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

Journals

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

Search Results (614)

Search Parameters:
Keywords = major histocompatibility complex I

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 2636 KB  
Article
Pro-Inflammatory Macrophage Phenotype Skewing Induced by Tumor Treating Fields (TTFields)
by Tal Kan, Yiftah Barsheshet, Tharwat Haj Khalil, Boris Brant, Tali Voloshin, Kerem Ben-Meir, Simona Zisman Rosen, Moshe Giladi, Uri Weinberg and Yoram Palti
Int. J. Mol. Sci. 2025, 26(24), 12086; https://doi.org/10.3390/ijms262412086 - 16 Dec 2025
Abstract
Tumor-associated macrophages (TAMs) are abundant in the tumor microenvironment (TME) and often adopt an M2-like immunosuppressive phenotype that promotes tumor growth. Reprogramming TAMs toward an M1-like pro-inflammatory state is an attractive therapeutic strategy. Tumor Treating Fields (TTFields), an FDA-approved, electric-field–based therapy, has recently [...] Read more.
Tumor-associated macrophages (TAMs) are abundant in the tumor microenvironment (TME) and often adopt an M2-like immunosuppressive phenotype that promotes tumor growth. Reprogramming TAMs toward an M1-like pro-inflammatory state is an attractive therapeutic strategy. Tumor Treating Fields (TTFields), an FDA-approved, electric-field–based therapy, has recently been suggested to modulate immune responses in addition to its established anti-mitotic activity. Here, we investigated the direct effects of TTFields on macrophage activation and function. Murine bone marrow–derived macrophages (BMDMs) were polarized toward a pro-inflammatory M1-like phenotype or an anti-inflammatory M2-like phenotype and exposed to TTFields. TTFields rapidly activated guanine nucleotide exchange factor-H1 (GEF-H1), and downstream nuclear factor kappa B (NF-κB) and activator protein-1 (AP-1, via c-Jun N-terminal kinase [JNK]) signaling. Functionally, TTFields reprogrammed M2-like macrophages by increasing major histocompatibility complex class II (MHC-II) and cluster of differentiation 80 (CD80); reducing arginase-1 (Arg1); and elevating secretion of chemokine (C-X-C motif) ligand 1 (CXCL1), interleukin-6 (IL-6), IL-1β, and IL-12 subunit p70 (IL-12p70). In interferon gamma (IFN-γ)-primed macrophages, TTFields provided a secondary signal, driving myeloid differentiation primary response 88 (MyD88)-dependent expression of inducible nitric oxide synthase (iNOS). In vivo, TTFields reduced tumor burden in an orthotopic murine lung cancer model and increased iNOS expression in both M1-like and a subset of M2-like TAMs. These findings demonstrate that TTFields directly reprogram macrophages toward a pro-inflammatory phenotype, suggesting a novel immunomodulatory mechanism that may enhance anti-tumor immunity in the TME. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

23 pages, 1268 KB  
Review
CD74 and Proteases: Impact of Location on Immune and Cellular Functions
by Iztok Dolenc
Cells 2025, 14(24), 1960; https://doi.org/10.3390/cells14241960 - 10 Dec 2025
Viewed by 257
Abstract
Proteases represent a diverse family of enzymes that catalyze the hydrolysis of peptide bonds, modulating numerous biological processes. Among their substrates, CD74—also known as the invariant chain—has received increased research attention due to its multifunctional roles in both innate and adaptive immunity. This [...] Read more.
Proteases represent a diverse family of enzymes that catalyze the hydrolysis of peptide bonds, modulating numerous biological processes. Among their substrates, CD74—also known as the invariant chain—has received increased research attention due to its multifunctional roles in both innate and adaptive immunity. This review provides an overview of current knowledge on protease-mediated interactions with CD74. The protein was originally identified as a chaperone for major histocompatibility complex class II (MHC-II) molecules. Proteolytic cleavage of CD74, most notably by cathepsin S, is essential for the release of MHC-II and the initiation of antigen presentation. However, CD74 has since emerged as a central regulator of processes extending well beyond antigen presentation. More recent findings reveal that CD74, acting as a receptor of macrophage migration inhibitory factor, also participates in signaling pathways in non-immune cells, independent of its classical chaperone function. Proteolytic processing of CD74 can trigger signaling cascades that modulate gene expression, underscoring its multifunctionality. Dysregulation of CD74 cleavage and its interaction with proteases has been linked to diverse pathological conditions, including cancer and autoimmune diseases, where aberrant protease activity disrupts CD74 function and promotes disease progression. Full article
Show Figures

