[go: up one dir, main page]

 
 
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
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

Search Results (11,035)

Search Parameters:
Keywords = macrophage

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
12 pages, 3824 KiB  
Article
Surface Display of Cholera Toxin B Subunit Recombinant Escherichia coli Ghosts Further Enhances Resistance to Chlamydia abortus Infection in Mice
by Huaiyu Zhang, Yunhui Li, Wei Li, Zhaocai Li, Jizhang Zhou and Dewen Tong
Microorganisms 2024, 12(8), 1656; https://doi.org/10.3390/microorganisms12081656 (registering DOI) - 13 Aug 2024
Viewed by 36
Abstract
Chlamydia abortus (C. abortus) is an important zoonotic pathogen that seriously endangers the development of animal husbandry. Vaccination is the most effective approach to preventing C. abortus infection. We previously reported a recombinant Escherichia coli ghost (rECG)-based C. abortus vaccine that [...] Read more.
Chlamydia abortus (C. abortus) is an important zoonotic pathogen that seriously endangers the development of animal husbandry. Vaccination is the most effective approach to preventing C. abortus infection. We previously reported a recombinant Escherichia coli ghost (rECG)-based C. abortus vaccine that demonstrated outstanding protective efficacy. In this study, we further attempted to fuse the cholera toxin B subunit (CTB), a widely studied potent mucosal immune adjuvant, with macrophage infectivity potentiator (MIP), a candidate antigen of C. abortus, on the surface of the rECG and explore its protective effect against C. abortus infection. The MIP fusion protein was highly expressed in the rECGs, and the CTB-modified rECGs significantly induced the activation of mouse bone marrow-derived dendritic cells in vitro. Intranasal immunization with rECGs induced a Th1-biased cellular immune response. Compared to the rECGs without CTB, the CTB-modified rECGs induced higher concentrations of IgA in the serum and vaginal wash solution. Moreover, in a mouse infection model, the CTB-modified rECGs significantly improved the clearance efficiency of C. abortus and reduced the pathological damage to the uterus. This study demonstrates that incorporating CTB into rECGs significantly enhances the immunogenic potential of the rECG vaccine and can significantly enhance its protective efficacy against a C. abortus challenge. Full article
(This article belongs to the Section Medical Microbiology)
Show Figures

Figure 1

Figure 1
<p>Construction of surface display plasmid and identification of fusion protein. (<b>A</b>) Schematic diagram of recombinant plasmids pA-lpp’-MIP and pA-lpp’-MIP-CTB. Fusion protein expression was detected by Western blotting using monoclonal antibodies against MIP (<b>B</b>) and monoclonal antibodies against CTB (<b>C</b>). M: protein marker.</p>
Full article ">Figure 2
<p>Analysis of display of MIP fusion proteins on bacterial surfaces. (<b>A</b>) MIP antibody was used to incubate bacteria, followed by Alexa Fluor 594-labeled anti-mouse IgG antibody (red) for indirect immunofluorescence analysis. Bacterial genomes were labeled with DAPI (blue). (<b>B</b>) Proteinase K-treated (+PK) or untreated C43-lpp’-MIP and C43-lpp’-MIP-CTB were incubated with MIP antibodies, followed by flow cytometry detection with Alexa Fluor 594-labeled anti-mouse IgG antibodies. Final data are shown as MFI. Results are expressed as mean ± standard deviation (SD). ** <span class="html-italic">p</span> &lt; 0.01; ns, no significance.</p>
Full article ">Figure 3
<p>Effects of rECGs on BMDCs’ maturation and activation. (<b>A</b>,<b>B</b>) After BMDCs were stimulated by rECGs in vitro, the CD80 and MHC II expression frequencies of CD11c<sup>+</sup> cells were detected by flow cytometry. The final data are shown as the MFI. Content of cytokines IL-6 (<b>C</b>), IL-12p70 (<b>D</b>), and TNF-α (<b>E</b>) in BMDC culture supernatants. Results are expressed as means ± SD. * <span class="html-italic">p</span> &lt; 0.5, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001; ns, no significance.</p>
Full article ">Figure 4
<p>Humoral immune response induced by rECGs in mice. (<b>A</b>) Dynamics of MIP-specific IgG in serum samples collected 0, 14, 28, 42, and 56 days after the first immunization. Titers of MIP-specific IgG2a (<b>B</b>) and IgG1 (<b>C</b>) in serum samples 42 days after the first immunization. MIP-specific IgA titers in serum samples (<b>D</b>) and vaginal lavage fluid (<b>E</b>) 42 days after the first immunization. Results are expressed as mean ± SD. * <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; ns, no significance.</p>
Full article ">Figure 5
<p>Cellular immune response induced by rECGs in mice. (<b>A</b>) Specific splenic lymphocyte proliferation response in immunized mice, with concanavalin A (ConA)-treated group serving as a positive control. Content of IFN-γ (<b>B</b>), IL-12p70 (<b>C</b>), TNF-α (<b>D</b>), IL-4 (<b>E</b>), and IL-10 (<b>F</b>) in lymphocyte culture supernatants. Results are expressed as mean ± SD. * <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; ns, no significance.</p>
Full article ">Figure 6
<p>Protective effect of rECGs on mice infected with <span class="html-italic">C. abortus</span>. (<b>A</b>) Vaginal swabs were collected every 3 d post-challenge to enumerate the <span class="html-italic">C. abortus</span>-recoverable IFUs. The mean recoverable IFUs are expressed as log<sub>10</sub> IFU/mL ± SD. (<b>B</b>) Histopathological analysis of uterine tissue sections. Lysis and necrosis of the lamina propria are indicated with black arrows, uterine gland cavity occlusion is indicated with red arrows, and lamina propria edema is indicated with yellow arrows. (<b>C</b>) Quantitative isolation of <span class="html-italic">C. abortus</span> in uterine homogenates of mice 7 days after challenge. * <span class="html-italic">p</span> &lt; 0.05, *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">
14 pages, 5113 KiB  
Article
Extracellular Vesicle Transplantation Is Beneficial for Acute Kidney Injury
by Amankeldi A. Salybekov, Shigeaki Okamura, Takayasu Ohtake, Sumi Hidaka, Takayuki Asahara and Shuzo Kobayashi
Cells 2024, 13(16), 1335; https://doi.org/10.3390/cells13161335 - 12 Aug 2024
Viewed by 208
Abstract
Under vasculogenic conditioning, certain pro-inflammatory subsets within peripheral blood mononuclear cells (PBMCs) undergo phenotypic transformation into pro-regenerative types, such as vasculogenic endothelial progenitor cells, M2 macrophages, and regulatory T cells. These transformed cells are collectively termed regeneration-associated cells (RACs). In this study, we [...] Read more.
Under vasculogenic conditioning, certain pro-inflammatory subsets within peripheral blood mononuclear cells (PBMCs) undergo phenotypic transformation into pro-regenerative types, such as vasculogenic endothelial progenitor cells, M2 macrophages, and regulatory T cells. These transformed cells are collectively termed regeneration-associated cells (RACs). In this study, we aimed to investigate the therapeutic efficacy of RAC-derived extracellular vesicles (RACev) compared with a vehicle-treated group in the context of renal ischemia-reperfusion injury (R-IRI). Human PBMCs were cultured with defined growth factor cocktails for seven days to harvest RACs. EV quantity and size were characterized by nanoparticle tracking analysis. Notably, the systemic injection of RACev significantly decreased serum creatinine and blood urine nitrogen at day three compared to the control group. Histologically, the treatment group showed less fibrosis in the cortex and medullary areas (p < 0.04 and p < 0.01) compared to the control group. The CD31 staining confirmed enhanced capillary densities in the treatment group compared to the control group (p < 0.003). These beneficial effects were accompanied by angiogenesis, anti-fibrosis, anti-inflammation, and anti-apoptosis RACev miR delivery to ischemic injury to control inflammatory, endothelial mesenchymal transition, and hypoxia pathways. In vivo bioluminescence analysis demonstrated a preferential accumulation of RACev in the IR-injured kidney. The systemic transplantation of RACev beneficially restored kidney function by protecting from tissue fibrosis and through anti-inflammation, angiogenesis, and anti-apoptosis miR delivery to the ischemic tissue. Full article
(This article belongs to the Special Issue Extracellular Vesicles in Tissue Repair and Regeneration)
Show Figures

