[go: up one dir, main page]
More Web Proxy on the site http://driver.im/
You seem to have javascript disabled. Please note that many of the page functionalities won't work as expected without javascript enabled.
 
 
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
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
remove_circle_outline
remove_circle_outline

Search Results (7,441)

Search Parameters:
Keywords = innate immunity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
18 pages, 20003 KiB  
Article
ST8SIA6 Sialylates CD24 to Enhance Its Membrane Localization in BRCA
by Jinxia He, Fengchao Zhang, Baihai Wu and Wengong Yu
Cells 2025, 14(1), 9; https://doi.org/10.3390/cells14010009 (registering DOI) - 26 Dec 2024
Abstract
CD24, a highly sialylated glycosyl-phosphatidyl-inositol (GPI) cell surface protein that interacts with sialic acid-binding immunoglobulin-like lectins (Siglecs), serves as an innate immune checkpoint and plays a crucial role in inflammatory diseases and tumor progression. Recently, cytoplasmic CD24 has been observed in samples from [...] Read more.
CD24, a highly sialylated glycosyl-phosphatidyl-inositol (GPI) cell surface protein that interacts with sialic acid-binding immunoglobulin-like lectins (Siglecs), serves as an innate immune checkpoint and plays a crucial role in inflammatory diseases and tumor progression. Recently, cytoplasmic CD24 has been observed in samples from patients with cancer. However, whether sialylation governs the subcellular localization of CD24 in cancer remains unclear, and the impact of CD24 expression and localization on the clinical prognosis of cancer remains controversial. Here, we performed a systematic pan-cancer analysis of the gene expression levels and clinical correlation of CD24. Our analysis revealed that CD24 was highly expressed in breast tumor tissues and tumor cells, significantly shortening patient survival time. However, this correlation was not evident in other types of cancer. Additionally, a correlation analysis of CD24 levels with sialyltransferases (STs) revealed that ST8SIA6 is the key ST affecting CD24 sialylation. Further investigation demonstrated that ST8SIA6 directly modified CD24, promoting its localization to the cell membrane. Taken together, these findings elucidate, for the first time, the mechanisms by which ST8SIA6 regulates CD24 subcellular localization, providing new insights into the biological functions and applications of CD24. Full article
Show Figures

Figure 1

Figure 1
<p>The flowchart illustrates the pan-cancer analysis of CD24. Abbreviations: IHC, immunohistochemistry; RNA-seq, RNA sequencing; DBs, databases; TILs, tumor-infiltrating lymphocytes.</p>
Full article ">Figure 2
<p>Expression analysis of the <span class="html-italic">CD24</span> gene in various human cancers. (<b>A</b>) The RNA expression levels of <span class="html-italic">CD24</span> in human cancers were analyzed using the TIMER algorithm. The statistical significance computed by the Wilcoxon test is annotated by the number of asterisks. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) The RNA expression levels of <span class="html-italic">CD24</span> in various human cancer cell lines were analyzed using data from the Human Protein Atlas database.</p>
Full article ">Figure 3
<p>The clinical relevance of <span class="html-italic">CD24</span> gene expression levels across various cancer types. (<b>A</b>) The clinical correlation between <span class="html-italic">CD24</span> gene expression levels and various types of cancer was analyzed using the TIMER algorithm, incorporating key clinical factors, including age, tumor stage, and tumor purity. A heatmap was drawn to show the normalized coefficient of the gene in the Cox model. Z-Score &gt; 0, <span class="html-italic">p</span> &lt; 0.05, increased risk; Z-Score &lt; 0, <span class="html-italic">p</span> &lt; 0.05, decreased risk; <span class="html-italic">p</span> &gt; 0.05, not significant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) The impact of <span class="html-italic">CD24</span> levels on the survival rates of patients with BRCA, MESO, and SKCM. HR represents the hazard ratio. (<b>C</b>) The impact of <span class="html-italic">CD24</span> levels on the survival rates of patients COAD, LGG, and UCS. HR represents the hazard ratio. (<b>D</b>) Spearman’s correlations between <span class="html-italic">CD24</span> gene expression levels and TILs, including activated CD8<sup>+</sup> T cells, activated CD4<sup>+</sup> T cells, natural killer cells, activated dendritic cells, macrophages, monocytes, and neutrophils, across various types of human cancers were obtained from the TISIDB database. Rho represents the Spearman’s correlation coefficient. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>The subcellular localization of CD24 protein was analyzed using data from the Human Protein Atlas database. (<b>A</b>) The 13 cancer tissues with the highest levels of CD24 in the cytoplasm and membrane, namely, THCA, HNSC, TGCT, CESC, STAD, BLCA, OV, lung cancer (LUAD and LUSC), PRAD, renal cancer (KICH, KIRC, and KIRP), UCEC, PAAD, and BRCA. (<b>B</b>) Representative immunohistochemical staining images of changes in membranous and cytoplasmic CD24 levels in high- and low-stage tumors from BLCA and PARD patients are shown (scale bar, 100 μm and 25 μm). (<b>C</b>) Representative immunohistochemical staining images of CD24 in the nucleus (scale bar, 100 μm and 20 μm). (<b>D</b>) Representative immunohistochemical staining images of membranous and cytoplasmic CD24 in THCA, TGCT, OV, and BRCA (scale bar, 100 μm and 25 μm).</p>
Full article ">Figure 5
<p>The correlations between <span class="html-italic">CD24</span> levels and the levels of ST8 family members in human cancer were analyzed using the TIMER database. (<b>A</b>) <span class="html-italic">CD24</span> levels were positively correlated with <span class="html-italic">ST8SIA2</span> and <span class="html-italic">ST8SIA6</span> levels in BRCA. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001. (<b>B</b>) The correlations between <span class="html-italic">CD24</span> and <span class="html-italic">ST8SIA6</span> levels were analyzed in various human cancers. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 6
<p>The correlations between <span class="html-italic">Siglec-7/9/10</span> levels and the abundance of tumor-infiltrating macrophages in BRCA (<b>A</b>) and other human cancers (<b>B</b>) were analyzed using the TIMER database. Spearman’s rho &gt; 0, <span class="html-italic">p</span> &lt; 0.05, positive correlation; Spearman’s rho &lt; 0, <span class="html-italic">p</span> &lt; 0.05, negative correlation; <span class="html-italic">p</span> &gt; 0.05, not significant. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 7
<p>CD24 is directly modified by ST8SIA6. (<b>A</b>) The co-localization of CD24 and Siglec-E Fc fluorescence was observed in 4T1 cells (scale bar, 10 μm). (<b>B</b>) The co-localization of CD24 and Siglec-E fluorescence was observed in TNBC tumor tissues (scale bar, 20 μm and 10 μm). (<b>C</b>) The co-localization of CD24 and ST8SIA6 fluorescence was observed in 4T1 cells (scale bar, 10 μm). (<b>D</b>) The co-localization of CD24 and ST8SIA6 fluorescence was observed in TNBC tumor tissues (scale bar, 50 μm and 10 μm).</p>
Full article ">Figure 8
<p>The impact of ST8SIA6 on the subcellular localization and expression of CD24. (<b>A</b>) The expression of ST8SIA6 was detected by western blotting in <span class="html-italic">St8sia6</span>-overexpressing (OE-<span class="html-italic">St8sia6</span>) and <span class="html-italic">St8sia6</span>-knockout (sg<span class="html-italic">St8sia6</span>) 4T1 cells. (<b>B</b>) The expression of ST8SIA6 was detected by immunofluorescence in OE-<span class="html-italic">St8sia6</span> and sg<span class="html-italic">St8sia6</span> 4T1 cells (scale bar, 25 μm). (<b>C</b>) The effects of ST8SIA6 on CD24 expression were detected by flow cytometry. (<b>D</b>) Quantitative analysis of CD24 expression in 4T1 cells. The graph shows the median fluorescence intensity (MFI) of CD24. Data are presented as the mean ± standard error of the mean. The <span class="html-italic">p</span>-values were calculated using a one-way analysis of variance. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01. (<b>E</b>) The effect of ST8SIA6 on the subcellular localization of CD24 was determined by immunofluorescence (scale bar, 10 μm).</p>
Full article ">
20 pages, 1221 KiB  
Review
Toward Mycobacterium tuberculosis Virulence Inhibition: Beyond Cell Wall
by Maria S. Kotliarova, Mikhail S. Shumkov and Anna V. Goncharenko
Microorganisms 2025, 13(1), 21; https://doi.org/10.3390/microorganisms13010021 - 26 Dec 2024
Abstract
Mycobacterium tuberculosis (Mtb) is one of the most successful bacterial pathogens in human history. Even in the antibiotic era, Mtb is widespread and causes millions of new cases of tuberculosis each year. The ability to disrupt the host’s innate and adaptive immunity, as [...] Read more.
Mycobacterium tuberculosis (Mtb) is one of the most successful bacterial pathogens in human history. Even in the antibiotic era, Mtb is widespread and causes millions of new cases of tuberculosis each year. The ability to disrupt the host’s innate and adaptive immunity, as well as natural persistence, complicates disease control. Tuberculosis traditional therapy involves the long-term use of several antibiotics. Treatment failures are often associated with the development of resistance to one or more drugs. The development of medicines that act on new targets will expand treatment options for tuberculosis caused by multidrug-resistant or extensively drug-resistant Mtb. Therefore, the development of drugs that target virulence factors is an attractive strategy. Such medicines do not have a direct bacteriostatic or bactericidal effect, but can disarm the pathogen so that the host immune system becomes able to eliminate it. Although cell wall-associated targets are being actively studied for anti-TB drug development, other virulence factors important for adaptation and host interaction are also worth comprehensive analysis. In this review, specific Mtb virulence factors (such as secreted phosphatases, regulatory systems, and the ESX-1 secretion system) are identified as promising targets for novel anti-virulence drug development. Additionally, models for the search of virulence inhibitors are discussed, such as virtual screening in silico, in vitro enzyme inhibition assay, the use of recombinant Mtb strains with reporter constructs, phenotypic analysis using in vitro cell infection models and specific environments. Full article
(This article belongs to the Section Antimicrobial Agents and Resistance)
Show Figures