Figure 1

22 pages, 2395 KB  
Article
Characterization of Stimulated γδ T Cells: Phenotypic Analysis and Implications for Allogeneic Cellular Immunotherapy
by Anna Bold, Heike Gross, Marco Bardenbacher, Elisabeth Holzmann, Stefan Knop and Martin Wilhelm
Cells 2025, 14(23), 1917; https://doi.org/10.3390/cells14231917 - 2 Dec 2025
Viewed by 377
Abstract
Due to their anti-tumor activity and non-major histocompatibility complex (MHC) binding T cell receptor, γδ T cells are suitable candidates for allogeneic cellular immunotherapy in cancer. Recently, we developed a new protocol called Ko-Op for stimulation of γδ T cells (specifically Vy9Vδ2 T [...] Read more.
Due to their anti-tumor activity and non-major histocompatibility complex (MHC) binding T cell receptor, γδ T cells are suitable candidates for allogeneic cellular immunotherapy in cancer. Recently, we developed a new protocol called Ko-Op for stimulation of γδ T cells (specifically Vy9Vδ2 T cells) that generates a cell product consisting mainly of γδ T cells with preserved anti-tumor activity targeted for clinical-grade application. In this study, we investigated the phenotype of stimulated γδ T cells and correlated this with results of functional assays to obtain a deeper understanding of the characteristics of stimulated γδ T cells. Additionally, an intensive analysis of surface molecules of unstimulated and stimulated γδ T cells is presented. Since heterogeneous results regarding the response to therapy with γδ T cells observed in earlier clinical trials could be a consequence of various extents of γδ T cell adhesion and migration ability, we addressed surface molecules associated with cellular activity and adhesion and migration functions as well. By investigating correlations between the phenotype of unstimulated γδ T cells and cellular cytotoxicity, as well as the degranulation ability of stimulated γδ T cells, we could draw conclusions about optimal donors for further allogeneic cellular therapies. Finally, we demonstrated that the phenotype varies over the time of culture and is clearly modifiable by changing the stimulation protocol. Full article
(This article belongs to the Special Issue Unconventional T Cells in Health and Disease)
Show Figures

Figure 1

17 pages, 8700 KB  
Article
Designing a Novel Multi-Epitope Trivalent Vaccine Against NDV, AIV and FAdV-4 Based on Immunoinformatics Approaches
by Jiashuang Ji, Xiaofeng Dong, Xiangyi Liu, Mengchun Ding, Yating Lin, Yunhang Zhang, Wuchao Zhang, Baishi Lei, Wanzhe Yuan and Kuan Zhao
Microorganisms 2025, 13(12), 2744; https://doi.org/10.3390/microorganisms13122744 - 2 Dec 2025
Viewed by 325
Abstract
The diseases caused by genotype VII Newcastle disease virus (NDV), H9N2 avian influenza virus (AIV), and fowl adenovirus serotype 4 (FAdV-4) continue to threaten the global poultry industry. However, no broad-spectrum vaccines provide simultaneous protection against these three pathogens. This study employed bioinformatics [...] Read more.
The diseases caused by genotype VII Newcastle disease virus (NDV), H9N2 avian influenza virus (AIV), and fowl adenovirus serotype 4 (FAdV-4) continue to threaten the global poultry industry. However, no broad-spectrum vaccines provide simultaneous protection against these three pathogens. This study employed bioinformatics and immunoinformatics approaches to design a multi-epitope vaccine, named NFAF, which consists of B-cell, cytotoxic T lymphocyte (CTL) epitopes, and helper T lymphocyte (HTL) epitopes derived from hemagglutinin-neuraminidase (HN) and fusion (F) proteins of genotype VII NDV, hemagglutinin (HA) protein of H9N2, and Fiber2 protein of FAdV-4. The vaccine candidate was predicted to have non-allergenic properties, non-toxicity, high antigenicity, and favorable solubility. Each of its constituent antigenic epitopes has a high degree of conservation. Molecular docking demonstrated stable binding between NFAF and chicken Toll-like receptor (TLRs) and major histocompatibility complex (MHC) molecules. NFAF was expressed in soluble form in Escherichia coli and purified. Polyclonal antibodies against all three target viruses showed specific binding to NFAF. In vitro experiments revealed that NFAF effectively stimulated chicken peripheral blood mononuclear cells (PBMCs) and induced Th1, Th2, and pro-inflammatory cytokine production, confirming its immunogenicity, and increased the mRNA expression of the key signaling molecules MyD88 and NF-κB. These results suggested that NFAF could therefore be an efficacious multi-epitope vaccine against genotype VII NDV, H9N2, and FAdV-4 infections. Full article
(This article belongs to the Special Issue The Host Response to Animal Virus Infection)
Show Figures