Figure 1

Figure 1
<p><b>Characterization of RACs.</b> (<b>A</b>) Total stem and progenitor levels increased after vasculogeneic conditioning. (<b>B</b>) The EPCs were quantitatively and (<b>C</b>,<b>D</b>) qualitatively enhanced in post-vasculogenic culture (the majority by definitive EPC expansion). (<b>E</b>,<b>F</b>) Vasculogenic conditioning dramatically accelerated M1 macrophage phenotype conversion to regenerative macrophage type 2 (<b>G</b>). The level of regulatory T cells. * <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; 0.0001 vs. the control group; Statistical significance was determined using a Mann–Whitney test. n = 10 per group. The results are presented as mean ± SEM. * <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; 0.0001.</p>
Full article ">Figure 2
<p><b>Characterization of RAC-derived extracellular vesicles.</b> (<b>A</b>) EV-specific anti-CD63 and anti-CD9 biomarker expression in RACev. (<b>B</b>) Representative transmission-electron microscopy figures showed the lipid bilayer structure in RACev. (<b>C</b>) (<b>a</b>) Quantification of one million RAC-derived EVs, (<b>b</b>) average size, and (<b>c</b>) protein amount.</p>
Full article ">Figure 3
<p><b>RACev transplantation restored kidney function.</b> (<b>A</b>) Serum creatinine level at day three significantly decreased in RACev vs. control. (<b>B</b>) Similarly, serum BUN level was dramatically diminished in the RACev transplanted group compared to the Control group. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; ns is not significant vs. the control group; statistical significance was determined using a 2-way ANOVA followed by Tukey’s multiple comparison test. The results are presented as mean ± SEM.</p>
Full article ">Figure 4
<p><b>RACev transplantation preserved renal interstitial fibrosis.</b> (<b>A</b>) Representative Masson trichrome staining depicts reduced or preserved fibrosis area in RACev-transplanted group in comparison to control groups. (<b>B</b>) Fibrosis area quantification in cortex area and (<b>C</b>) medullary area. (<b>D</b>) Anti-fibrosis miRs were significantly upregulated in RACev. (<b>E</b>) Fibrosis-related genes markedly upregulated RACev vs. control four days after the onset of R-IRI. * <span class="html-italic">p</span> &lt; 0.05; ** <span class="html-italic">p</span> &lt; 0.01; ns is not significant vs. the control group. Transcriptome analysis at day four after the onset of AKI demonstrated fibrosis-related gene upregulation in control group vs. RACev. Statistical significance was determined using a one-way ANOVA followed by Dunn’s multiple comparison test. The results are presented as mean ± SEM (n = 8–10 per group).</p>
Full article ">Figure 5
<p><b>Regulation of inflammatory and apoptosis pathways.</b> (<b>A</b>) Gene co-expression network analysis of control group revealed (<b>B</b>) inflammation, EMT, and hypoxia pathway upregulation. (<b>C</b>) RACev-transplanted group demonstrated regeneration-associated pathway upregulation. (<b>D</b>) Anti-inflammatory miRs are abundantly expressed in RACev. (<b>E</b>) Anti-apoptotic and proliferation-associated miR expression in RACev. Differentially expressed miRs were determined using a threshold of absolute values of fold change ≥ 2.</p>
Full article ">Figure 6
<p><b>Enhanced angiogenesis in infarcted tissues.</b> (<b>A</b>) Microvascular density was enhanced in ischemic injured kidney tissue in the RACev-transplanted group. (<b>B</b>) CD31 positive capillary count at day four and (<b>C</b>) day 28 after onset of R-IRI. (<b>D</b>) Angiogenic miRs, also known as angiomiRs, are markedly expressed in RACev. (<b>E</b>) Angiogenesis-related gene expression of RACev vs. control groups’ kidney tissues. ** <span class="html-italic">p</span> &lt; 0.01; ns is not significant vs. control group. Statistical significance was determined using one-way ANOVA with Dunn’s multiple comparisons test. Results are presented as the mean ± SEM (n = 8–10 per group). Differentially expressed miRs were determined using a threshold of absolute values of fold change ≥ 2.</p>
Full article ">Figure 7
<p><b>Selective accumulation of RACev.</b> (<b>A</b>) Schematic design of an in vivo study. (<b>B</b>) Systemic transplantation of labeled RACev preferentially accumulated into the ischemia-injured kidneys. (<b>C</b>,<b>D</b>) Transcriptome cell annotation and mapping showed possible RACev accumulation in the kidney.</p>
Full article ">
14 pages, 16172 KiB  
Article
Topical Application of Cha-Koji, Green Tea Leaves Fermented with Aspergillus Luchuensis var Kawachii Kitahara, Promotes Acute Cutaneous Wound Healing in Mice
by Yasuhiro Katahira, Jukito Sonoda, Miu Yamagishi, Eri Horio, Natsuki Yamaguchi, Hideaki Hasegawa, Izuru Mizoguchi and Takayuki Yoshimoto
Sci. Pharm. 2024, 92(3), 44; https://doi.org/10.3390/scipharm92030044 - 12 Aug 2024
Viewed by 375
Abstract
“Koji” is one of the most well-known probiotic microorganisms in Japan that contribute to the maintenance of human health. Although the beneficial effects of some probiotics on ulcer healing have been demonstrated, there have been no reports on the wound healing effects of [...] Read more.
“Koji” is one of the most well-known probiotic microorganisms in Japan that contribute to the maintenance of human health. Although the beneficial effects of some probiotics on ulcer healing have been demonstrated, there have been no reports on the wound healing effects of koji to date. In the present study, we investigated the effects of “cha-koji”, green tea leaves fermented with Aspergillus luchuensis, on cutaneous wound healing, using a linear incision wound mouse model. Topical application of autoclave-sterilized cha-koji suspension on the dorsal incision wound area healed the wound significantly faster and, notably, with less scarring than did the green tea or the control distilled water treatment. Further in vitro experiments revealed that the accelerated effects of cha-koji could be attributed to its increased anti-bacterial activity, enhanced epidermal cell proliferation and migration, augmented expression of the anti-inflammatory cytokine transforming growth factor-β1, reduced expression of inflammatory cytokine interleukin-6 in macrophages, and decreased endoplasmic reticulum stress. In addition, we conducted a skin sensitizing potential test, which revealed that cha-koji had no adverse effects that posed a sensitizing risk. Thus, cha-koji may have a potent therapeutic effect on cutaneous wound healing, opening up a new avenue for its clinical application as a medical aid. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Autoclave-sterilized cha-koji suspension promotes cutaneous wound healing. A cutaneous incision wound was made on the dorsal skin of the mice; coated with gauze soaked in green tea, cha-koji suspension, or distilled water; and covered with a transparent film dressing, Tegaderm, daily until day 13 (<b>A</b>,<b>B</b>). Photographs of the incision were taken over time. Representative photographs of the healing process are shown (<b>C</b>). The length of the incision wound in each photograph was measured using FIJI, and the relative length at each time point to the initial length of 10 mm was calculated (<b>D</b>). Data are shown as the mean ± SD (<span class="html-italic">n</span> = 3) and are representative of five independent experiments. <span class="html-italic">p</span>-values were determined via two-way analysis of variance with Tukey’s multiple comparison test. The red and blue asterisks marked on day 6, 8, 10, and 12 in the figure (<b>D</b>) mean that there are significant differences between cha-koji and green tea and between cha-koji and distilled water, respectively. * <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. Scale bar = 5 mm.</p>
Full article ">Figure 2
<p>Autoclave-sterilized cha-koji suspension showing anti-bacterial activity. <span class="html-italic">E. coli</span> was cultured in LB containing various concentrations of autoclave-sterilized cha-koji, green tea (0.5, 1.0, 2.0, 4.0, and 5.0%) or distilled water for 8 h at 37 °C (<b>A</b>). As a positive control, raw green tea was used. Bacterial growth was examined according to the turbidity of the LB medium, which was determined by measuring optical density at 600 nm (OD600) (<b>B</b>). Data are shown as the mean ± SD (<span class="html-italic">n</span> = 3) and are representative of three independent experiments. <span class="html-italic">p</span>-values were determined via one-way analysis of variance with Tukey’s multiple comparison test. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>Autoclave-sterilized cha-koji suspension promotes skin epidermal cell proliferation and migration. Keratinocyte PAM212 cells (<b>A</b>,<b>B</b>) or fibroblast NIH3T3 cells (<b>C</b>,<b>D</b>) were seeded in 24-well plates and cultured to semi-confluence in DMEM containing 10% FBS. The medium was then exchanged with DMEM containing 0.05% green tea or 0.05% cha-koji and 1.0% FBS. A cross-shaped scratch was made in the center of the well, and photographs were taken over time. Representative photographs are shown (<b>A</b>,<b>C</b>). The remaining wound area size was determined using FIJI, and the ratio relative to the initial wound area on day 0 was calculated (<b>B</b>,<b>D</b>). Data are shown as the mean ± SD (<span class="html-italic">n</span> = 6) and are representative of four (<b>A</b>,<b>B</b>) and three (<b>C</b>,<b>D</b>) independent experiments. <span class="html-italic">p</span>-values were determined via one-way analysis of variance with Tukey’s multiple comparison test. ** <span class="html-italic">p</span> &lt; 0.01 and *** <span class="html-italic">p</span> &lt; 0.001. Scale bar = 500 µm.</p>
Full article ">Figure 4
<p>Autoclave-sterilized cha-koji suspension promotes the expression of the anti-inflammatory cytokine TGF-β1. Macrophage RAW264.7 cells were stimulated with 0.3% cha-koji, 0.3% green tea, and distilled water for 24 and 48 h. RT-qPCR was then performed to analyze the mRNA expression of <span class="html-italic">TGF-β1</span> (<b>A</b>,<b>B</b>) and <span class="html-italic">IL-6</span> (<b>C</b>,<b>D</b>). <span class="html-italic">HPRT</span> was used as an internal control, and the relative expression of <span class="html-italic">TGF-β1</span> or <span class="html-italic">IL-6</span> to <span class="html-italic">HPRT</span> was calculated. Data are shown as the mean ± SD (<span class="html-italic">n</span> = 4−6) and are representative of three (<b>A</b>,<b>B</b>) and five (<b>C</b>,<b>D</b>) independent experiments. <span class="html-italic">p</span>-values were determined via one-way analysis of variance with Dunnett’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01.</p>
Full article ">Figure 5
<p>Autoclave-sterilized cha-koji suspension suppresses ER stress. Fibroblast NIH3T3 cells were stimulated with tunicamycin (1.0 μg/mL) in the presence or absence of 0.25% cha-koji and 0.25% green tea for 12 h. RNA was extracted, and RT-qPCR analysis was performed to examine the mRNA expression of ER stress-related factors, namely, <span class="html-italic">HSPA5</span> (<b>A</b>), <span class="html-italic">XBP1</span> (<b>B</b>), and <span class="html-italic">CHOP</span> (<b>C</b>). <span class="html-italic">HPRT</span> was used as an internal control, and its relative expression of <span class="html-italic">HPRT</span> was calculated. Data are shown as the mean ± SD (<span class="html-italic">n</span> = 3) and are representative of two independent experiments. <span class="html-italic">p</span>-values were determined via one-way analysis of variance with Dunnett’s multiple comparison test. After 48 h, photographs of each well were taken (<b>D</b>), and cell growth activity was determined by measuring the remaining viable cell area relative to the tunicamycin-untreated, distilled water-treated cell area using FIJI (<b>E</b>). Data are shown as the mean ± SD (<span class="html-italic">n</span> = 3) and are representative of three independent experiments. <span class="html-italic">p</span>-values were determined via one-way analysis of variance with Dunnett’s multiple comparison test. * <span class="html-italic">p</span> &lt; 0.05 and *** <span class="html-italic">p</span> &lt; 0.001. Scale bar = 250 µm.</p>
Full article ">Figure 6