Figure 1

Figure 1
<p>Schematic representation of mycobacterium tuberculosis influence on the key components of the phagocyte antimicrobial response. Details are given in the text.</p>
Full article ">Figure 2
<p>The selection of an appropriate screening model for screening virulence factor inhibitors. <b>Left panel</b>. The scheme of virulence realization represents events that can provide the output in a screening model. Once the bacterium infects the host, it is exposed to a variety of stressors. The signal from the external environment (the internal environment of the host in this case) is transmitted into bacterial cytoplasm, activates specific transcription factors and thus initiates the transcription of genes (1) that are essential for survival. Some of these genes encode effector proteins (2) that are secreted (3) across the cell membrane and/or cell wall and perform effector functions in the host (4), while some of the expressed proteins are involved in adaptation mechanisms within the bacterial cells (5). <b>Right panel</b>. The activity of regulatory systems and associated transcription factors (1) can be evaluated through the use of reporter strains, wherein the reporter gene is under the control of the promoter of the regulated region. The binding and blocking of a known reporter protein can be directly modelled in silico and screened in model systems with recombinant purified proteins (2). The activity of secretion systems can also be evaluated directly in strains where a known substrate of the secretion system is fused to a reporter (3). In vitro models using infected cells facilitate the assessment of parameters like intracellular survival of Mtb (5) and specific responses of infected cells, such as cell death (4). By in vitro simulating of aggressive conditions of the infection site and assessing the survival of Mtb cells, the function of genes responsible for adaptation and its disturbances could be evaluated.</p>
Full article ">
21 pages, 310 KiB  
Review
Employing the Oncolytic Vesicular Stomatitis Virus in Cancer Virotherapy: Resistance and Clinical Considerations
by Alaa A. Abdelmageed, Stephen Dewhurst and Maureen C. Ferran
Viruses 2025, 17(1), 16; https://doi.org/10.3390/v17010016 - 25 Dec 2024
Abstract
Vesicular Stomatitis Virus (VSV) has emerged as a promising candidate for various clinical applications, including vaccine development, virus pseudotyping, and gene delivery. Its broad host range, ease of propagation, and lack of pre-existing immunity in humans make it ideal for therapeutic use. VSV’s [...] Read more.
Vesicular Stomatitis Virus (VSV) has emerged as a promising candidate for various clinical applications, including vaccine development, virus pseudotyping, and gene delivery. Its broad host range, ease of propagation, and lack of pre-existing immunity in humans make it ideal for therapeutic use. VSV’s potential as an oncolytic virus has garnered attention; however, resistance to VSV-mediated oncolysis has been observed in some cell lines and tumor types, limiting its effectiveness. This review provides a detailed analysis of recent advances in VSV-based oncolysis, focusing on resistance mechanisms such as sustained type-I IFN signaling, upregulation of ISGs, immune cell activation, the tumor microenvironment (TME), and tumor-intrinsic factors. Strategies to overcome resistance include enhancing viral oncoselectivity, inhibiting IFN responses, modulating the TME, and combining VSV with chemotherapies, radiation, and immune checkpoint inhibitors. Several VSV-based phase I/II clinical trials show promise; however, addressing resistance and developing novel strategies to enhance therapeutic efficacy are essential for realizing the full potential of VSV oncolytic virotherapy. Future research should focus on patient-specific approaches, as tumor heterogeneity implies varying resistance mechanisms. Personalized treatments tailored to tumor molecular profiles, along with identifying biomarkers predictive of resistance to VSV oncolysis, will enhance patient selection and enable more effective, individualized VSV-based therapies. Full article
(This article belongs to the Special Issue Vesicular Stomatitis Virus (VSV))
Show Figures

Graphical abstract

Graphical abstract
Full article ">
12 pages, 2917 KiB  
Article
Cytogenetic Identification and Molecular Marker Analysis of Two Wheat–Thinopyrum ponticum Translocations with Stripe Rust Resistance
by Guotang Yang, Yi Han, Huihui Yin, Xingfeng Li, Honggang Wang and Yinguang Bao
Plants 2025, 14(1), 27; https://doi.org/10.3390/plants14010027 (registering DOI) - 25 Dec 2024
Abstract
Stripe rust, induced by Puccinia striiformis f. sp. tritici (Pst), is one of the most destructive fungal diseases of wheat worldwide. Thinopyrum ponticum, a significant wild relative for wheat improvement, exhibits innate immunity to this disease. To transfer the stripe [...] Read more.
Stripe rust, induced by Puccinia striiformis f. sp. tritici (Pst), is one of the most destructive fungal diseases of wheat worldwide. Thinopyrum ponticum, a significant wild relative for wheat improvement, exhibits innate immunity to this disease. To transfer the stripe rust resistance gene from Th. ponticum to wheat, two translocation lines, SN21171 and SN52684, were produced through distant hybridization techniques. Disease evaluation results showed that these two lines were immune to Pst species CYR32 at the adult plant stage. Molecular cytogenetic analyses and specific intron-targeting markers amplification results revealed that SN21171 and SN52684 harbor several T3Eb-3DS·3DL and T1Eb-1BS·1BL translocation chromosomes. Furthermore, the comparison of the chromosome karyotype from two translocation lines and their recurrent parent YN15, revealed that structural variation occurred in chromosomes 2A, 5A, 2B, 4B, 5B, and 6B in SN21171 and chromosomes 5A, 3B, 4B, 5B, 6B, and 7B in SN52684. Agronomic trait assessments uncovered advantageous properties in both lines, with SN21171 matching the recurrent parent and SN52684 exhibiting elevated higher grain number per main spike and increased thousand grain weight. These two translocation lines and specific markers may apply to wheat stripe rust-resistance breeding. Full article
(This article belongs to the Special Issue Crop Genetics and Breeding)
Show Figures

Figure 1

Figure 1
<p>The disease resistance evaluation of SN21171, SN52684, and their parent YN15 at the adult plant stage. (<b>a</b>) <span class="html-italic">Th. ponticum</span>; (<b>b</b>) SN21171; (<b>c</b>) SN52684; (<b>d</b>) YN15; (<b>e</b>) HXH.</p>
Full article ">Figure 2
<p>Cytogenetic analyses of SN21171 and SN52684. The GISH patterns of SN21171 (<b>a</b>) and SN52684 (<b>c</b>) with <span class="html-italic">Th. ponticum</span> gDNA as a probe. The mc-FISH patterns of SN21171 (<b>b</b>) and SN52684 (<b>d</b>) using eight single-strand oligos. The arrows note a pair of translocated chromosomes. Bar = 10 μm.</p>
Full article ">Figure 3
<p>Comparison of chromosome karyotypes in SN21171 SN52684, and YN15. (<b>a</b>) YN15; (<b>b</b>) SN21171; (<b>c</b>) SN52684. The arrows note locations with different FISH bands.</p>
Full article ">Figure 4
<p>Specific intron-targeting marker amplification in SN21171 and SN52684. (<b>a</b>) CINAU1185; (<b>b</b>) CINAU1195; (<b>c</b>) CINAU1214; (<b>d</b>) CINAU957; (<b>e</b>) CINAU959. M: marker; 1: <span class="html-italic">Th. ponticum</span>; 2: <span class="html-italic">Th. elongatum</span>; 3: <span class="html-italic">Th. bessarabicum</span>; 4: <span class="html-italic">Pseudoroegneria</span>; 5: YN15; 6: SN21171; 7: SN52684. The arrows indicate specific bands.</p>
Full article ">Figure 5
<p>The agronomic performance of SN21171, SN52684, and YN15. (<b>a</b>) SN21171; (<b>b</b>) SN52684; (<b>c</b>) YN15.</p>
Full article ">
23 pages, 1162 KiB  
Review
Biological Functions and Clinical Significance of the ABCG1 Transporter
by Stanislav Kotlyarov and Anna Kotlyarova
Biology 2025, 14(1), 8; https://doi.org/10.3390/biology14010008 - 25 Dec 2024
Abstract
ATP-binding cassette (ABC) transporters are a large family of proteins that transport various substances across cell membranes using energy from ATP hydrolysis. ATP-binding cassette sub-family G member 1 (ABCG1) is a member of the ABCG subfamily of transporters and performs many important functions, [...] Read more.
ATP-binding cassette (ABC) transporters are a large family of proteins that transport various substances across cell membranes using energy from ATP hydrolysis. ATP-binding cassette sub-family G member 1 (ABCG1) is a member of the ABCG subfamily of transporters and performs many important functions, such as the export of cholesterol and some other lipids across the membranes of various cells. Cholesterol transport is the mechanism that links metabolism and the innate immune system. Due to its lipid transport function, ABCG1 may contribute to the prevention of atherosclerosis and is involved in the functioning of the lung, pancreas, and other organs and systems. However, the full clinical significance of ABCG1 is still unknown and is a promising area for future research. Full article
(This article belongs to the Special Issue ABCG1 and HDL in Health and Disease)
Show Figures