Figure 1

25 pages, 5688 KB  
Article
Lacticaseibacillus rhamnosus MS27 Potentially Prevents Ulcerative Colitis Through Modulation of Gut Microbiota
by Jie Zhang, Jiakun Shen, Linbao Ji, Peng Tan, Chunchen Liu, Xiujun Zhang and Xi Ma
Int. J. Mol. Sci. 2025, 26(23), 11397; https://doi.org/10.3390/ijms262311397 - 25 Nov 2025
Viewed by 392
Abstract
(1) This study explored Lacticaseibacillus rhamnosus MS27, a newly isolated strain, as a potential probiotic candidate for alleviating the onset and severity of ulcerative colitis (UC). (2) L. rhamnosus MS27 was isolated and subjected to biochemical identification, antibiotic sensitivity testing, and antibacterial activity [...] Read more.
(1) This study explored Lacticaseibacillus rhamnosus MS27, a newly isolated strain, as a potential probiotic candidate for alleviating the onset and severity of ulcerative colitis (UC). (2) L. rhamnosus MS27 was isolated and subjected to biochemical identification, antibiotic sensitivity testing, and antibacterial activity assessment. Dextran sulfate sodium (DSS) colitis model mice were used to evaluate its alleviating effects. In this study, 16S rRNA microbiome and eukaryotes reference transcriptome analyses were conducted to investigate its impact on intestinal microbial ecology and potential molecular mechanisms. (3) L. rhamnosus MS27 exhibits high acid tolerance at pH 3.23 and maintains a high viable bacterial count for 24 h. It can utilize sucrose, lactose, maltose, inulin, esculin, salicin, and mannitol but not raffinose, and it is sensitive to carbenicillin, erythromycin, tetracycline, chloramphenicol, clindamycin, and penicillin. It effectively increases the abundance of beneficial microbes, particularly Akkermansia, Muribaculaceae, and Limosilactobacillus reuteri (p < 0.05), while significantly reducing microorganisms linked to human pathogens causing diarrhea and gastroenteritis (p < 0.05). Transcriptomic analysis demonstrated that the expression levels of Igkv16-104 and C1qtnf3 were significantly downregulated in the presence of L. rhamnosus MS27 treatment compared to DSS treatment alone (p < 0.05). Further analysis revealed significant differences in genes related to immune functions, antigen presentation, and immune cell markers, indicating potential protein–protein interaction networks, particularly among genes of the major histocompatibility complex (MHC). (4) L. rhamnosus MS27, as a novel strain, demonstrates a significant capacity to alleviate inflammatory phenotypes. L. rhamnosus MS27 exhibits distinctive metabolic characteristics in lactic acid utilization, acetic acid and oleic acid production. Furthermore, it contributes to systemic homeostasis regulation by modulating Turicibacter to link intestinal microbiota composition with host immune function. Full article
Show Figures