<p>Autoclave-sterilized cha-koji suspension poses no potential skin sensitizing risk. Human monocytic THP-1 cells were stimulated with cha-koji or green tea suspension (0.25, 1.0, and 4.0%) together with the positive control, conditioned medium from rhododenol-treated melanoma SK-MEL-37 cells. After 24 h, CD86 expression in the THP-1 cells was analyzed via flow cytometry using anti-CD86 (red-shaded histogram) or a control antibody (blue-shaded histogram). Conditioned medium from the rhododenol-treated melanoma (SK-MEL-37) was used as a positive control. Representative histograms for the cell surface expression of CD86 are shown (<b>A</b>). The RFI values of each sample were calculated and compared (<b>B</b>). An RFI higher than 150% is considered to be positive for skin sensitizing potential.</p>
Full article ">
15 pages, 1073 KiB  
Review
Revisiting the Immunometabolic Basis for the Metabolic Syndrome from an Immunonutritional View
by César Jeri Apaza, Juan Francisco Cerezo, Aurora García-Tejedor, Juan Antonio Giménez-Bastida and José Moisés Laparra-Llopis
Biomedicines 2024, 12(8), 1825; https://doi.org/10.3390/biomedicines12081825 - 12 Aug 2024
Viewed by 241
Abstract
Metabolic syndrome (MetS) implies different conditions where insulin resistance constitutes a major hallmark of the disease. The disease incurs a high risk for the development of cardiovascular complications, and takes its toll in regard to the gut–liver axis (pancreas, primary liver and colorectal)-associated [...] Read more.
Metabolic syndrome (MetS) implies different conditions where insulin resistance constitutes a major hallmark of the disease. The disease incurs a high risk for the development of cardiovascular complications, and takes its toll in regard to the gut–liver axis (pancreas, primary liver and colorectal)-associated immunity. The modulation of immunometabolic responses by immunonutritional factors (IFs) has emerged as a key determinant of the gut–liver axis’ metabolic and immune health. IFs from plant seeds have shown in vitro and pre-clinical effectiveness primarily in dealing with various immunometabolic and inflammatory diseases. Only recently have immunonutritional studies established the engagement of innate intestinal immunity to effectively control immune alterations in inflamed livers preceding the major features of the MetS. However, integrative analyses and the demonstration of causality between IFs and specific gut–liver axis-associated immunometabolic imbalances for the MetS remain ill-defined in the field. Herein, a better understanding of the IFs with a significant role in the MetS, as well as within the dynamic interplay in the functional differentiation of innate immune key effectors (i.e., monocytes/macrophages), worsening or improving the disease, could be of crucial relevance. The development of an adequate intermediary phenotype of these cells can significantly contribute to maintaining the function of Tregs and innate lymphoid cells for the prevention and treatment of MetS and associated comorbidities. Full article
(This article belongs to the Special Issue Immunometabolic Determinants of Gut–Liver Axis Health)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram for the interaction of the genetic and environmental factors influencing the control of hepatic fat accumulation by macrophages. ILCs, innate lymphoid cells; FA, fatty acids.</p>
Full article ">Figure 2
<p>Schematic hypothesis to explain the potential different contribution of TLR4 to the inflammatory phenotype of macrophages in the metabolic syndrome. SETIs, serine-type protease inhibitors; LPS, bacterial lipopolysaccharide; IRF3, interferon regulatory factor 3; IFNs, interferons; IS, insulin sensitivity. Small-dotted line represents the sequence of events triggered by the activation of TLR4 by the prototypical agonist LPS (red symbols and arrows). Solid line represents the sequence of events triggered by the activation of TLR4 by SETIs (green symbols and arrows). Large-dotted line represents the influence of insulin on the NRLP3 expression.</p>
Full article ">
11 pages, 5038 KiB  
Article
The Potential of Immunotherapy for SMARCA4-Deficient Undifferentiated Uterine Sarcoma (SDUS)
by Xiaohong Yao, Ying He, Chaoxin Xiao, Ruihan Zhou, Chengjian Zhao and Wei Wang
Biomolecules 2024, 14(8), 987; https://doi.org/10.3390/biom14080987 (registering DOI) - 11 Aug 2024
Viewed by 250
Abstract
(1) Background: SMARCA4-deficient undifferentiated uterine sarcoma (SDUS) is a rare and aggressive cancer that urgently requires novel therapeutic strategies. Despite the proven efficacy of immunotherapy in various cancer types, its application in SDUS remains largely unexplored. This study aims to investigate the immune [...] Read more.
(1) Background: SMARCA4-deficient undifferentiated uterine sarcoma (SDUS) is a rare and aggressive cancer that urgently requires novel therapeutic strategies. Despite the proven efficacy of immunotherapy in various cancer types, its application in SDUS remains largely unexplored. This study aims to investigate the immune microenvironment of SDUS to evaluate the feasibility of utilizing immunotherapy. (2) Methods: Multiplex immunofluorescence (mIF) was employed to examine the immune microenvironment in two cases of SDUS in comparison to other subtypes of endometrial stromal sarcomas (ESSs). This research involved a comprehensive evaluation of immune cell infiltration, cellular interactions, and spatial organization within the tumor immune microenvironment (TiME). Statistical analysis was performed to assess differences in immune cell densities and interactions between SDUS and other ESSs. (3) Results: SDUS exhibited a significantly higher density of cytotoxic T lymphocytes (CTLs), T helper (Th) cells, B cells, and macrophages compared to other ESSs. Notable cellular interactions included Th–CTL and Th–B cell interactions, which were more prominent in SDUS. The spatial analysis revealed distinct immune niches characterized by lymphocyte aggregation and a vascular-rich environment, suggesting an active and engaged immune microenvironment in SDUS. (4) Conclusions: The results suggest that SDUS exhibits a highly immunogenic TiME, characterized by substantial lymphocyte infiltration and dynamic cellular interactions. These findings highlight the potential of immunotherapy as an effective treatment approach for SDUS. However, given the small number of samples evaluated, these conclusions should be drawn with caution. This study underscores the importance of additional investigation into immune-targeted therapies for this challenging cancer subtype, with a larger sample size to validate and expand upon these preliminary findings. Full article
Show Figures