Figure 1

Figure 1
<p>Structure of «half» transporter ABCG1 and «full» ABC transporters (<b>A</b>) and the ABCG1 transport function (<b>B</b>). Note: Unlike classical «full» ABC transporters, which consist of two repeating modules, ABCG1 is a «half» transporter containing one intracellular ATP-binding nucleotide-binding domain (NBD) and one transmembrane domain (TMD).</p>
Full article ">Figure 2
<p>Biological and potential clinical significance of the ABCG1 transporter. Through the regulation of cholesterol transport, ABCG1 is involved in the regulation of inflammation involving macrophages (<b>A</b>), with potential clinical relevance to atherosclerosis, tumor progression, pancreatic, adrenal, brain, and breast function (<b>B</b>).</p>
Full article ">
19 pages, 995 KiB  
Review
Extracellular Cold-Inducible RNA-Binding Protein and Hemorrhagic Shock: Mechanisms and Therapeutics
by Naureen Rashid, Zhijian Hu, Asha Jacob and Ping Wang
Biomedicines 2025, 13(1), 12; https://doi.org/10.3390/biomedicines13010012 - 25 Dec 2024
Abstract
Hemorrhagic shock is a type of hypovolemic shock and a significant cause of trauma-related death worldwide. The innate immune system has been implicated as a key mediator in developing severe complications after shock. Inflammation from the innate immune system begins at the time [...] Read more.
Hemorrhagic shock is a type of hypovolemic shock and a significant cause of trauma-related death worldwide. The innate immune system has been implicated as a key mediator in developing severe complications after shock. Inflammation from the innate immune system begins at the time of initial insult; however, its activation is exaggerated, resulting in early and late-stage complications. Hypoxia and hypoperfusion lead to the release of molecules that act as danger signals known as damage-associated molecular patterns (DAMPs). DAMPs continue to circulate after shock, resulting in excess inflammation and tissue damage. We recently discovered that cold-inducible RNA-binding protein released into the extracellular space acts as a DAMP. During hemorrhagic shock, hypoperfusion leads to cell necrosis and the release of CIRP into circulation, triggering both systemic inflammation and local tissue damage. In this review, we discuss extracellular cold-inducible RNA-binding protein (eCIRP)’s role in sterile inflammation, as well as its various mechanisms of action. We also share our more newly developed anti-eCIRP agents with the eventual goal of producing drug therapies to mitigate organ damage, reduce mortality, and improve patient outcomes related to hemorrhagic shock. Finally, we suggest that future preclinical studies are required to develop the listed therapeutics for hemorrhagic shock and related conditions. In addition, we emphasize on the challenges to the translational phase and caution that the therapy should allow the immune system to continue to function well against secondary infections during hospitalization. Full article
(This article belongs to the Special Issue Molecular Mechanisms and Therapeutics in Hemorrhagic Shock)
Show Figures

Figure 1

Figure 1
<p>Mechanisms of action of eCIRP in hemorrhagic shock. Damaged vessels are unable to deliver blood to tissue due to hemorrhage. The loss of perfusion and resultant hypoxemia result in ischemic damage to cells. Due to hypoxemia, cellular metabolism shifts to anaerobic metabolism via glycolysis which leads to increasing lactate production. The accumulation of lactate creates an acidotic environment, causing further cell damage. The accumulation of these factors results in cell death by varying mechanisms including apoptosis, necrosis, and necroptosis. As a result of cell death, the cell membrane is disrupted, and intracellular CIRP is released into the extracellular space, becoming eCIRP, as shown in the top right. During the shock state, blood flow is shunted towards vital organs. eCIRP, now in systemic circulation, will also be shunted towards these organs, causing increased inflammation. eCIRP will bind to TLR4 and triggering receptor expressed on myeloid cells (TREM1) receptors on both tissue and circulating macrophages, demonstrated in the bottom left. As a result of this binding, the macrophage will release pro-inflammatory cytokines into circulation, worsening inflammation in the already ischemic tissues. A detailed image shown in the bottom right illustrates eCIRP acting on TLR4/MyD88 and inducing damage to the mitochondria, causing an increase in cytosolic DNA. This cytosolic DNA via the cGAS pathway activates stimulator of interferon genes (STING) on the endoplasmic reticulum membrane. This results in an increase in pIRF, a transcription factor responsible for increasing expression of type I <span class="html-italic">IFNs</span>. The activation of this pathway increases the release of type I IFNs into the circulation. These pathways collectively lead to inflammation and injury in hemorrhagic shock.</p>
Full article ">Figure 2
<p>eCIRP as a therapeutic target in hemorrhagic shock. Both C23 and M3 are small peptides derived from the human sequence of CIRP. C23 has high affinity to the TLR4/MD2 complex, and M3 has specific binding for the TREM1 receptor. Thus, both peptides block the binding of eCIRP to their respective receptors. In contrast, A12 is a synthetic oligonucleotide consisting of a poly(A) tail mimic that binds to eCIRP, blocking its binding site to the TLR4/MD2 complex. PS-OME miR 130 is a microRNA with stabilizing adjustments, including 3 phosphorothioate (PS) bonds at the 5′ and 3′ ends and 2′Omethyl (2′Ome) bases. It has a strong binding affinity to eCIRP, thereby preventing eCIRP from signaling via the TLR4/MD2 signaling pathway. As a result, these small molecule peptides and oligonucleotides prevent the interaction between endogenous eCIRP and its target receptors on macrophages. As a result, the inflammatory cascade is inhibited, and the hyperinflammatory response at the initial insult from hemorrhagic shock is prevented.</p>
Full article ">
54 pages, 696 KiB  
Review
Cathelicidins: Opportunities and Challenges in Skin Therapeutics and Clinical Translation
by Lenka Dzurová, Edita Holásková, Hana Pospíšilová, Gabriela Schneider Rauber and Jitka Frébortová
Antibiotics 2025, 14(1), 1; https://doi.org/10.3390/antibiotics14010001 - 24 Dec 2024
Abstract
Cathelicidins are a group of cationic, amphipathic peptides that play a vital role in the innate immune response of many vertebrates, including humans. Produced by immune and epithelial cells, they serve as natural defenses against a wide range of pathogens, including bacteria, viruses, [...] Read more.
Cathelicidins are a group of cationic, amphipathic peptides that play a vital role in the innate immune response of many vertebrates, including humans. Produced by immune and epithelial cells, they serve as natural defenses against a wide range of pathogens, including bacteria, viruses, and fungi. In humans, the cathelicidin LL-37 is essential for wound healing, maintaining skin barrier integrity, and combating infections. Cathelicidins of different origins have shown potential in treating various skin conditions, including melanoma, acne, and diabetic foot ulcers. Despite their promising therapeutic potential, cathelicidins face significant challenges in clinical application. Many peptide-based therapies have failed in clinical trials due to unclear efficacy and safety concerns. Additionally, the emergence of bacterial resistance, which contradicts initial claims of non-resistance, further complicates their development. To successfully translate cathelicidins into effective clinical treatments, therefore, several obstacles must be addressed, including a better understanding of their mechanisms of action, sustainable large-scale production, optimized formulations for drug delivery and stability, and strategies to overcome microbial resistance. This review examines the current knowledge of cathelicidins and their therapeutic applications and discusses the challenges that hinder their clinical use and must be overcome to fully exploit their potential in medicine. Full article
(This article belongs to the Section Antimicrobial Peptides)
14 pages, 1891 KiB  
Article
Balneotherapy (Mud-Bath Therapy) with a Peloid Enriched with Rosmarinic Acid Enhances Clinical Outcomes and Innate Immune Benefits in Elderly Patients with Osteoarthritis: A Pilot Study
by Eduardo Ortega-Collazos, María Dolores Hinchado, Eduardo Otero, Casimiro Fermín López-Jurado, Isabel Gálvez, José Luis Legido, Juan Francisco Sánchez Muñoz-Torrero, Eduardo Ortega and Silvia Torres-Piles
Appl. Sci. 2024, 14(24), 12017; https://doi.org/10.3390/app142412017 - 22 Dec 2024
Viewed by 385
Abstract
Osteoarthritis (OA) is a common chronic condition that causes pain and disability, particularly in the elderly, resulting in significant limitations on mobility and overall quality of life. Balneotherapy using peloids (mud therapy) is an effective, non-pharmacological treatment for OA that improves symptoms and [...] Read more.
Osteoarthritis (OA) is a common chronic condition that causes pain and disability, particularly in the elderly, resulting in significant limitations on mobility and overall quality of life. Balneotherapy using peloids (mud therapy) is an effective, non-pharmacological treatment for OA that improves symptoms and function. This pilot study aimed to assess whether a controlled-matured peloid, supplemented with rosmarinic acid (RosA), could enhance clinical outcomes, functional status, and immune response in OA patients. The study involved 42 elderly OA patients (mean age 70), comparing a 10-day balneotherapy cycle using either a RosA-fortified or non-fortified peloid. The effects on pain (Visual Analogue Scale), functional status (WOMAC, knee flexion/extension), quality of life (EUROQOL), and innate immune response (neutrophil phagocytic and microbicidal activity) were evaluated. Both treatments resulted in significant improvements in pain (by approximately 60%), function, and quality of life, but the RosA-fortified peloid led to greater benefits, particularly in the anxiety/depression dimension of the EUROQOL questionnaire and in enhancing neutrophil immune responsiveness. These findings suggest that RosA supplementation may further improve the therapeutic effects of mud therapy for OA management. Full article
Show Figures