Graphical abstract

29 pages, 5088 KB  
Hypothesis
Molecular Mechanism for the Selective Presentation of Antigenic Peptides by Major Histocompatibility Complex Class I and Class II Molecules: A Hypothesis
by Bao Ting Zhu
Curr. Issues Mol. Biol. 2025, 47(11), 945; https://doi.org/10.3390/cimb47110945 - 13 Nov 2025
Viewed by 705
Abstract
The major histocompatibility complex (MHC) class I and class II molecules (abbreviated as MHC-I and MHC-II, respectively) are specialized in antigen presentation. Unlike the T cell receptors (TCRs), which have great variability, the MHC-I and MHC-II molecules essentially have no variability at all. [...] Read more.
The major histocompatibility complex (MHC) class I and class II molecules (abbreviated as MHC-I and MHC-II, respectively) are specialized in antigen presentation. Unlike the T cell receptors (TCRs), which have great variability, the MHC-I and MHC-II molecules essentially have no variability at all. It is apparent that the MHC-I and MHC-II molecules per se do not have the built-in ability to distinguish the huge populations of self-peptides from antigenic non-self-peptides. At present, the precise mechanism underlying the selective presentation of antigenic peptides by both MHC-I and MHC-II molecules is unclear. For an MHC-II molecule to gain the ability to selectively present antigenic (mostly foreign) peptides, it is hypothesized herein that all naïve CD4+ T cells in the body will release extracellular vesicles (EVs), which are specially designed for antigen-presenting cells (APCs); these EVs contain mRNAs that will be delivered to APCs and translated into an intracellular version of the TCR proteins (iTCRII), which will help select antigenic peptides for presentation by the MHC-II molecules. Similarly, it is hypothesized that the fully activated CD4+ T cells will also release EVs, and these EVs contain different mRNAs for another intracellular version of the TCR proteins (iTCRI), which will help pathogen-infected somatic cells to select the antigenic peptides (mostly from invading pathogens) for presentation by the MHC-I molecules. Understandably, while the iTCRII proteins will work closely with the MHC-II molecules in the exogenous endocytic pathway, the iTCRI proteins will work closely with the MHC-I molecules in the endogenous pathway. In this paper, a few other related hypotheses are also proposed, which jointly offer a plausible mechanistic explanation for the selective presentation of antigenic peptides by both MHC-I and MHC-II molecules. While the proposed hypotheses are partly supported by some experimental observations, it is hoped that these hypotheses will promote discussion and experimental testing of the mechanisms underlying the complex process of selective antigen presentation. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Figure 1

21 pages, 1541 KB  
Article
Comparative Analysis of Genetic Risk for Viral-Induced Axonal Loss in Genetically Diverse Mice
by Tae Wook Kang, Aracely Perez-Gomez, Koedi Lawley, Colin R. Young, C. Jane Welsh and Candice L. Brinkmeyer-Langford
Int. J. Mol. Sci. 2025, 26(21), 10727; https://doi.org/10.3390/ijms262110727 - 4 Nov 2025
Cited by 1 | Viewed by 563
Abstract
Among mouse models of neurological disease, Theiler’s murine encephalomyelitis virus (TMEV) provides a unique platform by using a naturally occurring viral trigger, paralleling the role of infections like Epstein–Barr virus in multiple sclerosis (MS). Just as not all individuals with predisposing viral infections [...] Read more.
Among mouse models of neurological disease, Theiler’s murine encephalomyelitis virus (TMEV) provides a unique platform by using a naturally occurring viral trigger, paralleling the role of infections like Epstein–Barr virus in multiple sclerosis (MS). Just as not all individuals with predisposing viral infections develop the same neurological disease, not all mouse strains develop the same diseases following TMEV infection, so susceptibility is dictated by genetic background. For example, certain sets of alleles, called haplotypes, of the major histocompatibility complex (MHC) region have been associated with susceptibility to TMEV-induced demyelination (TVID) and MS. However, our previous work revealed that these MHC susceptibility haplotypes are not the sole contributors to TMEV-induced diseases in all mice. We infected mice from the genetically diverse Collaborative Cross (CC), a resource designed to reflect human population-level genetic variation. All 15 CC strains tested exhibited some form of neurological phenotype or CNS lesion following TMEV infection. However, chronic radiculoneuropathy characterized by axonal degeneration with myelin loss was observed in the CNS of only two strains, CC002 and CC023, which had markedly different immune responses and clinical profiles throughout the course of infection. Moreover, the pathology seen in CC002 and CC023 was not the same as what is typically seen in TVID. We used previous results from RNA sequencing of the hippocampus and spinal cord to test our hypothesis that myelin loss in these strains resulted from the convergent biological effects of multiple genetic risk variants, many previously unassociated with TMEV-induced diseases. These findings identify novel genetic targets and demonstrate the utility of genetically diverse models for uncovering complex neuroimmune interactions. Full article
(This article belongs to the Special Issue Latest Advances in Comparative Genomics)
Show Figures