Figure 1

Figure 1
<p>mIF defines the TiME of ESS. (<b>A</b>) A schematic diagram depicting mIF-multiplexed images from 9 patients with ESS. The images were created with FigDraw. Immune cells were identified using fluorescent dyes: CD8 (yellow), CD4 (red), CD20 (green), and CD68 (magenta), while nuclei were counterstained with DAPI, appearing as dark blue. (<b>B</b>) A schematic representation of high immune infiltration TiME characteristic of SDUS. (<b>C</b>) A schematic representation of low immune infiltration TiME commonly found in both LG-ESS and HG-SS. (<b>D1</b>) Comparison of cell densities (per mm<sup>2</sup>) for four markers (Th cell, CTL, B cell, and Mac) across various ESSs and SDUS. (<b>D2</b>) The comparative statistical analysis graph illustrates the distinctions in immune cell infiltration between ESSs with low immune cell infiltration and SDUS with high immune cell infiltration. A two-way ANOVA with multiple comparison test was used for statistical analysis by GraphPad Prism10.1.2.</p>
Full article ">Figure 2
<p>An immunofluorescent staining schematic diagram of SDUS. (<b>A</b>,<b>B</b>) The immunofluorescent staining included markers for lymphocytes (CD3, CD4, CD8, and CD20), myeloid cells (CD68 and CD14), immune checkpoint inhibitors (PD-1 and PD-L1), stromal markers (α-SMA, CD31, and FAP), and markers for cell cytotoxicity and proliferation (Ki67 and Granzyme B). (<b>C</b>–<b>G</b>) Fluorescence imaging of proliferating and exhausted cells, with a graphical representation of the proportion of proliferative cells.</p>
Full article ">Figure 3
<p>Interaction analysis of SDUS. A mIF heatmap of cell–cell interactions in patient SDUS-case2, with fluorescence images and rendering of interacting cells. Corresponding images for another patient (SDUS-case1) are provided in <a href="#app1-biomolecules-14-00987" class="html-app">Supplementary Figure S1</a>.</p>
Full article ">Figure 4
<p>A comprehensive analysis of cellular niches in the SDUS TiME. (<b>A</b>) A niche schematic diagram [<a href="#B16-biomolecules-14-00987" class="html-bibr">16</a>] (<b>B</b>,<b>C</b>) presenting heatmaps displaying the cellular composition percentages and proportional distribution of niches. (<b>D</b>,<b>E</b>) depict schematic representations of the two most prevalent niches.</p>
Full article ">
46 pages, 2259 KiB  
Review
Help or Hinder: Protein Host Factors That Impact HIV-1 Replication
by Michael Rameen Moezpoor and Mario Stevenson
Viruses 2024, 16(8), 1281; https://doi.org/10.3390/v16081281 - 10 Aug 2024
Viewed by 575
Abstract
Interactions between human immunodeficiency virus type 1 (HIV-1) and the host factors or restriction factors of its target cells determine the cell’s susceptibility to, and outcome of, infection. Factors intrinsic to the cell are involved at every step of the HIV-1 replication cycle, [...] Read more.
Interactions between human immunodeficiency virus type 1 (HIV-1) and the host factors or restriction factors of its target cells determine the cell’s susceptibility to, and outcome of, infection. Factors intrinsic to the cell are involved at every step of the HIV-1 replication cycle, contributing to productive infection and replication, or severely attenuating the chances of success. Furthermore, factors unique to certain cell types contribute to the differences in infection between these cell types. Understanding the involvement of these factors in HIV-1 infection is a key requirement for the development of anti-HIV-1 therapies. As the list of factors grows, and the dynamic interactions between these factors and the virus are elucidated, comprehensive and up-to-date summaries that recount the knowledge gathered after decades of research are beneficial to the field, displaying what is known so that researchers can build off the groundwork of others to investigate what is unknown. Herein, we aim to provide a review focusing on protein host factors, both well-known and relatively new, that impact HIV-1 replication in a positive or negative manner at each stage of the replication cycle, highlighting factors unique to the various HIV-1 target cell types where appropriate. Full article
(This article belongs to the Special Issue Cellular Mechanisms Regulating HIV Replication)
Show Figures

Figure 1

Figure 1
<p>Host factors and restriction factors interact with the HIV-1 capsid to either assist or prevent successful cytoplasmic transport, nuclear entry, and capsid uncoating. (1) After attachment and envelope fusion with the plasma membrane, the capsid is released into the cytoplasm. Factors found within the capsid (CypA, red spheres; IP6, green spheres) as well as outside of the capsid (Sec24c, yellow spheres; PDZD8, blue spheres) bind the capsid to promote stability during cytoplasmic transport. Restriction factors (TRIM5α, black squares; TRIM11, purple squares; PQBP1, red triangles) facilitate capsid disassembly and/or immune detection of the viral genome. Transport of the capsid can occur in a microtubule-dependent (2a) or microtubule-independent (2b) manner. Using the adaptor protein BICD2, the microtubule-associated protein dynein moves the capsid inwards towards the nucleus; FEZ1 and kinesin-1 (not shown in Figure), though outwards-bound from the nucleus, still positively impact capsid localization to the nucleus. (3) At the nucleus, capsid entry through the nuclear pore complex is aided by host factors (Nup358, Nup 153, CPSF6) and prevented by restriction factors (MX2). (4) Once inside the nucleus, host factors promote capsid destabilization and disassembly (TNPO1, TNPO3, Pin1, CypA), ensuring proper viral genome release, followed by localization to sites of open chromatin by CPSF6. At the same time, restriction factors (MX2, Daxx) will attempt to prevent uncoating and genome release.</p>
Full article ">Figure 2
<p>Various components of the Gag polyprotein interact with host factors to facilitate the recruitment of ESCRT pathway machinery, leading to productive and efficient virion budding. See text for further details.</p>
Full article ">
21 pages, 10664 KiB  
Article
Overexpression of the β-Subunit of Acid Ceramidase in the Epidermis of Mice Provokes Atopic Dermatitis-like Skin Symptoms
by Miho Sashikawa-Kimura, Mariko Takada, Md Razib Hossain, Hidetoshi Tsuda, Xiaonan Xie, Mayumi Komine, Mamitaro Ohtsuki and Genji Imokawa
Int. J. Mol. Sci. 2024, 25(16), 8737; https://doi.org/10.3390/ijms25168737 (registering DOI) - 10 Aug 2024
Viewed by 410
Abstract
We previously reported that a pathogenic abnormality in the barrier and water-holding functions of the stratum corneum (SC) in the skin of patients with atopic dermatitis (AD) is mainly attributable to significantly decreased levels of total ceramides in the SC. That decrease is [...] Read more.
We previously reported that a pathogenic abnormality in the barrier and water-holding functions of the stratum corneum (SC) in the skin of patients with atopic dermatitis (AD) is mainly attributable to significantly decreased levels of total ceramides in the SC. That decrease is mediated by the abnormal expression of a novel ceramide-reducing enzyme, sphingomyelin/glucosylceramide deacylase (SGDase), which is the β-subunit (ASAH1b) of acid ceramidase. In this study, we determined whether mice overexpressing ASAH1b in their epidermis develop AD-like skin symptoms. We generated transgenic (TG) mice overexpressing ASAH1b, regulated by the involucrin promoter, to localize its expression in the upper epidermis. After hair removal using a depilatory cream containing glycolic acid, the TG mice without any visible skin inflammation at 8 weeks of age had increased levels of ASAH1b and decreased levels of SC ceramide, with disrupted barrier functions measured by trans-epidermal water loss compared to the wild-type (WT) mice. Interestingly, enzymatic assays revealed that SGDase activity was not detectable in the skin of the TG mice compared to WT mice. Immunological staining revealed that there was an increased expression level of IL-33 in the epidermis and an accumulation of macrophages in the dermis of TG mice compared to WT mice, which are phenotypic characteristics of AD, that were exacerbated by tape-stripping of the skin. In the skin of the TG mice, the mRNA levels of IL-5, CCL11, IL-22, CXCL10, and IFNγ were significantly upregulated compared to the WT mice, and tape-stripping significantly increased the mRNA levels of IL-4, IL-33, CXCL1, CXCL12, TLR9, and CD163 compared to WT mice. These findings strongly indicate that the skin of the depilatory cream-treated TG mice exists in an atopic dry skin condition that is highly sensitive to various environmental stimuli. The sum of our results suggests that ASAH1b itself, even in the absence of its enzymatic activity, is a major etiologic factor for atopic dry skin symptoms via an unknown mechanism. Full article
(This article belongs to the Section Molecular Immunology)
Show Figures