Figure 1

Figure 1
<p>Flowchart of patients through the study.</p>
Full article ">Figure 2
<p>Knee flexion (<b>A</b>) and extension (<b>B</b>) in OA patients after the mud therapy intervention. CMT = Group of patients receiving controlled mud therapy; CMRT = group of patients receiving controlled mud therapy fortified with RosA. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.001, compared to the corresponding baseline values.</p>
Full article ">Figure 3
<p>Perceived pain in OA patients after the mud therapy intervention. VAS = visual analogue scale; CMT = group of patients receiving controlled mud therapy; CMRT = group of patients receiving controlled mud therapy fortified with RosA. *** <span class="html-italic">p</span> &lt; 0.001, compared to the corresponding baseline values.</p>
Full article ">Figure 4
<p>Pain (<b>B</b>), stiffness (<b>C</b>,<b>D</b>), and physical function (<b>E</b>) (WOMAC indices and total scores in (<b>A</b>)) in OA patients after the mud therapy intervention. CMT = group of patients receiving controlled mud therapy; CMRT = group of patients receiving controlled mud therapy fortified with RosA. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, compared to the corresponding baseline values.</p>
Full article ">Figure 5
<p>Health-related quality of life in OA patients after the mud therapy intervention. Total scores (<b>A</b>), mobility (<b>B</b>), self-care (<b>C</b>), usual activities (<b>D</b>), pain/discomfort (<b>E</b>), and anxiety/depression (<b>F</b>) dimensions of the EuroQol-5D questionnaire are represented in the corresponding histograms. CMT = group of patients receiving controlled mud therapy; CMRT = group of patients receiving controlled mud therapy fortified with RosA. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, compared to the corresponding baseline values.</p>
Full article ">Figure 6
<p>Innate immune response in OA patients after the mud therapy intervention. Percentage of neutrophils with phagocytic (<b>A</b>) and oxygen-dependent microbicidal (<b>B</b>) capacities are represented in the corresponding histograms. CMT = group of patients receiving controlled mud therapy; CMRT = group of patients receiving controlled mud therapy fortified with RosA. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001, compared to the corresponding baseline values. <sup>###</sup> <span class="html-italic">p</span> &lt; 0.001 (two-way ANOVA).</p>
Full article ">
23 pages, 10599 KiB  
Article
Microbubble-Protected Oncolytic Virotherapy Targeted by Sonoporation Induces Tumor Necrosis and T-Lymphocyte Infiltration in Humanized Mice Bearing Triple-Negative Breast Cancer
by Juliana Sitta, Flavia De Carlo, Imani Kirven, John H. Tackett, Patrice Penfornis, George Clement Dobbins, Mallory Barbier, Luis Del Valle, Clayton T. Larsen, Ernest G. Schutt, Rhodemann Li, Candace M. Howard and Pier Paolo Claudio
Int. J. Mol. Sci. 2024, 25(24), 13697; https://doi.org/10.3390/ijms252413697 - 21 Dec 2024
Viewed by 333
Abstract
Oncolytic virotherapy has shown great promise in mediating targeted tumor destruction through tumor-selective replication and induction of anti-tumor immunity; however, obstacles remain for virus candidates to reach the clinic. These include avoiding neutralizing antibodies, preventing stimulation of the adaptive immune response during intravenous [...] Read more.
Oncolytic virotherapy has shown great promise in mediating targeted tumor destruction through tumor-selective replication and induction of anti-tumor immunity; however, obstacles remain for virus candidates to reach the clinic. These include avoiding neutralizing antibodies, preventing stimulation of the adaptive immune response during intravenous administration, and inducing sufficient apoptosis and immune activation so that the body’s defense can work to eradicate systemic disease. We have developed a co-formulation of oncolytic viruses (OVs) with Imagent® lipid-encapsulated, perfluorocarbon microbubbles (MBs) to protect the OVs from the innate and adaptive immune system. Once inside the MB, the viral particles become acoustically active such that external ultrasound can target the delivery of the virus locally within the tumor. Humanized NSG female mice (Hu-CD34+ NSG-SGM3) engrafted in their flanks with MDA-MB-231-Luc triple-negative breast cancer (TNBC) cells were transduced with MB/OVs, with or without adjuvant Pembrolizumab treatment, and tumor sizes and tumor necrosis were assessed. The presence of CD8+ (cytotoxic T-cells), CD4+ (helper T-cells), and CD25+ (Tregs) tumor-infiltrating lymphocytes (TILs) was quantified in the tumor samples by immunohistochemistry. In an in vivo model of humanized mice engrafted with a human immune system, we observed significantly greater tumor necrosis and smaller tumor mass in human TNBC xenografts systemically treated with MB/OV complexes in the presence or absence of pembrolizumab adjuvant treatment, compared to controls. Additionally, we observed a low ratio of CD4+/CD8+ TILs and a high ratio of CD8+/CD25+ TILs in the MDA-MB-231 xenografts treated with MB/OVs complexes with or without pembrolizumab adjuvant treatment, compared to controls. Our study demonstrated the feasibility of using MBs to target OVs to TNBC through diagnostic ultrasound, which decreased tumor mass by increasing tumor necrosis and stimulated a local and systemic antitumoral immune response by increasing intratumoral CD8+ T-cytotoxic lymphocyte infiltration and decreasing CD25+ Treg cells. Full article
(This article belongs to the Special Issue Molecular Research in Triple-Negative Breast Cancer)
Show Figures

