poorly understood. Methods: We [...] Read more.
Background: Treatment with androgen receptor (AR) signaling inhibitors, such as enzalutamide, can induce neural lineage plasticity in prostate cancer, potentially progressing to t-NEPC. However, the molecular mechanisms underlying this enzalutamide-driven plasticity, particularly the contribution of immune signaling pathways, remain poorly understood. Methods: We analyzed transcriptomic profiles of patient samples and prostate cancer cell lines to investigate changes in immune signaling pathways. Interferon gamma (IFNγ), interferon alpha (IFNα), and interleukin 6 (IL6)-Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) signaling were assessed in enzalutamide-sensitive and -resistant prostate cancer cells. Functional assays were conducted to examine cell responsiveness to cytokine stimulation and susceptibility to STAT1 inhibition using fludarabine. Results: Immune-related pathways, including IFNγ, IFNα, IL6-JAK-STAT3, and inflammatory responses, were significantly suppressed in NEPC patient samples compared to those with castration-resistant prostate cancer (CRPC). Enzalutamide-resistant and NEPC cells exhibited markedly impaired IFNγ and IL6 signaling. In contrast, early-stage enzalutamide treatment paradoxically enhanced IFNγ and IL6 responsiveness. Transcriptomic profiling revealed coordinated upregulation of E2F target genes and activation of IFNα/IFNγ and JAK/STAT signaling pathways during early treatment. Importantly, these early-stage cells remained highly sensitive to IFNγ and IL6 stimulation and showed increased susceptibility to STAT1 inhibition by fludarabine, a sensitivity that was lost in resistant cells. Conclusions: Early enzalutamide treatment enhances immune responsiveness, while the development of resistance is associated with suppressed immune signaling and increased lineage plasticity. These results suggest a therapeutic window where combining enzalutamide with STAT inhibitors may delay or prevent lineage plasticity and resistance. Full article
Show Figures

Figure 1

29 pages, 4385 KB  
Review
The Dual Role of Astrocytes in CNS Homeostasis and Dysfunction
by Aarti Tiwari, Satyabrata Rout, Prasanjit Deep, Chandan Sahu and Pradeep Kumar Samal
Neuroglia 2025, 6(4), 38; https://doi.org/10.3390/neuroglia6040038 - 29 Sep 2025
Abstract
Astrocytes are the most common type of glial cell in the central nervous system (CNS). They have many different functions that go beyond just supporting other cells. Astrocytes were once thought of as passive parts of the CNS. However, now they are known [...] Read more.
Astrocytes are the most common type of glial cell in the central nervous system (CNS). They have many different functions that go beyond just supporting other cells. Astrocytes were once thought of as passive parts of the CNS. However, now they are known to be active regulators of homeostasis and active participants in both neurodevelopmental and neurodegenerative processes. This article looks at the both sides of astrocytic function: how they safeguard synaptic integrity, ion and neurotransmitter balance, and blood-brain barrier (BBB) stability, as well as how astrocytes can become activated and participate in the immune response by releasing cytokines, upregulating interferons, and modulating the blood–brain barrier and inflammation disease condition. Astrocytes affect and influence neuronal function through the tripartite synapse, gliotransmission, and the glymphatic system. When someone is suffering from neurological disorders, reactive astrocytes become activated after being triggered by factors such as pro-inflammatory cytokines, chemokines, and inflammatory mediators, these reactive astrocytes, which have higher levels of glial fibrillary acidic protein (GFAP), can cause neuroinflammation, scar formation, and the loss of neurons. This review describes how astrocytes are involved in important CNS illnesses such as Alzheimer’s disease, Parkinson’s disease, multiple sclerosis, amyotrophic lateral sclerosis, and ischemia. It also emphasizes how these cells can change from neuroprotective to neurotoxic states depending on the situation. Researchers look at important biochemical pathways, such as those involving toll-like receptors, GLP-1 receptors, and TREM2, to see if they can change how astrocytes respond. Astrocyte-derived substances, including BDNF, GDNF, and IL-10, are also essential for protecting and repairing neurons. Astrocytes interact with other CNS cells, especially microglia and endothelial cells, thereby altering the neuroimmune environment. Learning about the molecular processes that control astrocytic plasticity opens up new ways to treat glial dysfunction. This review focuses on the importance of astrocytes in the normal and abnormal functioning of the CNS, which has a significant impact on the development of neurotherapeutics that focus on glia. Full article
Show Figures