Figure 1

Figure 1
<p>Appearance of the ear skin, H&amp;E staining, skin score and thickness of the epidermis before and after repeated tape-stripping. (<b>A</b>) (<b>a</b>) WT ears at day 0 before tape-stripping, (<b>b</b>) WT ears at day 2 after tape-stripping of the right ear (red arrow); the left ear (blue arrow) was not tape-stripped as a control, (<b>c</b>) H&amp;E staining of the left WT ear at day 2, (<b>d</b>) H&amp;E staining of right WT ear at day 2 after tape-stripping, (<b>e</b>) TG ears at day 0 before tape-stripping, (<b>f</b>) TG ears at day 2 after tape-stripping of the right ear (red arrow); the left ear (blue arrow) was not tape-stripped as a control, (<b>g</b>) H&amp;E staining of the left TG ear at day 2, (<b>h</b>) H&amp;E staining of the right TG ear at day 2 after tape-stripping. (<b>B</b>) (<b>a</b>) Total visible score on the outside ear skin of TG and WT mice at the age of 8 weeks before and day 2 after tape-stripping. n = 7 for WT, n = 5 for TG, *: <span class="html-italic">p</span> &lt; 0.05 by Mann–Whitney comparisons test. (<b>b</b>) Thickness of the epidermis of the outside of the ear measured using Image J software (imageJ 1.54g). n = 5~7, ***: <span class="html-italic">p</span> &lt; 0.001, ****: <span class="html-italic">p</span> &lt; 0.0001 by Tukey’s multiple comparisons test. nd: not significant difference.</p>
Full article ">Figure 2
<p>Surface appearance of the dorsal skin of TG and WT mice at the age of 8 weeks after hair removal using electric hair clippers and barrier function values three days after hair removal. (<b>A</b>) Skin appearance three days after hair removal. (<b>B</b>) Barrier function measured by TEWL three days after hair removal, TG; n = 6, WT; n = 4, ns: not significant, by Mann–Whitney test.</p>
Full article ">Figure 3
<p>Surface appearance of the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks and barrier function before and at day 2 after tape-stripping. (<b>A</b>) Skin appearance at Days 0, 1 and 2, <b>right</b> and <b>left</b> sides (separated by red line) of the dorsal skin show tape-stripped and non-tape-stripped areas, respectively. (<b>B</b>) Barrier function measured by TEWL before and at day 2 after tape-stripping, n = 5, ****: <span class="html-italic">p</span> &lt; 0.0001, ***: <span class="html-italic">p</span> &lt; 0.001 by Tukey’s multiple comparisons test.</p>
Full article ">Figure 4
<p>H&amp;E staining of the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and after tape-stripping. (<b>A</b>) H&amp;E staining, (<b>a</b>) WT dorsal skin before tape-stripping, (<b>b</b>) WT dorsal skin at day 2 after tape-stripping, (<b>c</b>) TG dorsal skin before tape-stripping, (<b>d</b>) TG dorsal skin at day 2 after tape-stripping, bars = 100 μm, (<b>B</b>) Epidermal thickness, analysis by measurement using Image J software (imageJ 1.54g). n = 6, ****: <span class="html-italic">p</span> &lt; 0.0001 by Tukey’s multiple comparisons test.</p>
Full article ">Figure 5
<p>Immunostaining of ASAH1 with an anti-ASAH1b antibody in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. (<b>A</b>) Immunostaining, bars = 100 μm, (<b>B</b>) Stained area, analysis by measurement with a Keyence Image Analyzer. n = 6, ****: <span class="html-italic">p</span> &lt; 0.0001, **: <span class="html-italic">p</span> &lt; 0.01 by Tukey’s multiple comparisons test. TS = tape-stripped.</p>
Full article ">Figure 6
<p>Immunostaining of ceramide with an anti-ceramide antibody in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. (<b>A</b>) Immunostaining, bars = 100 μm, (<b>a</b>) WT/TS (−), (<b>b</b>) WT/TS (+), (<b>c</b>) TG/TS (−), (<b>d</b>) TG/TS (+), (<b>B</b>) Stained area, analysis by measurement with a Keyence Image Analyzer, n = 6, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test. TS: tape-stripped.</p>
Full article ">Figure 7
<p>Western blotting of ASAH1b protein in epidermal homogenates of the depilatory cream-treated dorsal skin and abdominal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. ME = treated with 2-mercaptoethanol. The epidermal homogenates were obtained at 8 weeks of age or at day 2 after tape-stripping.</p>
Full article ">Figure 8
<p>Immunostaining of macrophages using various macrophage markers in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. (<b>A</b>) Immunostaining of F4/80, bars = 100 μm, analysis by measurement with a Keyence Image Analyzer, n = 5, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test. (<b>B</b>) Immunostaining of CD80, bars = 100 μm, analysis by measurement with a Keyence Image Analyzer, n = 3, ***: <span class="html-italic">p</span> &lt; 0.001, **: <span class="html-italic">p</span> &lt; 0.01 by Tukey’s multiple comparisons test. (<b>C</b>) Immunostaining of CD163, bars = 100 μm, analysis by measurement with a Keyence Image Analyzer, n = 5, *: <span class="html-italic">p</span> &lt; 0.05, by Tukey’s multiple comparisons test. (<b>a</b>) WT/TS (−), (<b>b</b>) WT/TS (+), (<b>c</b>) TG/TS (−), (<b>d</b>) TG/TS (+), TS: Tape-stripped. ns: not significant.</p>
Full article ">Figure 9
<p>Toluidine blue staining of mast cells (<b>A</b>) and immunostaining with an anti-IL-33 antibody (<b>B</b>) in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. bars = 100 μm, analysis by measurement with a Keyence Image Analyzer. n = 6, ***: <span class="html-italic">p</span> &lt; 0.001, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test. (<b>a</b>) WT/TS (−), (<b>b</b>) WT/TS (+), (<b>c</b>) TG/TS (−), (<b>d</b>) TG/TS (+), TS: Tape-stripping.</p>
Full article ">Figure 10
<p>Effects of the overexpression of ASAH1b on the activity of SGDase in the epidermis. (<b>A</b>): LC-MS-MS spectrum for released SPC by sphingolipid ceramide N-deacylase from <span class="html-italic">Pseudomonas</span> sp. as a positive control. (<b>B</b>): LC-MS-MS spectrum for released SPC by SGDase in the epidermal homogenate from TG/TS (+). (<b>C</b>): Time course of the released SPC after enzymatic reaction with epidermal homogenate or sphingolipid ceramide N-deacylase from <span class="html-italic">Pseudomonas</span> sp. as positive control.An epidermal homogenate was incubated for 5 h at 37 °C with varying amounts of SM. The enzymatic activities expressed as released SPC for WT/TS(-), WT/TS(+), TG/TS(-), and TG/TS(+) were located at O level. The final reaction mixtures contained 50 mM potassium acetate buffer (pH 4.7), the enzyme source, the substrate, 0.1% Triton X-100 and 20 mM CaCl<sub>2</sub>. The rate of SPC generation was measured as a function of SGDase by LC-MS-MS analysis. PC: positive control using sphingolipid ceramide N-deacylase from <span class="html-italic">Pseudomonas</span> sp. as SGDase. TS: Tape-stripping.</p>
Full article ">Figure 11
<p>Effects of the overexpression of ASAH1b on the activity of ASAH1 in the epidermis. (<b>A</b>): LC-MS-MS spectrum of released sphingosine (SPH) after enzymatic reaction with epidermal homogenate. (<b>B</b>): Enzymatic activity of ASAH1 expressed as released SPH in epidermal homogenates of WT and TG mice. An epidermal homogenate was incubated for 12 h at 37 °C with varying amounts of ceramide substrates. The final reaction mixtures contained 50 mM potassium acetate buffer (pH 4.7), the enzyme source, the substrate, 0.1% Triton X-100 and 20 mM CaCl<sub>2</sub>. Blue rectangles indicate TG mice. The rate of sphingosine (SPH) generation was measured as a function of ASAH1 by LC-MS-MS analysis. n = 6, ****: <span class="html-italic">p</span> &lt; 0.0001, **: <span class="html-italic">p</span> &lt; 0.01, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test.</p>
Full article ">Figure 12
<p>mRNA expression levels of inflammatory cytokines and chemokines in the depilatory cream-treated dorsal skin of TG and WT mice at the age of 8 weeks before and at day 2 after tape-stripping. n = 3~9, ****: <span class="html-italic">p</span> &lt; 0.0001, ***: <span class="html-italic">p</span> &lt; 0.001, **: <span class="html-italic">p</span> &lt; 0.01, *: <span class="html-italic">p</span> &lt; 0.05 by Tukey’s multiple comparisons test.</p>
Full article ">
19 pages, 1292 KiB  
Review
A Review on Canine and Human Soft Tissue Sarcomas: New Insights on Prognosis Factors and Treatment Measures
by Filippo Dell’Anno, Roberta Giugliano, Valeria Listorti and Elisabetta Razzuoli
Vet. Sci. 2024, 11(8), 362; https://doi.org/10.3390/vetsci11080362 - 10 Aug 2024
Viewed by 269
Abstract
Soft tissue sarcomas (STSs) represent a diverse group of tumors arising from mesenchymal cells, affecting both humans and animals, including dogs. Although STSs represent a class of rare tumors, especially in humans, they pose significant clinical challenges due to their potential for local [...] Read more.
Soft tissue sarcomas (STSs) represent a diverse group of tumors arising from mesenchymal cells, affecting both humans and animals, including dogs. Although STSs represent a class of rare tumors, especially in humans, they pose significant clinical challenges due to their potential for local recurrence and distant metastasis. Dogs, as a model for human STSs, offer several advantages, including exposure to similar environmental risk factors, genetic diversity among breeds, and the spontaneous development of tumors. Furthermore, canine tumors closely mimic the heterogeneity and complexity of human tumors, making them valuable for research into disease progression and treatment effectiveness. Current treatment approaches for STSs in both dogs and humans primarily involve surgery, radiation therapy, and chemotherapy, with treatment decisions based on tumor characteristics and patient factors. However, the development of novel therapeutic strategies is essential, given the high failure rate of new drugs in clinical trials. To better design new tailored treatments, comprehension of the tumor microenvironment (TME) is fundamental, since it plays a crucial role in STS initiation and progression by modulating tumor behavior, promoting angiogenesis, and suppressing immune responses. Notably, TME features include cancer-associated fibroblasts (CAFs), extracellular matrix (ECM) alterations, and tumor-associated macrophages (TAMs) that, depending on their polarization state, can affect immune responses and thus the patient’s prognosis. In this review, new therapeutical approaches based on immunotherapy will be deeply explored as potential treatment options for both dogs and humans with STSs. In conclusion, this review provides an overview of the current understanding of STSs in dogs and humans, emphasizing the importance of the TME and potential treatment strategies. Full article
(This article belongs to the Special Issue Focus on Tumours in Pet Animals)
Show Figures