Figure 1

Figure 1
<p>Study treatment schedule. Treatments started when tumors were 150 mm<sup>3</sup>. Created with <a href="http://Biorender.com" target="_blank">Biorender.com</a>.</p>
Full article ">Figure 2
<p>Detection of Adenovirus-5 immunohistochemistry. Adenovirus-5 Hexon protein expression was negative in the controls (MBs and Pembrolizumab groups). Adenovirus-5 Hexon expression significantly increased in the OV group, with a 50–75% rate in infected cells in the IV groups. The magnification of the upper panels is 100×, and the lower panels is 200×.</p>
Full article ">Figure 3
<p>Tumor measurements by caliper and by IVIS.</p>
Full article ">Figure 4
<p>Hematoxylin and eosin staining of tumor xenografts (right flank). A montage of the treated tumor is shown on the upper panels, where the areas of necrosis are evident; lower panels show a low magnification view of the tumors in which the different sizes of the necrotic areas can be observed (original magnification 100×).</p>
Full article ">Figure 5
<p>Detection of tumor-infiltrating CD4<sup>+</sup> cells by immunohistochemistry. CD4-positive immune cells are few in the control (saline) group and sporadic in the Pembrolizumab-treated group, but their numbers significantly increase in the OV groups and dramatically increase in the MB groups. The upper panel magnification is 100×, and the lower panel magnification is 200×.</p>
Full article ">Figure 6
<p>Immunohistochemical detection of CD8<sup>+</sup> T-cells in tumors. CD8<sup>+</sup> T cell tumor infiltrates (TILs) are few in the control (saline) group, and sporadic in the Pembrolizumab treated group; however, their expression significantly increases in both OV groups, and their number is dramatically increased in both MB groups. The magnification of the upper panels is 100×, and the magnification of the lower panels is 200×.</p>
Full article ">Figure 7
<p>Immunohistochemistry for IL-R2 alpha (CD25) in tumors. CD25<sup>+</sup> cells are regularly distributed in the control MB group and are abundant in the Pembrolizumab group, mainly located within or surrounding areas of necrosis; however, their number plummeted in both MB groups, coinciding with these tumors having significantly smaller areas of necrosis. The magnification of the upper panels is 100×, and the magnification of the lower panels is 200×.</p>
Full article ">
31 pages, 1122 KiB  
Review
Therapeutic Significance of NLRP3 Inflammasome in Cancer: Friend or Foe?
by Aliea M. Jalali, Kenyon J. Mitchell, Christian Pompoco, Sudeep Poludasu, Sabrina Tran and Kota V. Ramana
Int. J. Mol. Sci. 2024, 25(24), 13689; https://doi.org/10.3390/ijms252413689 - 21 Dec 2024
Viewed by 417
Abstract
Besides various infectious and inflammatory complications, recent studies also indicated the significance of NLRP3 inflammasome in cancer progression and therapy. NLRP3-mediated immune response and pyroptosis could be helpful or harmful in the progression of cancer, and also depend on the nature of the [...] Read more.
Besides various infectious and inflammatory complications, recent studies also indicated the significance of NLRP3 inflammasome in cancer progression and therapy. NLRP3-mediated immune response and pyroptosis could be helpful or harmful in the progression of cancer, and also depend on the nature of the tumor microenvironment. The activation of NLRP3 inflammasome could increase immune surveillance and the efficacy of immunotherapy. It can also lead to the removal of tumor cells by the recruitment of phagocytic macrophages, T-lymphocytes, and other immune cells to the tumor site. On the other hand, NLRP3 activation can also be harmful, as chronic inflammation driven by NLRP3 supports tumor progression by creating an environment that facilitates cancer cell proliferation, migration, invasion, and metastasis. The release of pro-inflammatory cytokines such as IL-1β and IL-18 can promote tumor growth and angiogenesis, while sustained inflammation may lead to immune suppression, hindering effective anti-tumor responses. In this review article, we discuss the role of NLRP3 inflammasome-mediated inflammatory response in the pathophysiology of various cancer types; understanding this role is essential for the development of innovative therapeutic strategies for cancer growth and spread. Full article
(This article belongs to the Section Molecular Pathology, Diagnostics, and Therapeutics)
Show Figures

Figure 1

Figure 1
<p>Canonical and non-canonical activation of NLRP3 inflammasome. The canonical pathway involves the activation of NLRP3 inflammasomes through signals such as mitochondrial ROS, calcium influx, and potassium efflux, leading to NF-κB activation and the production of pro-inflammatory cytokines (IL-1β and IL-18). This pathway ultimately activates caspase-1, resulting in cytokine release and pyroptosis. The non-canonical pathway involving LPS from Gram-negative bacteria triggers caspase-11, which indirectly activates NLRP3, leading to similar inflammasome responses, cytokine production, and pyroptosis. Both pathways generally play critical roles in innate immunity and inflammation.</p>
Full article ">Figure 2
<p>Significance of NLRP3 inflammasome in the melanoma progression. Various factors such as UV radiation, tumor microenvironment, and melanoma cells contribute to oxidative stress and cause DNA damage, immune cell activation, and cytokine release. Oxidative stress, in turn, triggers reactive oxygen species (ROS), mitochondrial DNA damage, potassium efflux, and NF-κB activation, which influence the activation of the NLRP3 inflammasome. NLRP3 activation promotes melanoma progression, metastasis, immune evasion, and therapy resistance. In contrast, inhibition of NLRP3 could enhance immunotherapy, inhibit tumor growth, and reduce metastasis.</p>
Full article ">Figure 3
<p>Significance of NLRP3 inflammasome in leukemias. Oxidative stress and mitochondrial dysfunction in leukemia cells could activate NLRP3 inflammasomes through mitochondrial ROS, potassium efflux, and NF-κB signaling pathways. NLRP3 activation leads to the generation of active IL-1β and IL-18 cytokines, and could cause pyroptosis. Further, NLRP3 activation plays various roles in different leukemias. For example, in Acute Myeloid Leukemia (AML), NLRP3 promotes immune evasion and survival, while inhibition reduces the disease burden. In Chronic Myeloid Leukemia (CML), NLRP3 is linked to KRAS mutations and therapy resistance. In Acute Lymphoblastic Leukemia (ALL), NLRP3 activation is correlated with glucocorticoid resistance, and in Chronic Lymphocytic Leukemia (CLL), P2X7R overexpression leads to increased NLRP3.</p>
Full article ">Figure 4
<p>Role of NLRP3 inflammasome in breast cancer growth and spread. Several factors such as reactive oxygen species (ROS)-induced mitochondrial damage, BRCA1-associated genetic mutations causing mitochondrial dysfunction, extracellular ATP leading to P2X7R overexpression, and inflammatory cytokines that activate NF-κB-mediated inflammasome components could lead to activation of NLRP3 inflammasome. NLRP3-mediated release of IL-1β and IL-18 promotes cancer cell proliferation, survival, migration, immune evasion, and resistance to therapy. Further, the outcomes also include increased tumor growth, metastasis, compromised immune surveillance, and drug resistance.</p>
Full article ">Figure 5
<p>Significance of NLRP3 inflammasome activation in cigarette smoke and COPD-induced lung cancer development. Cigarette smoke leads to reactive oxygen species (ROS), mitochondrial damage, and tissue dysfunction, activating the NLRP3 inflammasome. The release of pro-inflammatory cytokines IL-1β and IL-18 contributes to prolonged inflammation. COPD-induced inflammation and immune cell recruitment, such as macrophages, further amplify this process. The persistent inflammation and oxidative stress promote DNA damage, genetic mutations, and genomic instability, ultimately leading to lung carcinogenesis.</p>
Full article ">Figure 6