Figure 1

23 pages, 27325 KB  
Article
Extracellular Vesicles from Streptococcus suis Promote Bacterial Pathogenicity by Disrupting Macrophage Metabolism
by Wenjie Jin, Jinpeng Li, Zhaoyu Yi, Zhiheng Chang, Yue Li, Yamin Shen, Yingying Quan, Yuxin Wang, Baobao Liu, Li Yi and Yang Wang
Microorganisms 2025, 13(11), 2469; https://doi.org/10.3390/microorganisms13112469 - 29 Oct 2025
Viewed by 563
Abstract
Streptococcus suis (S. suis) is an important zoonotic pathogen that causes severe disease in both humans and pigs, leading to substantial economic losses in the swine industry. Extracellular vesicles (EVs), as critical mediators of host–pathogen interactions, play key roles in bacterial [...] Read more.
Streptococcus suis (S. suis) is an important zoonotic pathogen that causes severe disease in both humans and pigs, leading to substantial economic losses in the swine industry. Extracellular vesicles (EVs), as critical mediators of host–pathogen interactions, play key roles in bacterial virulence and disease progression. This study aimed to investigate the biological properties of S. suis EVs and elucidate their role in the bacterium’s pathogenesis. We isolated and characterized S. suis EVs, which were found to contain diverse protein molecules. EVs were efficiently internalized by mammalian cells, and concentrations below 50 μg/mL did not affect cell viability. Following uptake, EVs suppressed the production of key pro-inflammatory cytokines (TNF-α, IL-1β, and IL-8) by modulating macrophage metabolism. They also downregulated the expression of major histocompatibility complex class II (MHC-II) and adhesion molecules (VCAM-1 and ICAM-1) during subsequent infections, potentially impairing macrophage-mediated clearance. In addition, EVs served as vectors for efficient cargo delivery and facilitated S. suis adhesion to and invasion of endothelial cells. In infection models, EVs markedly enhanced lethality in Galleria mellonella larvae and promoted tissue colonization in murine models. These findings suggest that S. suis EVs are key mediators of host–pathogen interactions, contributing to colonization and disease pathogenesis. Moreover, they offer novel insights and potential strategies for the prevention and control of S. suis infections. Full article
Show Figures

Figure 1

19 pages, 383 KB  
Article
HLA Class II Alleles and Suicidal Behavior: Evidence from a Case–Control Study
by Mihaela Elvira Cîmpianu, Mihaela Laura Vică Matei, Ștefana Bâlici, Gheorghe Zsolt Nicula, Elena Maria Domșa, Teodora Cîmpianu, Sergiu Ionica Rusu, Horia George Coman and Costel Vasile Siserman
Int. J. Mol. Sci. 2025, 26(20), 10181; https://doi.org/10.3390/ijms262010181 - 20 Oct 2025
Viewed by 498
Abstract
Suicidality is a complex multifactorial phenomenon strongly associated with major depression and other psychiatric disorders. Building on evidence implicating the Major Histocompatibility Complex (MHC) in modulating the immune and inflammatory processes characterizing psychiatric disorders, we hypothesized that specific HLA-DQB1 and HLA-DRB1 variants may [...] Read more.
Suicidality is a complex multifactorial phenomenon strongly associated with major depression and other psychiatric disorders. Building on evidence implicating the Major Histocompatibility Complex (MHC) in modulating the immune and inflammatory processes characterizing psychiatric disorders, we hypothesized that specific HLA-DQB1 and HLA-DRB1 variants may contribute to an increased genetic susceptibility to suicidal behavior. Human Leucocyte Antigen (HLA) typing by sequence-specific primers (PCR-SSP) was performed on a sample of 196 individuals, including 70 non-lethal suicide attempters, 28 cases of completed suicide, and matched controls. The *HLA-DQB1 02/06 (RR 1.60, CI95% 1.22–2.09, p = 0.03 *) and *HLA-DRB1 11/15 (RR 1.70, CI95% 1.3–2.24, p = 0.04 *) genotypes and the HLA-DRB115~DQB103 haplotype (RR 1.58, CI95% 1.22–2.04, p = 0.03 *) were found to favor suicidal behavior. Psychosocial determinants associated with an increased suicidal risk were bereavement of close relatives (linked with HLA-DQB1*02), memory dysfunction (HLA-DQB1*06), disillusionment (HLA-DRB1*07 and HLA-DRB1*15), and self-harm (HLA-DRB1*15). Our findings support the contributory role of HLA polymorphisms in shaping susceptibility to suicidal behavior. Full article
(This article belongs to the Section Molecular Biology)
15 pages, 882 KB  
Article
Evidence of Mixed Selection Acting on the MHC Class II DQA Gene in Captive Thai Elephant Populations
by Trifan Budi, Marie Roselle Enguito, Worapong Singchat, Thitipong Panthum, Ton Huu Duc Nguyen, Aingorn Chaiyes, Narongrit Muangmai, Darren K. Griffin, Prateep Duengkae and Kornsorn Srikulnath
Genes 2025, 16(10), 1180; https://doi.org/10.3390/genes16101180 - 10 Oct 2025
Viewed by 717
Abstract
Background: The health and viability of captive elephants, which are central to off-site conservation efforts and health management in Thailand, is threatened by emerging infectious diseases. This is partly due to genetic differences in immune-related genes, especially in the major histocompatibility complex (MHC) [...] Read more.
Background: The health and viability of captive elephants, which are central to off-site conservation efforts and health management in Thailand, is threatened by emerging infectious diseases. This is partly due to genetic differences in immune-related genes, especially in the major histocompatibility complex (MHC) and, among these, loci such as DQA play a crucial role in immune surveillance. Data pertaining to MHC polymorphisms in elephants are scarce, and thus this study investigated such polymorphisms and selection signatures in a partial fragment of exon 2 of the MHC Class II DQA gene. Methods: The approach we used targeted next-generation sequencing and diversity analyses of individuals from three captive elephant camps in Northern Thailand. Results: Eight alleles containing 11 SNPs were identified in the exon 2 fragment, encompassing both silent and missense mutations, some of which may influence immune function. Notably, the allele Elma-DQA*TH3, which is identical to Loaf-DQA*01 and Elma-DQA*01, previously reported as the most common alleles in Loxodonta and Elephas, was found at low frequencies. This shift may reflect local selective pressures that shape MHC allele distributions. Evidence of mixed selection (both positive and balancing) was detected in the partial fragment of DQA exon 2, suggesting a dynamic interplay between evolutionary forces. Positive selection likely reflects an adaptation to emerging or locally prevalent pathogens, whereas balancing selection maintains allelic diversity over time to enable a broad immunological response. Conclusions: Our findings reveal immunogenetic variations in captive Thai elephants, and provides insights into host–pathogen interactions that inform conservation and health strategies with the aim of improving disease resilience. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