Figure 1

Figure 1
<p>A resume of the available therapies for humans and dogs in the treatment of soft tissue sarcomas.</p>
Full article ">Figure 2
<p>Tumor microenvironment in human and canine soft tissue sarcomas. (<b>A</b>) Tumor-associated macrophages (TAMs) can undergo two different polarizations, M1 or M2, starting from M0 populations defined as non-activated macrophages. M0 populations differentiate into the M1 subtype after stimulation with granulocyte–monocyte colony-stimulating factor (GM-CSF), lipopolysaccharide (LPS), or interferon-gamma (IFNγ). M2 polarization is activated by M-CSF, IL-4, IL 10, and IL 13, and it induces a pro-tumoral environment. (<b>B</b>) M2 can release signaling molecules and growth factors such as basic fibroblast growth factor (bFGF), platelet-derived growth factor (PDGF), and vascular endothelial growth factor (VEGF). (<b>C</b>) The activation of the VEGF pathway is responsible for angiogenesis and tumor growth. (<b>D</b>) VEGF can be used as a prognostic marker to predict tumor recurrence or survival in STSs. Indeed, there is a positive correlation between increased VEGF expression and a higher tumor grade. Moreover, TAMs negatively regulate cytotoxic effector cells, such as CD8+ and natural killer (NK) cells, and they often express programmed death-ligand 1 (PD-L1)/L2.</p>
Full article ">Figure 3
<p>Immunotherapy approaches specific to treat canine and human soft tissue sarcomas. See <a href="#vetsci-11-00362-t001" class="html-table">Table 1</a> for a more detailed description.</p>
Full article ">
17 pages, 719 KiB  
Review
Endothelial Cell Dysfunction Due to Molecules Secreted by Macrophages in Sepsis
by Heng He, Wei Zhang, Luofeng Jiang, Xirui Tong, Yongjun Zheng and Zhaofan Xia
Biomolecules 2024, 14(8), 980; https://doi.org/10.3390/biom14080980 (registering DOI) - 9 Aug 2024
Viewed by 426
Abstract
Sepsis is recognized as a syndrome of systemic inflammatory reaction induced by dysregulation of the body’s immunity against infection. The multiple organ dysfunction associated with sepsis is a serious threat to the patient’s life. Endothelial cell dysfunction has been extensively studied in sepsis. [...] Read more.
Sepsis is recognized as a syndrome of systemic inflammatory reaction induced by dysregulation of the body’s immunity against infection. The multiple organ dysfunction associated with sepsis is a serious threat to the patient’s life. Endothelial cell dysfunction has been extensively studied in sepsis. However, the role of macrophages in sepsis is not well understood and the intrinsic link between the two cells has not been elucidated. Macrophages are first-line cells of the immune response, whereas endothelial cells are a class of cells that are highly altered in function and morphology. In sepsis, various cytokines secreted by macrophages and endothelial cell dysfunction are inextricably linked. Therefore, investigating how macrophages affect endothelial cells could offer a theoretical foundation for the treatment of sepsis. This review links molecules (TNF-α, CCL2, ROS, VEGF, MMP-9, and NO) secreted by macrophages under inflammatory conditions to endothelial cell dysfunction (adhesion, permeability, and coagulability), refining the pathophysiologic mechanisms of sepsis. At the same time, multiple approaches (a variety of miRNA and medicines) regulating macrophage polarization are also summarized, providing new insights into reversing endothelial cell dysfunction and improving the outcome of sepsis treatment. Full article
(This article belongs to the Special Issue The Role of Modulating Intracellular Signaling in Immune System)
Show Figures