<p>Role of NLRP3 inflammasome activation in promoting colon cancer development. During inflammatory bowel disease (IBD), such as Crohn’s disease and ulcerative colitis, the inflammation of the gut lining activates immune cells, causing oxidative stress, mitochondrial damage, and the release of DAMPs. Gut microbiome imbalance (dysbiosis) leads to pathogenic bacterial growth and loss of gut-barrier integrity, allowing pathogen and toxin leakage, further driving oxidative stress. These pathways cause NLRP3 activation and trigger IL-1β and IL-18 release. Increased inflammasome response results in immune system imbalance, epithelial-barrier dysfunction, and neoplasia initiation, ultimately contributing to colon cancer growth and metastasis.</p>
Full article ">
20 pages, 328 KiB  
Review
Therapeutic Potential of Nutritional Aryl Hydrocarbon Receptor Ligands in Gut-Related Inflammation and Diseases
by Fu-Chen Huang
Biomedicines 2024, 12(12), 2912; https://doi.org/10.3390/biomedicines12122912 - 20 Dec 2024
Viewed by 229
Abstract
A solid scientific foundation is required to build the concept of personalized nutrition developed to promote health and a vision of disease prevention. Growing evidence indicates that nutrition can modulate the immune system through metabolites, which are either generated via microbiota metabolism or [...] Read more.
A solid scientific foundation is required to build the concept of personalized nutrition developed to promote health and a vision of disease prevention. Growing evidence indicates that nutrition can modulate the immune system through metabolites, which are either generated via microbiota metabolism or host digestion. The aryl hydrocarbon receptor (AhR) plays a crucial role in regulating immune responses, particularly in the gut, and has emerged as a key modulator of gut-mediated inflammation and related diseases. AhR is a ligand-activated transcription factor that responds to environmental, dietary, and microbial-derived signals, influencing immune balance and maintaining intestinal homeostasis. Nutritional AhR ligands play a significant role in modulating intestinal immunity and the function of mucosal immune cells, thereby exerting clinical effects on colitis and innate immunity. Additionally, they have the capacity to orchestrate autophagy, phagocytic cell function, and intestinal epithelial tight junctions. Therapeutic strategies aimed at enhancing AhR activity, restoring gut integrity, and optimizing immune responses hold promise as avenues for future research and potential treatments for critically ill patients. Full article
(This article belongs to the Special Issue Feature Reviews in Gastrointestinal Diseases)
24 pages, 1561 KiB  
Review
Association Between Diabetes Mellitus–Tuberculosis and the Generation of Drug Resistance
by Axhell Aleid Cornejo-Báez, Roberto Zenteno-Cuevas and Julieta Luna-Herrera
Microorganisms 2024, 12(12), 2649; https://doi.org/10.3390/microorganisms12122649 - 20 Dec 2024
Viewed by 616
Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains one of the leading infectious causes of death globally, with drug resistance presenting a significant challenge to control efforts. The interplay between type 2 diabetes mellitus (T2DM) and TB introduces additional complexity, as [...] Read more.
Tuberculosis (TB), caused by Mycobacterium tuberculosis (Mtb), remains one of the leading infectious causes of death globally, with drug resistance presenting a significant challenge to control efforts. The interplay between type 2 diabetes mellitus (T2DM) and TB introduces additional complexity, as T2DM triples the risk of active TB and exacerbates drug resistance development. This review explores how T2DM-induced metabolic and immune dysregulation fosters the survival of Mtb, promoting persistence and the emergence of multidrug-resistant strains. Mechanisms such as efflux pump activation and the subtherapeutic levels of isoniazid and rifampicin in T2DM patients are highlighted as key contributors to resistance. We discuss the dual syndemics of T2DM–TB, emphasizing the role of glycemic control and innovative therapeutic strategies, including efflux pump inhibitors and host-directed therapies like metformin. This review underscores the need for integrated diagnostic, treatment, and management approaches to address the global impact of T2DM–TB comorbidity and drug resistance. Full article
(This article belongs to the Special Issue Prevention, Treatment and Diagnosis of Tuberculosis, 2nd Edition)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>The image presents three cases: one of a person with T2DM, another with TB, and a third with both T2DM–TB simultaneously. This figure illustrates how the immune response is altered in each condition, showing changes in the concentration of immune response cells and cytokines. These changes are represented with blue arrows for increases and red arrows for decreases. Additionally, it highlights that the presence of the T2DM–TB comorbidity promotes the development of DR, which worsens the clinical condition of patients.</p>
Full article ">
35 pages, 1935 KiB  
Review
Vaccination as a Promising Approach in Cardiovascular Risk Mitigation: Are We Ready to Embrace a Vaccine Strategy?
by Georgios Tsioulos, Natalia G. Vallianou, Alexandros Skourtis, Maria Dalamaga, Evangelia Kotsi, Sofia Kargioti, Nikolaos Adamidis, Irene Karampela, Iordanis Mourouzis and Dimitris Kounatidis
Biomolecules 2024, 14(12), 1637; https://doi.org/10.3390/biom14121637 - 20 Dec 2024
Viewed by 423
Abstract
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and [...] Read more.
Cardiovascular disease (CVD) remains a leading global health concern, with atherosclerosis being its principal cause. Standard CVD treatments primarily focus on mitigating cardiovascular (CV) risk factors through lifestyle changes and cholesterol-lowering therapies. As atherosclerosis is marked by chronic arterial inflammation, the innate and adaptive immune systems play vital roles in its progression, either exacerbating or alleviating disease development. This intricate interplay positions the immune system as a compelling therapeutic target. Consequently, immunomodulatory strategies have gained increasing attention, though none have yet reached widespread clinical adoption. Safety concerns, particularly the suppression of host immune defenses, remain a significant barrier to the clinical application of anti-inflammatory therapies. Recent decades have revealed the significant role of adaptive immune responses to plaque-associated autoantigens in atherogenesis, opening new perspectives for targeted immunological interventions. Preclinical models indicate that vaccines targeting specific atherosclerosis-related autoantigens can slow disease progression while preserving systemic immune function. In this context, numerous experimental studies have advanced the understanding of vaccine development by exploring diverse targeting pathways. Key strategies include passive immunization using naturally occurring immunoglobulin G (IgG) antibodies and active immunization targeting low-density lipoprotein cholesterol (LDL-C) and apolipoproteins, such as apolipoprotein B100 (ApoB100) and apolipoprotein CIII (ApoCIII). Other approaches involve vaccine formulations aimed at proteins that regulate lipoprotein metabolism, including proprotein convertase subtilisin/kexin type 9 (PCSK9), cholesteryl ester transfer protein (CETP), and angiopoietin-like protein 3 (ANGPTL3). Furthermore, the literature highlights the potential for developing non-lipid-related vaccines, with key targets including heat shock proteins (HSPs), interleukins (ILs), angiotensin III (Ang III), and a disintegrin and metalloproteinase with thrombospondin motifs 7 (ADAMTS-7). However, translating these promising findings into safe and effective clinical therapies presents substantial challenges. This review provides a critical evaluation of current anti-atherosclerotic vaccination strategies, examines their proposed mechanisms of action, and discusses key challenges that need to be overcome to enable clinical translation. Full article
Show Figures