20 pages, 2459 KB  
Review
The Immunoregulatory Mechanisms of Human Cytomegalovirus from Primary Infection to Reactivation
by Xiaodan Liu, Chang Liu and Ting Zhang
Pathogens 2025, 14(10), 998; https://doi.org/10.3390/pathogens14100998 - 2 Oct 2025
Cited by 1 | Viewed by 1880
Abstract
Human cytomegalovirus (HCMV) establishes lifelong latency following primary infection, residing within myeloid progenitor cells and monocytes. To achieve this, the virus employs multiple immune evasion strategies. It suppresses innate immune signaling by inhibiting Toll-like receptor and cGAS-STING pathways. In addition, the virus suppresses [...] Read more.
Human cytomegalovirus (HCMV) establishes lifelong latency following primary infection, residing within myeloid progenitor cells and monocytes. To achieve this, the virus employs multiple immune evasion strategies. It suppresses innate immune signaling by inhibiting Toll-like receptor and cGAS-STING pathways. In addition, the virus suppresses major histocompatibility complex (MHC)-dependent antigen presentation to evade T cell recognition. As the downregulation of MHC molecules may trigger NK cell activation, the virus compensates for this by expressing proteins such as UL40 and IL-10, which engage inhibitory NK cell receptors and block activating signals, thereby suppressing NK cell immune surveillance. Viral proteins like UL36 and UL37 block host cell apoptosis and necroptosis, allowing HCMV to persist undetected and avoid clearance. In settings of profound immunosuppression, such as after allogeneic hematopoietic stem cell transplantation (allo-HSCT) or solid organ transplantation, slow immune reconstitution creates a window for viral reactivation. Likewise, immunosenescence and chronic low-grade inflammation during aging increases the risk of reactivation. Once reactivated, HCMV triggers programmed cell death, releasing viral PAMPs (pathogen-associated molecular patterns) and host-derived DAMPs (damage-associated molecular patterns). This release fuels a potent inflammatory response, promoting further viral reactivation and exacerbating tissue damage, creating a vicious cycle. This cycle of inflammation and reactivation contributes to both transplant-related complications and the decline of antiviral immunity in the elderly. Therefore, understanding the immune regulatory mechanisms that govern the switch from latency to reactivation is critical, especially within the unique immune landscapes of transplantation and aging. Elucidating these pathways is essential for developing strategies to prevent and treat HCMV-related disease in these high-risk populations. Full article
(This article belongs to the Special Issue Pathogen–Host Interactions: Death, Defense, and Disease)
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
Viewed by 840
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