Figure 1

10 pages, 2311 KB  
Article
Transcriptome Analysis Reveals Host Peripheral Blood Mononuclear Cells Response to Mpox Virus Infection
by Chaode Gu, Caiyun Wang, Chenlu Zhang, Jie Ni, Yun Xia and Hongwei Wang
Viruses 2025, 17(10), 1317; https://doi.org/10.3390/v17101317 - 28 Sep 2025
Abstract
Mpox virus (MPXV), a member of the Orthopoxvirus genus in the Poxviridae family, has long been endemic in Africa. The interaction between MPXV infection and peripheral immune responses is of great significance. However, the activation of signaling pathways and molecular changes in peripheral [...] Read more.
Mpox virus (MPXV), a member of the Orthopoxvirus genus in the Poxviridae family, has long been endemic in Africa. The interaction between MPXV infection and peripheral immune responses is of great significance. However, the activation of signaling pathways and molecular changes in peripheral blood mononuclear cells (PBMCs) following MPXV infection remain poorly understood. This study evaluated the transcriptomic profiles of rabbit PBMCs during the mpox acute and recovery phases. The results showed that MPXV infection significantly altered the transcriptomic profiles of PBMCs. At 6 days post-infection, pathways related to pathogenic infection and IL-1 response were enriched, while at 14 days post-infection, the T cell receptor signaling pathway was enriched. During the mpox acute phase, inflammatory cytokines in serum such as IL-1α, IL-1β, IL-8, and IL-21 were upregulated, while MMP-9 and NCAM-1 were downregulated. In rabbits and rhesus monkeys, key genes upregulated in common during the mpox acute period were associated with the interferon pathway (e.g., the ISG15, OAS, and IFIT families), while downregulated genes were related to B-cell activation and differentiation (e.g., the MS4A1 and FCRL families). Additionally, rabbits developed protective immunity against reinfection, with neutralizing antibodies effectively activated. These findings provide insights into the molecular characteristics of PBMCs changes in in vivo models of MPXV infection, and offer references for the diagnosis, vaccine development, and therapeutic research of mpox. Full article
Show Figures

Figure 1

22 pages, 3777 KB  
Article
Comparative Transcriptomics Reveals Novel and Differential Circular RNA Responses Underlying Interferon-Mediated Antiviral Regulation in Porcine Alveolar Macrophages
by Jiuyi Li, Oluwaseun Adeyemi, Laura C. Miller and Yongming Sang
Viruses 2025, 17(10), 1307; https://doi.org/10.3390/v17101307 - 27 Sep 2025
Abstract
Porcine Reproductive and Respiratory Syndrome (PRRS) causes significant economic losses in the swine industry. Circular RNAs (circRNAs), a class of stable non-coding RNAs, are increasingly recognized as regulators in immune responses and host–virus interactions. This study investigated the genome-wide circRNA responses in porcine [...] Read more.
Porcine Reproductive and Respiratory Syndrome (PRRS) causes significant economic losses in the swine industry. Circular RNAs (circRNAs), a class of stable non-coding RNAs, are increasingly recognized as regulators in immune responses and host–virus interactions. This study investigated the genome-wide circRNA responses in porcine alveolar macrophages (PAMs), key cell targets of PRRSV, following treatment with a modified live virus (MLV) vaccine or two interferon (IFN) subtypes (IFN-α1, IFN-ω5). Using RNA sequencing, we identified over 1000 differentially expressed circRNAs across treatment groups, revealing both conserved and distinct expression profiles. Gene Ontology and KEGG pathway analyses indicated that circRNA-associated genes are significantly enriched in immune-related processes and pathways, including cytokine signaling and antiviral defense. Notably, IFN-ω5 treatment induced a pronounced circRNA response, aligning with its potent antiviral activity. We further explored the regulatory potential of these circRNAs by predicting miRNA binding sites, revealing complex circRNA-miRNA interaction networks. Additionally, we assessed the coding potential of differentially expressed circRNAs by identifying open reading frames (ORFs), internal ribosome entry sites (IRESs), and N6-methyladenosine (m6A) modification sites, suggesting a subset may undergo non-canonical translation. These findings provide a comprehensive landscape of circRNA expression in PAMs under different antiviral conditions, highlighting their potential roles as immune regulators and novel players in interferon-mediated antiviral responses, particularly downstream of IFN-ω5. This work contributes to understanding the non-coding RNA landscape in the PRRSV-swine model and suggests circRNAs as potential targets for future antiviral strategies. Full article
(This article belongs to the Special Issue Host Cell-Virus Interaction, 4th Edition)
Show Figures