Figure 1

Figure 1
<p>Schematic representation of adaptive immune cell involvement in the progression of atherosclerosis. In early atherosclerosis stages, Tregs accumulate in arterial walls, interacting with APCs like dendritic cells and macrophages, releasing anti-inflammatory cytokines (IL-10, TGF-β) to maintain immune tolerance and reduce inflammation. Circulating T cells also migrate to plaques, attracted by chemotactic signals (CCL5, CXCL16) from platelets and immune cells. As the disease progresses, APCs present atherosclerosis-related antigens to naïve T cells in regional lymph nodes, driving T cell polarization into pro-inflammatory Th cells and CTLs. These activated T cells return to plaques, exacerbating local inflammation. In advanced stages, Th1 cells dominate, secreting IFN-γ and TNF-α to promote plaque growth and instability. B cells modulate disease progression: B1 cells secrete protective IgM antibodies, while B2 cells release IgG antibodies, contributing to atherosclerosis progression. Overall, the adaptive immune response shifts from protective to pathogenic, driving plaque progression and vascular damage [<a href="#B14-biomolecules-14-01637" class="html-bibr">14</a>,<a href="#B15-biomolecules-14-01637" class="html-bibr">15</a>,<a href="#B16-biomolecules-14-01637" class="html-bibr">16</a>,<a href="#B17-biomolecules-14-01637" class="html-bibr">17</a>,<a href="#B18-biomolecules-14-01637" class="html-bibr">18</a>,<a href="#B19-biomolecules-14-01637" class="html-bibr">19</a>,<a href="#B20-biomolecules-14-01637" class="html-bibr">20</a>,<a href="#B21-biomolecules-14-01637" class="html-bibr">21</a>,<a href="#B22-biomolecules-14-01637" class="html-bibr">22</a>,<a href="#B23-biomolecules-14-01637" class="html-bibr">23</a>,<a href="#B24-biomolecules-14-01637" class="html-bibr">24</a>,<a href="#B25-biomolecules-14-01637" class="html-bibr">25</a>,<a href="#B26-biomolecules-14-01637" class="html-bibr">26</a>,<a href="#B27-biomolecules-14-01637" class="html-bibr">27</a>,<a href="#B28-biomolecules-14-01637" class="html-bibr">28</a>,<a href="#B29-biomolecules-14-01637" class="html-bibr">29</a>,<a href="#B30-biomolecules-14-01637" class="html-bibr">30</a>,<a href="#B31-biomolecules-14-01637" class="html-bibr">31</a>,<a href="#B32-biomolecules-14-01637" class="html-bibr">32</a>,<a href="#B33-biomolecules-14-01637" class="html-bibr">33</a>,<a href="#B34-biomolecules-14-01637" class="html-bibr">34</a>,<a href="#B35-biomolecules-14-01637" class="html-bibr">35</a>,<a href="#B36-biomolecules-14-01637" class="html-bibr">36</a>,<a href="#B37-biomolecules-14-01637" class="html-bibr">37</a>,<a href="#B38-biomolecules-14-01637" class="html-bibr">38</a>,<a href="#B39-biomolecules-14-01637" class="html-bibr">39</a>,<a href="#B40-biomolecules-14-01637" class="html-bibr">40</a>,<a href="#B41-biomolecules-14-01637" class="html-bibr">41</a>]. Abbreviations: APCs: antigen-presenting cells; B1 cells: B cell subtype 1; B2 cells: B cell subtype 2; CCL5: chemokine (C-C motif) ligand 5; CTLs: cytotoxic T lymphocytes; CXCL16: chemokine (C-X-C motif) ligand 16; DCs: dendritic cells; IFN-γ: interferon gamma; IgG: immunoglobulin G; IgM: immunoglobulin M; IL-10: interleukin-10; Mfs: macrophages; MHC: major histocompatibility complex; T cells: T lymphocytes; TGF-β: transforming growth factor beta; Th cells: T helper cells; TNF: tumor necrosis factor; Tregs: regulatory T cells. Created in BioRender. Kounatidis, D. (2024) <a href="https://BioRender.com/x00b831" target="_blank">https://BioRender.com/x00b831</a>, assessed on 24 November 2024.</p>
Full article ">Figure 2
<p>Experimental vaccine models targeting lipids to reduce inflammation in atherosclerosis. This figure depicts four distinct experimental vaccine strategies designed to mitigate the inflammation and progression of atherosclerosis by modulating immune responses. (<b>A</b>) OxLDL-C-pulsed dendritic cell transfer: LDLr−/− mice were treated with mature DCs pulsed with oxLDL-C before atherosclerosis induction via Western-type diet feeding. This approach promoted oxLDL-C-specific T cells and IgG production, diminishing foam cell recruitment and inflammation [<a href="#B84-biomolecules-14-01637" class="html-bibr">84</a>]. (<b>B</b>) PAM nanoparticle vaccine: PAM nanoparticles were used to deliver the p210 peptide in ApoE−/− mice models. This vaccine suppressed CD4<sup>+</sup> and CD8<sup>+</sup> effector T cells and shifted macrophage phenotypes, collectively reducing atherosclerotic burden and showing translational potential [<a href="#B99-biomolecules-14-01637" class="html-bibr">99</a>]. (<b>C</b>) Nanoliposome-based PCSK9-TP vaccine: A nanoliposome vaccine conjugated with PCSK9 and tetanus-derived peptides (IFPT peptide) was tested in atherosclerotic mice. The vaccine stimulated anti-inflammatory CD4<sup>+</sup> Th2 cells and IL-4 secretion, promoting atheroprotective immune responses [<a href="#B121-biomolecules-14-01637" class="html-bibr">121</a>]. (<b>D</b>) Oral TT/CETP vaccine in rabbits: A combined oral vaccine targeting TT and CETP upregulated anti-inflammatory cytokines IL-10 and TGF-β while suppressing pro-inflammatory cytokines TNF-α and IFN-γ [<a href="#B133-biomolecules-14-01637" class="html-bibr">133</a>]. Abbreviations: ApoE: apolipoprotein E; CETP: cholesteryl ester transfer protein; DC: dendritic cell; IFN-γ: interferon-gamma; IL: interleukin; LDLr: low-density lipoprotein receptor; oxLDL-C: oxidized low-density lipoprotein cholesterol; PAM: poly(amino acid)-based; PCSK9: proprotein convertase subtilisin/kexin type 9; TGF-β: transforming growth factor-beta; Th2: T-helper type 2; TNF-α: tumor necrosis factor-alpha; TP: tetanus peptide; TT: tetanus toxoid. Created in BioRender. Kounatidis, D. (2024) <a href="https://BioRender.com/f60u021" target="_blank">https://BioRender.com/f60u021</a>, assessed on 24 November 2024.</p>
Full article ">Figure 3
<p>Primary vaccination targets in atherosclerosis. Abbreviations: ADAMTS-7: A disintegrin and metalloproteinase with thrombospondin motifs 7; ANGII: angiotensin II; ANGPTL3: angiopoietin-like protein 3; ApoB100: apolipoprotein B100; ApoCIII: apolipoprotein CIII; CETP: cholesteryl ester transfer protein; HSP: heat shock protein; IgG: immunoglobulin G; LDL-C: low-density lipoprotein cholesterol; PCSK9: proprotein convertase subtilisin/kexin type 9. Created in BioRender. Kounatidis, D. (2024) <a href="https://BioRender.com/f98n410" target="_blank">https://BioRender.com/f98n410</a>, assessed on 24 November 2024.</p>
Full article ">
20 pages, 5468 KiB  
Article
Mucosal Bacterial Immunotherapy Attenuates the Development of Experimental Colitis by Reducing Inflammation Through the Regulation of Myeloid Cells
by Eva Jiménez, Alberto Vázquez, Sara González, Rosa Sacedón, Lidia M. Fernández-Sevilla, Alberto Varas, Jose L. Subiza, Jaris Valencia and Ángeles Vicente
Int. J. Mol. Sci. 2024, 25(24), 13629; https://doi.org/10.3390/ijms252413629 - 20 Dec 2024
Viewed by 557
Abstract
Ulcerative colitis is a chronic relapsing–remitting and potentially progressive form of inflammatory bowel disease in which there is extensive inflammation and mucosal damage in the colon and rectum as a result of an abnormal immune response. MV130 is a mucosal-trained immunity-based vaccine used [...] Read more.
Ulcerative colitis is a chronic relapsing–remitting and potentially progressive form of inflammatory bowel disease in which there is extensive inflammation and mucosal damage in the colon and rectum as a result of an abnormal immune response. MV130 is a mucosal-trained immunity-based vaccine used to prevent respiratory tract infections in various clinical settings. Additionally, MV130 may induce innate immune cells that acquire anti-inflammatory properties and promote tolerance, which could have important implications for chronic inflammatory diseases such as ulcerative colitis. This work demonstrated that the prophylactic administration of MV130 substantially mitigated colitis in a mouse model of acute colitis induced by dextran sulphate sodium. MV130 downregulated systemic and local inflammatory responses, maintained the integrity of the intestinal barrier by preserving the enterocyte layer and goblet cells, and reduced the oedema and fibrosis characteristic of the disease. Mechanistically, MV130 significantly reduced the infiltration of neutrophils and pro-inflammatory macrophages in the intestinal wall of the diseased animals and favoured the appearance of M2-polarised macrophages. These results suggest that MV130 might have therapeutic potential for the treatment of ulcerative colitis, reducing the risk of relapse and the progression of disease. Full article
(This article belongs to the Special Issue Inflammatory Bowel Disease: Molecular Insights)
Show Figures