18 pages, 979 KB  
Article
Genetic Diversity and Selection of MHC I-UAA in Clariid Catfish from Thailand: Implications for Breeding and Conservation
by Ton Huu Duc Nguyen, Piangjai Chalermwong, Chananya Patta, Wattanawan Jaito, Worapong Singchat, Thitipong Panthum, Trifan Budi, Kednapat Sriphairoj, Sittichai Hatachote, Prapansak Srisapoome, Narongrit Muangmai, Darren K. Griffin, Agostinho Antunes, Prateep Duengkae and Kornsorn Srikulnath
Genes 2025, 16(9), 1106; https://doi.org/10.3390/genes16091106 - 18 Sep 2025
Viewed by 663
Abstract
Background/Objectives: Understanding variabilities in the Major Histocompatibility Complex class I (MHC I) gene is essential for evaluating immunogenetic diversity in clariid catfish. MHC I plays a critical role in immune defense by presenting endogenous antigens to cytotoxic T cells. Therefore, we [...] Read more.
Background/Objectives: Understanding variabilities in the Major Histocompatibility Complex class I (MHC I) gene is essential for evaluating immunogenetic diversity in clariid catfish. MHC I plays a critical role in immune defense by presenting endogenous antigens to cytotoxic T cells. Therefore, we aimed to investigate the genetic diversity, selection patterns, and phylogenetic relationships of MHC I alleles in three important clariid catfish species (Clarias gariepinus, Clarias macrocephalus, and Clarias batrachus) across wild and hatchery populations in Thailand. Methods: Targeted next-generation sequencing of a 174 bp fragment partial exon 6 of MHC I-UAA gene was performed, along with phylogenetic analyses, neutrality tests and dN/dS analyses. Results: Overall, 91 novel alleles were identified in 674 individuals, all of which were novel (100% novelty), with none matching existing reference sequences, thereby revealing extensive variation in population-specific variants. Phylogenetic analyses revealed allele sharing among species, which was consistent with balanced selection. Neutrality tests and dN/dS analyses provided evidence of both purifying and diversifying selection, with episodic positive selection detected at multiple codon sites associated with the antigen-binding α1 domain. Distinct selection patterns among populations, influenced by local environmental conditions and human pressures, along with high allele richness, are reflected in the diversity of immunogenetic variations. Conclusions: These findings provide critical insights into immune adaptation and highlight the potential of MHC I as a functional marker for genetic monitoring. Although a causal relationship between MHC I polymorphism and disease resistance is debated, studies suggest associations with pathogen survival, indicating future implications for aquaculture breeding and conservation, particularly in marker-assisted selection for broodstock management in Thailand. Full article
(This article belongs to the Section Animal Genetics and Genomics)
Show Figures