Figure 1

17 pages, 2330 KB  
Article
MyD88 Contributes to TLR3-Mediated NF-κB Activation and Cytokine Production in Macrophages
by Zhuodong Chai, Yuqi Zhou, Ling Yang, Yan Zhang, Sukria Hossain, Sahelosadat Hajimirzaei, Jiaqian Qi, Guoying Zhang, Yinan Wei and Zhenyu Li
Cells 2025, 14(19), 1507; https://doi.org/10.3390/cells14191507 - 27 Sep 2025
Abstract
Toll-like receptor 3 (TLR3) initiates antiviral and inflammatory responses exclusively through the adaptor protein TRIF (TIR-domain-containing adapter-inducing interferon-β). In contrast, MyD88 (myeloid differentiation primary response 88), a central adaptor for most other TLRs, is traditionally considered dispensable for TLR3 signaling. Here, we demonstrate [...] Read more.
Toll-like receptor 3 (TLR3) initiates antiviral and inflammatory responses exclusively through the adaptor protein TRIF (TIR-domain-containing adapter-inducing interferon-β). In contrast, MyD88 (myeloid differentiation primary response 88), a central adaptor for most other TLRs, is traditionally considered dispensable for TLR3 signaling. Here, we demonstrate that MyD88 directly contributes to TLR3-mediated NF-κB activation and cytokine production in macrophages. Bone marrow-derived macrophages (BMDMs) from MyD88 deficient mice exhibited significantly attenuated NF-κB activation in response to the TLR3 agonist polyinosinic–polycytidylic acid (poly(I:C)) compared to wild-type cells, as evidenced by the reduced phosphorylation of NF-κB p65 and IκBα, as well as IκBα degradation. Consistently, pro-inflammatory cytokine production, including IL-6, TNF-α, and IFN-β, was attenuated in MyD88-deficient BMDMs in vitro following stimulation by poly(I:C) or poly(A:U), another TLR3 agonist. Blood concentrations of IL-6, TNF-α, and IFN-β were significantly reduced in both TRIF-deficient mice and MyD88-deficient mice challenged by the i.p. injection of poly(I:C). Mechanistic analyses revealed that MyD88 physically associates with activated TLR3 upon poly(I:C) stimulation, and that TLR3 engagement triggered MyD88 oligomerization, which was absent in TLR3 or TRIF deficient macrophages. Our findings highlight a previously unrecognized dual-adaptor mechanism for TLR3, wherein MyD88 recruitment amplifies NF-κB signaling dynamics by bridging TLR3 to the canonical NF-κB activation cascade and robust cytokine induction. This study expands the paradigm of TLR3 signaling by establishing MyD88 as a direct contributor to TLR3-driven innate immune responses, offering new insight into cross-talk between MyD88-dependent and -independent pathways. Full article
Show Figures