Figure 1

Figure 1
<p>Beneficial effect of prophylactic MV130 treatment on the development of acute colitis. (<b>A</b>) Schematic diagram of study design. Briefly, after 21 days of prophylactic MV130 treatment or vehicle, acute colitis was induced in the animals by treatment with 2.5% DSS for 7 days. (<b>B</b>) The disease activity index (DAI) was calculated as specified in the Materials and Methods and the mean value ± SEM for each day is plotted in the graph. Animals fed fresh water were used as controls. (<b>C</b>) The mean colon length ± SEM of 4 animals per group is shown. (<b>D</b>) Representative macroscopic image of the colon from one animal in each group. Graph shows the mean values ± SEM of the different groups of animals. Significance is indicated with respect to CTRL (*) or CTRL-DSS (#) (* <span class="html-italic">p</span> &lt; 0.05; **** <span class="html-italic">p</span> ≤ 0.0001; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01)—analysed by Kruskal–Wallis test for DAI and one-way ANOVA for colon length).</p>
Full article ">Figure 2
<p>Systemic effects of MV130 in colitis-induced animals. (<b>A</b>–<b>J</b>) Mice were treated as described in <a href="#ijms-25-13629-f001" class="html-fig">Figure 1</a>A, and samples were collected on day 4 and 7 after colitis induction. (<b>A</b>) Serum levels of several cytokines in the different groups of mice 7 days after DSS treatment (n = 4 per group) were measured by CBA. (<b>B</b>) Data represent TNFα/IL-10 ratio production in each experimental group. (<b>C</b>) Splenocytes were labelled with CFSE and stimulated with ConA + PMA for four days, as described in the Material and Methods. The percentage of proliferation of splenocytes from mice of different experimental groups is shown (n = 4 per group). Flow cytometry analysis of spleen (<b>D</b>–<b>G</b>) and mesenteric lymph node immune populations (<b>H</b>–<b>J</b>). (<b>D</b>) Representative dot plots of the Treg lymphocyte population (CD4<sup>+</sup>CD25<sup>+</sup>FoxP3<sup>+</sup>) in the spleen after four days of stimulation with ConA and PMA in CTRL-DSS and MV130-DSS mice (n = 4 per group). Absolute numbers of neutrophils (<b>E</b>,<b>H</b>) and monocytes (<b>F</b>,<b>I</b>) recovered from the spleen and MLN of mice on days 4 and 7 after colitis induction (n = 3–4 per group). (<b>G</b>,<b>J</b>) Representative dot plot of myeloid populations on day 7 after DSS induction. Neutrophils (Ly6G<sup>hi</sup>Ly6C<sup>lo/−</sup>) and monocyte/macrophage populations (Ly6G<sup>−</sup>Ly6C<sup>+</sup>) were gated within CD45<sup>+</sup> CD11b<sup>+</sup>. (<b>K</b>) Flow cytometry analysis of human PBMCs after culture with MV130-CFSE for 24 h. Dot plots show the percentage of uptake of CFSE-MV130 by lymphocytes, monocytes, and granulocytes. Data are representative of 3 independent experiments. (<b>L</b>) mRNA expression of several cytokines was studied in monocytes treated with MV130 for 24 h by RT-qPCR. The graphs show the mean values ± SEM and the significance is indicated with respect to CTRL (*) or CTRL-DSS (#). * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001; # <span class="html-italic">p</span> &lt; 0.05—analysed by (<b>A</b>,<b>B</b>,<b>E</b>,<b>F</b>,<b>H</b>,<b>I</b>) Kruskal–Wallis, (<b>C</b>) one-way ANOVA, and (<b>L</b>) Mann–Whitney test.</p>
Full article ">Figure 3
<p>MV130 treatment prevents colitis-induced tissue damage. (<b>A</b>) Haematoxylin and eosin staining was performed, and the areas of the different histological regions were measured. Graphs show the mean values ± SEM of the different groups of animals. Representative images of colon sections from each group of animals: (<b>B</b>) haematoxylin and eosin (scale bar: left column, 100 μm; right column, 50 μm; insert, 10 μm); (<b>C</b>) PAS–haematoxylin (scale bar, 100 μm); and (<b>D</b>) Masson’s trichrome (scale bar, 100 μm). (<b>E</b>) Percentage of area with loss of crypt cytoarchitecture in the mucosa. (<b>F</b>) Percentage of injured epithelium. (<b>G</b>) Quantification of goblet cells in the mucosa using PAS staining. Graphs represent mean ± SEM. The significance is indicated with respect to CTRL (*) or CTRL-DSS (#) (** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; # <span class="html-italic">p</span> &lt; 0.05; ## <span class="html-italic">p</span> &lt; 0.01—analysed by one-way ANOVA test).</p>
Full article ">Figure 4
<p>MV130 treatment reduces innate immune cell infiltration and modulates the macrophage phenotype. Colon cryosections from the different experimental groups of mice sacrificed on day 7 were immunostained for CD45 (green) and for (<b>A</b>) CD68 (red) or (<b>B</b>) CD206 (red). The isotype controls are also shown. Hoechst was used for nuclear counterstaining. Scale bars represent 100 mm. Images are representative of four animals per group. Measurement of (<b>C</b>) CD68<sup>+</sup> and (<b>D</b>) CD206<sup>+</sup> macrophages in the colonic mucosa and submucosa of mice from the different groups was performed. Relative CD68 or CD206 expression was calculated by dividing all individual data by the mean expression in the CTRL group of animals. Results represent the mean ± SEM of four different animals, and the significance is indicated with respect to CTRL-DSS (# <span class="html-italic">p</span> &lt; 0.05; ## <span class="html-italic">p</span> &lt; 0.01—analysed by Student’s <span class="html-italic">t</span> test).</p>
Full article ">Figure 5
<p>MV130 induces M2-like macrophage differentiation. Human CD14<sup>+</sup> monocytes were cultured for 6 days with GM-CSF to induce M1 macrophages or with M-CSF to induce M2 macrophages. MV130 treatment was added at the beginning of the culture. (<b>A</b>) Dot plots show the percentage of M2-like CD14<sup>+</sup>CD163<sup>+</sup> cells present in the different culture conditions (representative of 4 independent experiments). (<b>B</b>) Percentage of macrophage viability was measured by flow cytometry. Annexin V<sup>−</sup> PI<sup>−</sup> cells were considered as viable cells. (<b>C</b>–<b>E</b>) After 6 days of culture, supernatants were analysed for different immunomodulatory factors produced by macrophages. (<b>C</b>) Data represent TNFα/IL-10 ratio production, (<b>D</b>) IL-4, and (<b>E</b>) CXCL10 protein secretion by macrophages under the different experimental conditions. (<b>F</b>) <span class="html-italic">IDO1</span> mRNA expression levels quantified in macrophages under the different experimental conditions at day 6 of culture. (<b>B</b>–<b>F</b>) Data are mean ± SEM of four independent experiments. Significance is indicated relative to M1 control (* <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01—analysed by Kruskal–Wallis test).</p>
Full article ">Figure 6
<p>MV130 reduces the recruitment of neutrophils and tissue damage mediators in ulcerative colitis. (<b>A</b>) Immunodetection of CD45 (green) and CD15 (neutrophils, red) in colon cryosection 7 days after DSS induction. The isotype controls are also shown. Hoechst was used for nuclear counterstaining. Scale bars represent 100 μm. Images are representative of four animals per group. (<b>B</b>) Measurement of CD15<sup>+</sup> neutrophils in the colonic mucosa and submucosa of mice from the different groups was performed (dotted line). Relative CD15 expression was calculated by dividing all individual data by the mean expression in the CTRL group. (<b>C</b>) Relative MPO activity in colonic protein extracts on days 4 and 7 after colitis induction. (<b>D</b>) Protein levels in colonic mucosa measured by flow cytometry. (<b>B</b>–<b>D</b>) Results represent the mean ± SEM of four different animals per group and the significance is indicated with respect to CTRL (*) or CTRL-DSS (#). * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; **** <span class="html-italic">p</span> &lt; 0.0001; # <span class="html-italic">p</span> &lt; 0.05; ## <span class="html-italic">p</span> &lt; 0.01; ### <span class="html-italic">p</span> &lt; 0.001—analysed by (<b>B</b>) Student’s <span class="html-italic">t</span> test, (<b>C</b>) Kruskal–Wallis test, (<b>D</b>) and one-way ANOVA test.</p>
Full article ">
14 pages, 1463 KiB  
Article
Similarly to BmToll9-1, BmToll9-2 Is a Positive Regulator of the Humoral Immune Response in the Silkworm, Bombyx mori
by Jisheng Liu, Weijian Chen, Sihua Chen, Shuqiang Li and Luc Swevers
Insects 2024, 15(12), 1005; https://doi.org/10.3390/insects15121005 - 19 Dec 2024
Viewed by 280
Abstract
Toll receptors play important roles in the development and innate immunity of insects. Previously, we reported the immunological function of BmToll9-2 in silkworm, Bombyx mori, larvae. In this study, we focused on the role of BmToll9-2 as a regulator in the Toll [...] Read more.
Toll receptors play important roles in the development and innate immunity of insects. Previously, we reported the immunological function of BmToll9-2 in silkworm, Bombyx mori, larvae. In this study, we focused on the role of BmToll9-2 as a regulator in the Toll signaling pathway. The expressions of most signaling genes in the Toll pathway, as well as immune effectors, were reduced after the RNAi of BmToll9-2. Coincidentally, hemolymph from BmToll9-2-silenced larvae exhibited decreased antibacterial activity in the growth of Escherichia coli, demonstrated either by growth curve or inhibitory zone experiments. The oral administration of heat-inactivated E. coli and Staphylococcus aureus following the RNAi of BmToll9-2 up-regulated the expression of most signaling genes in the Toll pathway and downstream immune effectors. The above results indicate that BmToll9-2 is positively involved in the Toll signaling pathway. As a positive regulator, BmToll9-2 is shown to be activated preferentially against E. coli and, in turn, positively modulates the humoral immune response in antibacterial activity. Full article
(This article belongs to the Collection Insect Immunity: Evolution, Genomics and Physiology)
Show Figures

Figure 1

Figure 1
<p>The relative expression of signaling genes in the Toll pathway after the RNAi of <span class="html-italic">BmToll9-2</span>. Larvae of the 5th instar were injected with dsBmToll9-2, and dsGFP served as a control. Data are represented as the means ± standard deviations of three biological replications. Asterisks indicate significant differences from dsGFP injection groups: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; and **** <span class="html-italic">p</span> &lt; 0.0001. ns, not significant.</p>
Full article ">Figure 2
<p>The relative expression of immune effector genes after the RNAi of <span class="html-italic">BmToll9-2</span>. Larvae of the 5th instar were injected with dsBmToll9-2, and dsGFP served as a control. Data are represented as the means ± standard deviations of three biological replications. Asterisks indicate significant differences from dsGFP injection groups: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; and **** <span class="html-italic">p</span> &lt; 0.0001. ns, not significant.</p>
Full article ">Figure 3
<p>Antibacterial activity assays of <span class="html-italic">B. mori</span> hemolymph against <span class="html-italic">E. coli</span> and <span class="html-italic">S. aureus</span> after RNAi of <span class="html-italic">BmToll9-2.</span> Hemolymph was collected 24 h after dsRNA injection and tested for antibacterial activity. (<b>A</b>) Bacterial growth curve experiment. (<b>B</b>) Inhibition zone experiment. dsBmToll9-2: hemolymph from larvae injected with dsBmToll9-2; dsGFP: hemolymph from larvae injected with dsGFP; H<sub>2</sub>O: sterile water; antibiotic: ampicillin. Asterisks indicate significant differences from dsGFP injection groups: * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>The relative expression of signaling genes in the Toll pathway following challenges with heat-inactivated bacteria after the RNAi of <span class="html-italic">BmToll9-2</span>. Larvae of 5th instar were injected with dsBmToll9-2 or dsGFP. Then, the larvae were fed with heat-killed (<b>A</b>) <span class="html-italic">E. coli</span> or (<b>B</b>) <span class="html-italic">S. aureus.</span> Data are represented as the means ± standard deviations of three biological replications. Asterisks indicate significant differences from dsGFP injection groups: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; and **** <span class="html-italic">p</span> &lt; 0.0001. ns, not significant.</p>
Full article ">Figure 5
<p>The relative expression of immune effector genes following challenges with heat-inactivated bacteria after the RNAi of <span class="html-italic">BmToll9-2</span>. Larvae of 5th instar were injected with dsBmToll9-2 or dsGFP. Then, the larvae were fed with heat-killed (<b>A</b>) <span class="html-italic">E. coli</span> or (<b>B</b>) <span class="html-italic">S. aureus</span>. Data are represented as the means ± standard deviations of three biological replications. Asterisks indicate significant differences from dsGFP injection groups: * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001; and **** <span class="html-italic">p</span> &lt; 0.0001. ns, not significant.</p>
Full article ">
Back to TopTop