Figure 1

22 pages, 7902 KB  
Article
CD63-Mediated SARS-CoV-2 RBD Fusion Neoantigen DNA Vaccine Enhances Antitumor Immune Response in a Mouse Panc02 Model via EV-Targeted Delivery
by Guang Liu, Ziqing Yuan, Ziyi Wu, Qiyv Yang, Tingbo Ding, Ker Yu and Jibin Dong
Vaccines 2025, 13(9), 977; https://doi.org/10.3390/vaccines13090977 - 16 Sep 2025
Viewed by 1161
Abstract
Background: Although DNA vaccines offer a flexible platform for tumor immunotherapy, their weak immunogenicity remains a key limitation. This study aimed to improve the immunogenicity of DNA vaccines by enhancing the efficiency of tumor neoantigen delivery through extracellular vesicles (EVs), thereby promoting stronger [...] Read more.
Background: Although DNA vaccines offer a flexible platform for tumor immunotherapy, their weak immunogenicity remains a key limitation. This study aimed to improve the immunogenicity of DNA vaccines by enhancing the efficiency of tumor neoantigen delivery through extracellular vesicles (EVs), thereby promoting stronger dendritic cell (DC) activation and antitumor responses. Methods: A novel DNA vaccine (pCSP) was engineered by fusing tumor-specific neoantigens to the EV-associated protein CD63 and incorporating a SARS-CoV-2 receptor-binding domain (RBD) fragment to facilitate EV uptake by DCs. The resulting EVs were expected to carry neoantigens into the immunoproteasome for major histocompatibility complex I (MHC-I) presentation. The immunological and antitumor effects of pCSP were assessed through in vitro functional assays and in vivo experiments in a murine pancreatic cancer model. Safety was evaluated through histological and biochemical analyses. Results: In vitro, pCSP significantly promoted EV internalization by DCs by approximately twofold and enhanced their immune activation, as evidenced by elevated cytokine production. In vivo, pCSP markedly suppressed tumor growth with a decrease in volume by over 70% relative to controls, boosted CD8+ T cell responses, and increased immune infiltration into the tumor microenvironment. Safety assessments revealed that while liver/kidney function markers were within physiological ranges, mild inflammatory infiltrates were consistently observed in the lungs, indicating a localized safety concern that warrants further monitoring. Conclusions: The pCSP vaccine enhances the immunogenicity of neoantigen DNA vaccines by improving EV uptake and immune activation in DCs. These findings provide a potential strategy for improving DNA vaccine efficacy in the context of cancer immunotherapy while maintaining acceptable safety. Full article
(This article belongs to the Special Issue Advances in Immunotherapy of Cancer)
Show Figures

Figure 1

16 pages, 836 KB  
Article
mRNA Multipeptide-HLA Class II Immunotherapy for Melanoma
by Apostolos P. Georgopoulos, Lisa M. James and Matthew Sanders
Cells 2025, 14(18), 1430; https://doi.org/10.3390/cells14181430 - 12 Sep 2025
Cited by 2 | Viewed by 1110
Abstract
Human Leukocyte Antigen (HLA) Class II (HLA-II) molecules bind peptides of phagocytosed non-self proteins and present them on the cell surface to circulating CD4+ T lymphocytes. A successful binding of the presented peptide with the T cell receptor (TCR) activates the CD4+ T [...] Read more.
Human Leukocyte Antigen (HLA) Class II (HLA-II) molecules bind peptides of phagocytosed non-self proteins and present them on the cell surface to circulating CD4+ T lymphocytes. A successful binding of the presented peptide with the T cell receptor (TCR) activates the CD4+ T cell, leading to the production of antibodies against the peptide (and the protein of its origin) by the B cell and augmentation of the cytotoxic and memory functions of CD8+ T cells. The first and essential step in this process is the successful formation of a stable peptide-HLA-II complex (pHLA-II), which is achieved when the peptide binds with high affinity to the HLA-II molecule. Such highly antigenic non-self peptides occur in melanoma-associated proteins and could be used as antitumor agents when bound to a matching HLA-II molecule. The objective of this study was to identify such peptides from 15 melanoma-associated proteins. We determined in silico the predicted binding affinity (IC50) of all pHLA-II pairs between 192 common HLA-II molecules and all possible linear 15-amino acid (15-mer) peptides (epitopes) of 15 known melanoma-associated antigens (N = 3466 epitopes) for a total of 192 × 3466 = 665,472 determinations. From this set, we identified epitopes with strong antigenicity (predicted best binding affinity [PBBA] IC50 < 50 nM). Of a total of 665,472 pHLA-II tested, 5941 (0.89%) showed strong PBBA, stemming from 117 HLA-II alleles and 679 distinct epitopes. This set of 5941 pHLA-II pairs with predicted high antigenicity possesses the requisite information for devising multipeptide vaccines with those epitopes alone or in combination with the corresponding HLA-II molecules. The results obtained have a major implication for cancer therapy, namely that the administration of subsets of the 679 high antigenicity epitopes above, alone or in combination with their associated HLA-II molecules, would be successful in engaging CD4+ T helper lymphocytes to augment the cytotoxic action and memory of CD8+ T lymphocytes and induce the production of antitumor antibodies by B cells. This therapy would be effective in other solid tumors (in addition to melanoma) and would be enhanced by concomitant immunotherapy with immune checkpoint inhibitors. Full article
Show Figures

Figure 1

Back to TopTop