Figure 1

52 pages, 1456 KB  
Review
The Gut Microbiome in Enteric Viral Infections: Underlying Mechanisms and Therapeutic Approaches
by Alejandro Borrego-Ruiz and Juan J. Borrego
Microorganisms 2025, 13(10), 2247; https://doi.org/10.3390/microorganisms13102247 - 25 Sep 2025
Abstract
Despite growing recognition of the role of the gut microbiome in host health and in modulating pathogen activity, the dynamic and reciprocal relationship between enteric viruses and the gut microbial ecosystem remains insufficiently defined and requires further exploration. This comprehensive review examines the [...] Read more.
Despite growing recognition of the role of the gut microbiome in host health and in modulating pathogen activity, the dynamic and reciprocal relationship between enteric viruses and the gut microbial ecosystem remains insufficiently defined and requires further exploration. This comprehensive review examines the bidirectional interplay between the gut microbiome and enteric viral infections by addressing (i) viruses associated with gastrointestinal alterations, (ii) how enteric viral infections alter the composition and function of the gut microbiome, (iii) how the gut microbiome modulates viral infectivity and host susceptibility, and (iv) current microbial-based approaches for preventing or treating enteric viral infections. Gastrointestinal viral infections induce gut microbiome dysbiosis, marked by reductions in beneficial bacteria and increases in potentially pathogenic populations. Specific gut microorganisms can modulate host susceptibility, with certain bacterial genera increasing or decreasing infection risk and disease severity. Pattern recognition receptors in the intestinal epithelium detect microbial signals and trigger antimicrobial peptides, mucus, and interferon responses to control viral replication while maintaining tolerance to commensal bacteria. The gut microbiome can indirectly facilitate viral infections by creating a tolerogenic environment, suppressing antiviral antibody responses, and modulating interferon signaling, or directly enhance viral replication by stabilizing virions, promoting host cell attachment, and facilitating coinfection and viral recombination. In turn, commensal gut bacteria can inhibit viral entry, enhance host antiviral responses, and strengthen mucosal barrier function, contributing to protection against gastrointestinal viral infections. Probiotics and fecal microbiota transplantation constitute potential microbial-based therapeutics that support antiviral defenses, preserve epithelial integrity, and restore microbial balance. In conclusion, the role of the gut microbiome in modulating enteric viral infections represents a promising area of future investigation. Therefore, integrating microbiome insights with virology and immunology could enable predictive and personalized strategies for prevention and treatment. Full article
(This article belongs to the Special Issue Microbiota and Gastrointestinal Diseases)
Show Figures

Figure 1

11 pages, 2186 KB  
Article
MyD88 Plays an Important Role in UVB-Induced Suppression of Dendritic Cell Activity, T Cell Function, and Cutaneous Immune Response
by Mohammad Asif Sherwani, Carlos Alberto Mier Aguilar, Charlotte McRae, Gelare Ghajar-Rahimi, Aisha Anwaar, Ahmed Omar Jasser, Ariq Chandra, Hui Xu and Nabiha Yusuf
Int. J. Mol. Sci. 2025, 26(19), 9361; https://doi.org/10.3390/ijms26199361 - 25 Sep 2025
Abstract
Ultraviolet B (UVB) radiation triggers DNA damage and immune suppression, establishing conditions favorable for skin carcinogenesis. Previous studies have shown that a downstream adaptor for Toll-like receptors (TLRs), myeloid differentiation primary response 88 (MyD88), plays a role in UVB-induced DNA damage and immunosuppression. [...] Read more.
Ultraviolet B (UVB) radiation triggers DNA damage and immune suppression, establishing conditions favorable for skin carcinogenesis. Previous studies have shown that a downstream adaptor for Toll-like receptors (TLRs), myeloid differentiation primary response 88 (MyD88), plays a role in UVB-induced DNA damage and immunosuppression. However, specific mechanisms for the effects on dendritic cells and T cells remain poorly understood. The objective of this study is to determine the role of MyD88 and TIR-domain-containing adaptor inducing interferon-β (TRIF), another key TLR downstream adaptor, in UVB-induced suppression of dendritic cell activity and T cell function. MyD88−/−, Trif−/−, and wild-type (WT) mice were evaluated for UVB-induced effects on dendritic cell, T cells, and contact hypersensitivity responses in skin. MyD88−/− mice exhibited significant resistance to UVB-induced immune suppression, compared to Trif−/− mice and wild-type controls. The MyD88 deficiency significantly reduced UVB-induced Treg cells that were CD4+CD25+Foxp3+ and produced interleukin (IL)-10. Moreover, it significantly inhibited the UVB-induced suppression of IL-12/IL-23 producing CD11c+ dendritic cells. Further experiments confirmed that MyD88 conditional knockout (MyD88fl/flXCD11c.Cre) mice were protected against UVB-induced immune suppression. Dendritic cells from MyD88 genomic or conditional knockout mice were resistant to UVB-induced reduction of major histocompatibility complex (MHC) class II antigens. These findings show that MyD88 plays a key role in UVB-induced immune suppression. The deficiency in the MyD88 gene inhibits UVB-induced suppression of CD11c+ dendritic cell (DC) activity and reduces UVB-induced development of Treg cells. Our studies demonstrate a new mechanism for MyD88-mediated regulation of UVB-induced immune suppression. Full article
Show Figures

Figure 1

17 pages, 4834 KB  
Article
Activation of IRE1 Endonuclease Activity Regulates Zika Virus Replication and Antiviral Response During Infection in Human Microglia
by Tomás Hernández-Díaz, Aarón Oyarzún-Arrau, Aracelly Gaete-Argel, Delia López-Palma, Javier López-Schettini, Dominique Fernández, Fernando Valiente-Echeverría, Fabiola Osorio and Ricardo Soto-Rifo
Viruses 2025, 17(10), 1291; https://doi.org/10.3390/v17101291 - 24 Sep 2025
Viewed by 111
Abstract
Zika virus (ZIKV) can infect and replicate in the endoplasmic reticulum (ER) of different human cell types, including neural progenitor cells, radial glial cells, astrocytes, and microglia in the brain. ZIKV infection of microglia is expected to trigger both ER stress and the [...] Read more.
Zika virus (ZIKV) can infect and replicate in the endoplasmic reticulum (ER) of different human cell types, including neural progenitor cells, radial glial cells, astrocytes, and microglia in the brain. ZIKV infection of microglia is expected to trigger both ER stress and the induction of an antiviral response through production of type-I interferons and pro-inflammatory cytokines, contributing to neuroinflammation during infection. Despite their critical role in ZIKV pathogenesis, the interplay between ER stress and the antiviral response during infection has not been fully characterized in human microglia. In this work, we show that infection of a human microglia cell line with ZIKV triggers the induction of an antiviral response and the activation of the endonuclease activity of the unfolded protein response sensor IRE1. Interestingly, we observed that both IRE1 and XBP1 were sequestered to the viral replication sites during infection. Moreover, pharmacological inhibition or hyperactivation of the endonuclease activity of IRE1 resulted in reduced viral titers. As such, while inhibition of IRE1 resulted in an increased type-I interferon response, hyperactivation led to a decrease in ZIKV RNA levels and the appearance of ER-derived cytoplasmic structures containing NS3, IRE1, and XBP1. Together, our data indicate that regulation of the endonuclease activity of IRE1 is critical for both ZIKV replication and immune activation, highlighting the potential of the ER stress sensor as a target for the development of antivirals to treat ZIKV infections. Full article
(This article belongs to the Special Issue Virus-Host Protein Interactions)
Show Figures

Figure 1

26 pages, 2605 KB  
Article
Salivary Biomarker Analysis to Distinguish Between Health and Periodontitis Status: A Preliminary Study
by Carlo Bertoldi, Milena Nasi, Roberta Salvatori, Marcello Pinti, Silvia Montagna, Maurizio Tonetti, Luigi Generali, Elisa Bellei, Davide Zaffe, Valentina Selleri and Stefania Bergamini
Dent. J. 2025, 13(9), 436; https://doi.org/10.3390/dj13090436 - 22 Sep 2025
Viewed by 243
Abstract
Objective: This study aims to explore the feasibility of a non-invasive and simple method for discriminating between health and periodontitis (PRD), facilitating early and objective diagnosis of PRD before detectable periodontal attachment loss and monitoring treatment outcomes. Methods: Salivary samples were collected from [...] Read more.
Objective: This study aims to explore the feasibility of a non-invasive and simple method for discriminating between health and periodontitis (PRD), facilitating early and objective diagnosis of PRD before detectable periodontal attachment loss and monitoring treatment outcomes. Methods: Salivary samples were collected from 16 PRD-free patients (G1) and 10 patients with PRD (G2). The analysis included salivary matrix metalloproteinase-8 (MMP-8), major anti-inflammatory interleukins (IL-4 and IL-10), pro-inflammatory cytokines (IL-1β, IL-8, and interferon α [IFN-α]), and the cytokine IL-6. Clinical and salivary assessments were performed at baseline (TP0) for both groups and after periodontal treatment for G2 (TP1). Results: PRD indices were significantly higher in G2-TP0, lower in G1, and intermediate in G2-TP1. Except for IL-6, the biomarkers were significantly correlated with nearly all PRD clinical indices. Logistic regression and receiver operating characteristic (ROC) curve analyses showed statistical significance for MMP-8, IL-1β, IL-4, IL-8, and IL-10 when comparing G1 and G2 at TP0. MMP-8 was also significant when comparing G2-TP0 and G2-TP1, while IL-1β and IL-10 showed borderline significance. IL-8 was significant when comparing G1 and G2-TP1. Conclusions: The molecular network demonstrated great potential for early diagnosis and monitoring of therapy response, providing a promising basis for future research. Among the biomarkers, MMP-8, IL-1β, IL-4, IL-8, and IL-10 showed the strongest statistical correlations with the clinical indices. The inflammation-related biomolecules behaved differently among untreated PRD (G2-TP0), treated (G2-TP1), and healthy individuals (G1). Healthy individuals and those with treated PRD may regulate inflammation significantly differently from those with untreated PRD. Full article
Show Figures

Figure 1

22 pages, 4275 KB  
Article
Lumpy Skin Disease Virus ORF137 Protein Inhibits Type I Interferon Production by Interacting with and Decreasing the Phosphorylation of IRF3
by Qunhua Ke, Kaishen Yao, Min Qu, Zhengji Liang, Miaomiao Li, Xiangwei Wang, Xiangping Yin and Yuefeng Sun
Cells 2025, 14(18), 1475; https://doi.org/10.3390/cells14181475 - 22 Sep 2025
Viewed by 276
Abstract
Lumpy skin disease (LSD) is an invasive infectious disease caused by the lumpy skin disease virus (LSDV), which is detrimental to the production of cattle. LSDV encodes about 156 proteins, most of whose functions are still unknown. In this study, we found that [...] Read more.
Lumpy skin disease (LSD) is an invasive infectious disease caused by the lumpy skin disease virus (LSDV), which is detrimental to the production of cattle. LSDV encodes about 156 proteins, most of whose functions are still unknown. In this study, we found that the ORF137 protein was identified as one of the strongest inhibitors of IFN-β and ISG expression, determining LSDV ORF137 as a negative regulator of interferon (IFN) β signaling. Further evidence suggests that ORF137 interacts with the signal transduction factor IRF3 and inhibits the activation of IFN-β signaling by reducing Phospho-IRF3 (p-IRF3). Further investigation indicated that overexpression of ORF137 in BMEC could significantly inhibit the transcription of IFN-β and ISGs, thereby promoting the replication of LSDV. More importantly, through homologous recombination, we deleted the ORF137 gene from the LSDV/FJ/CHA/2021 strain and constructed the recombinant strain LSDV-ΔORF137-EGFP. Compared with the parental strain, LSDV-ΔORF137-EGFP showed a weakened effect on inhibiting the transcription of IFN-β and ISGs and a reduced replication level in infected MDBK cells. In summary, ORF137 facilitates LSDV replication by targeting IRF3 to inhibit IFN-β signaling. Our findings reveal a new mechanism by which LSDV suppresses the host antiviral response, which may facilitate the development of attenuated live vaccines for LSDV. Full article
Show Figures

Figure 1

40 pages, 3769 KB  
Article
Deciphering European Sea Bass (Dicentrarchus labrax) Resistance to Nervous Necrosis Virus by Transcriptome Analysis from Early Infection Towards Establishment of Virus Carrier State
by Dimitra K. Toubanaki, Odysseas-Panagiotis Tzortzatos, Antonia Efstathiou, Vasileios Bakopoulos and Evdokia Karagouni
Int. J. Mol. Sci. 2025, 26(18), 9220; https://doi.org/10.3390/ijms26189220 - 21 Sep 2025
Viewed by 255
Abstract
Viral nervous necrosis, caused by the nervous necrosis virus (NNV), is an important threat to aquaculture, causing great economic losses and a high environmental burden. European sea bass (Dicentrarchus labrax) is highly affected by NNV, and selective breeding programs for disease [...] Read more.
Viral nervous necrosis, caused by the nervous necrosis virus (NNV), is an important threat to aquaculture, causing great economic losses and a high environmental burden. European sea bass (Dicentrarchus labrax) is highly affected by NNV, and selective breeding programs for disease resistance have been established in order to achieve a sustainable aquaculture and minimize the need for vaccines, drugs and antibiotics. Resistant and susceptible European sea bass were experimentally challenged with NNV and their head kidney transcriptomes were analyzed at three time points, i.e., 3 hpi, 2 dpi and 14 dpi. Numerous differentially expressed genes (DEGs) were identified in the head kidneys of resistant and susceptible infected vs. non-infected sea bass. Gene ontology enrichment, pathway, and protein–protein interaction analyses revealed that the NNV-resistant fish control their response to viral infection more efficiently, utilizing different mechanisms compared to the susceptible fish. Resistant fish displayed higher levels of interferon-related elements, cytokines, antigen presentation, T-cell activity, apoptosis, and programmed cell death combined with a controlled inflammatory response and more active proteasome and lysosome functions. The susceptible fish appeared to have high immune responses at the early infection stages, accompanied by high expressions of inflammatory, complement and coagulation pathways. Insulin metabolism was better regulated in the resistant fish and the control of lipid metabolism was less effective in the susceptible family. The cytoskeleton- and cell adhesion-related pathways were mostly down-regulated in the susceptible fish, and the intracellular transport and motor proteins were utilized more efficiently by the resistant fish. The present study represents a thorough transcriptomic analysis of NNV infection effects on a resistant and a susceptible European sea bass head kidney. The obtained results provide valuable information on the mechanisms that offers pathogen resistance to a host, with many aspects that can be exploited to develop more efficient approaches to fighting viral diseases in aquaculture. Full article
Show Figures

Figure 1

29 pages, 7171 KB  
Article
Antibody-Dependent Enhancement of Porcine Reproductive and Respiratory Syndrome Virus Infection Antagonizes the Secretion of Type I Interferons in Porcine Alveolar Macrophages by Interfering with the Retinoic Acid-Inducible Gene I/Melanoma Differentiation-Associated Gene 5 Pathway via Fc Gamma Receptor I
by Liujun Zhang, Aiyang Wang, Weizhen Chen, Xing Feng, Bo Wang, Shaojun He and Hongjie Fan
Viruses 2025, 17(9), 1277; https://doi.org/10.3390/v17091277 - 20 Sep 2025
Viewed by 213
Abstract
Type I interferons (IFNs), mainly IFN-α and IFN-β, play an essential role in defending against viral invasion by inducing the host’s innate antiviral response. Porcine reproductive and respiratory syndrome virus (PRRSV) is known to impair the IFN responses of infected hosts through the [...] Read more.
Type I interferons (IFNs), mainly IFN-α and IFN-β, play an essential role in defending against viral invasion by inducing the host’s innate antiviral response. Porcine reproductive and respiratory syndrome virus (PRRSV) is known to impair the IFN responses of infected hosts through the antibody-dependent enhancement (ADE) infection pathway, but the precise mechanisms employed are poorly understood. In this study, we showed that PRRSV alone induced a strong secretion of IFN-α and IFN-β in infected porcine alveolar macrophages (PAMs) by activating the retinoic acid-inducible gene I (RIG-I)/melanoma differentiation-associated gene 5 (MDA5) signaling pathway. By contrast, ADE infection of PRRSV significantly down-regulated the production levels of IFN-α and IFN-β in PAMs by negatively regulating the RIG-I/MDA5 signaling pathway and considerably enhancing the replication level of PRRSV in PAMs. Next, small interfering RNA (siRNA) experiments revealed that Fc gamma receptor I (FcγRI) was responsible for the ADE infection of PRRSV in PAMs. In addition, we observed that FcγRI mediated the potent inhibition of IFN-α and IFN-β production through blocking the activation of the RIG-I/MDA5 signaling pathway in PAMs. Further, we found that FcγRI effectively inhibited PRRSV-induced synthesis of IFN-α and IFN-β by negatively regulating PRRSV-induced activation of the RIG-I/MDA5 signaling pathway in PAMs and significantly increased the viral production of PRRSV in PAMs. In conclusion, these results suggest that ADE infection of PRRSV may antagonize the secretion of type I IFNs (IFN-α/β) by interfering with the RIG-I/MDA5 pathway via FcγRI in PAMs, thereby facilitating the proliferation level of PRRSV in PAMs. Full article
(This article belongs to the Section Animal Viruses)
Show Figures

Figure 1

19 pages, 4885 KB  
Article
Induction of Sustained Immunity Following Vaccination with Live Attenuated Trypanosoma cruzi Parasites Combined with Saponin-Based Adjuvants
by Brenda A. Zabala, María Elisa Vázquez, Daniela E. Barraza, Andrea C. Mesías, Federico Ramos, Alejandro Uncos, Iván S. Marcipar, Leonardo Acuña and Cecilia Pérez Brandán
Biology 2025, 14(9), 1298; https://doi.org/10.3390/biology14091298 - 20 Sep 2025
Viewed by 266
Abstract
Chagas disease, caused by Trypanosoma cruzi, remains a major health concern in Latin America, particularly affecting low-income and rural communities. Among the many vaccine strategies explored, live attenuated parasites have shown the strongest ability to trigger protective immune responses. In this study, [...] Read more.
Chagas disease, caused by Trypanosoma cruzi, remains a major health concern in Latin America, particularly affecting low-income and rural communities. Among the many vaccine strategies explored, live attenuated parasites have shown the strongest ability to trigger protective immune responses. In this study, we investigated whether adding saponin-based adjuvants—Immunostimulant Particle Adjuvant (ISPA) and Quil-A—could improve the effectiveness and safety of a live parasite attenuated T. cruzi vaccine. Mice were vaccinated with a T. cruzi attenuated strain (TCC) alone or in combination with each adjuvant, and immunoglobulin G (IgG) subtypes in the serum of vaccinated mice, and interferon gamma (IFN-γ) and interleukin-10 (IL-10) in the supernatants of stimulated cells were measured at two weeks and twelve months post-vaccination. While protection levels were similar across all groups, the adjuvants assist in modulating the immune response over time: ISPA and Quil-A initially shifted antibody production toward IgG1 but later favored a balanced TH1/TH2 profile. ISPA also promoted long-term regulation through increased IL-10. Both adjuvants reduced tissue inflammation and enhanced clearance of vaccine-derived parasites. These findings suggest that while adjuvants may not boost protection directly, they significantly improve vaccine safety and immune quality, reinforcing their value in developing better vaccines for Chagas disease. Full article
Show Figures

Graphical abstract

Back to TopTop