Figure 1

Figure 1
<p>Comparison of endothelial cells under physiologic and pathologic conditions. (<b>A</b>) In the physiologic state, blood cells are in a relatively quiescent state. Endothelial cells are in a tightly packed state containing an intact surface glycocalyx, robust intercellular junctions, a relaxed cytoskeleton, and small amounts of adhesion molecules and selectins. (<b>B</b>) In the setting of LPS or sepsis, monocytes, macrophages, and neutrophils produce a large number of signaling molecules, including NO, VEGF, TNF-α, MMP-9, ROS, TF, and CCL-2. These molecules lead to structural and functional alterations in endothelial cells, including degradation of the glycocalyx, increased exposure to and expression of adhesion molecules and selectins, rupture of intercellular connections, and cytoskeleton shrinkage by phosphorylation, and apoptosis due to mitochondrial disruption. Neutrophils cross the endothelial cell barrier through an enlarged paracellular gap. These changes ultimately cause an increase in the adhesion and permeability of the endothelium.</p>
Full article ">
14 pages, 1208 KiB  
Review
The Effects of Gynecological Tumor Irradiation on the Immune System
by Jesus Romero Fernandez, Sofia Cordoba Largo, Raquel Benlloch Rodriguez and Beatriz Gil Haro
Cancers 2024, 16(16), 2804; https://doi.org/10.3390/cancers16162804 - 9 Aug 2024
Viewed by 220
Abstract
Radiobiology has evolved from a mechanistic model based on DNA damage and response factors into a more complex model that includes effects on the immune system and the tumor microenvironment (TME). Irradiation has an immunomodulatory effect that can manifest as increased anti-tumor immunity [...] Read more.
Radiobiology has evolved from a mechanistic model based on DNA damage and response factors into a more complex model that includes effects on the immune system and the tumor microenvironment (TME). Irradiation has an immunomodulatory effect that can manifest as increased anti-tumor immunity or immunosuppression. Irradiation promotes an inflammatory microenvironment through the release of pro-inflammatory cytokines and endothelial damage, which recruit immune system cells to the irradiated area. Radiation-induced immunogenic cell death (ICD), characterized by the release of damage-associated molecular patterns (DAMPs) and tumor antigens, triggers an anti-tumor immune response of both innate and adaptive immunity. Anti-tumor immunity can manifest at a distance from the irradiated area, a phenomenon known as the abscopal effect (AE), which involves dendritic cells and CD8+ T cells. Irradiation also produces an immunosuppressive effect mediated by tumor-associated macrophages (TAMs) and regulatory T lymphocytes (Tregs), which counterbalances the immunostimulatory effect. In this work, we review the mechanisms involved in the radiation-induced immune response, which support the combined treatment of RT and immunotherapy, focusing, where possible, on gynecologic cancer. Full article
(This article belongs to the Special Issue Radiotherapy in Gynecological Cancer: State of the Art)
Show Figures

Figure 1

Figure 1
<p>Therapeutic index: the therapeutic index is established by comparing the sigmoidal dose–response curve for tumor control (red) with that for toxicity (blue). The dashed lines represent the modification of the dose-response curves due to the antitumor immune response (red) or to techniques that spare healthy tissues such as IMRT or brachytherapy (blue).</p>
Full article ">Figure 2
<p>Linear–quadratic model. α parameter (red) represents a non-repairable lesion produced by a single irradiation event (linear component), and β parameter (blue) represents several repairable lesions produced by independent events (quadratic component).</p>
Full article ">Figure 3
<p>Schematic representation of the complex network of the immune response pathways triggered by irradiation. DSB: double-strand DNA break; cGAS: cyclic GMP-AMP synthase; STING: stimulator of interferon genes; IFN: interferon; CCL2, CCL19, CCL21, and CCL22: chemokines; TAAs: tumor antigens; ICD: immunogenic cell death; ATP: adenosine triphosphate; CALRT: calreticulin; HMGB1: high-mobility group protein B1; P2X, CD91, and TRL4: pattern recognition receptors; DC: dendritic cell; MHC: major histocompatibility complex; TCR: T-cell receptor; TNFα: tumor necrosis factor-alpha; ROS: reactive oxygen species; IL: interleukin; TGFβ: transforming growth factor-beta; NFκB: nuclear factor kappa B; TAM: tumor-associated macrophage; TAN: tumor-associated neutrophil; Treg: regulatory T lymphocyte.</p>
Full article ">
19 pages, 1360 KiB  
Article
Immunoregulatory Cells and Cytokines Discriminate Disease Activity Score 28-Remission Statuses and Ultrasound Grades in Rheumatoid Arthritis Patients with Non-High Disease Activity
by Lieh-Bang Liou, Yao-Fan Fang, Ping-Han Tsai, Yen-Fu Chen, Che-Tzu Chang, Chih-Chieh Chen and Wen-Yu Chiang
Int. J. Mol. Sci. 2024, 25(16), 8694; https://doi.org/10.3390/ijms25168694 - 9 Aug 2024
Viewed by 289
Abstract
It is not clear whether immunoregulatory cytokines and cells are associated with Disease Activity Score 28 (DAS28) scores and ultrasound grades/scores. Here, we investigated the relationships between immunoregulatory cytokines or cells and different DAS28 scores or ultrasound grades/scores in patients with rheumatoid arthritis [...] Read more.
It is not clear whether immunoregulatory cytokines and cells are associated with Disease Activity Score 28 (DAS28) scores and ultrasound grades/scores. Here, we investigated the relationships between immunoregulatory cytokines or cells and different DAS28 scores or ultrasound grades/scores in patients with rheumatoid arthritis (RA). This study enrolled 50 RA patients (with 147 visits) who had remission/low/moderate DAS28-ESR scores (92% in remission and low disease activity) at baseline. Blood was collected and an ultrasound was performed three times in a year. Percentages of regulatory B cells and T regulatory type 1 cells and M2 macrophage numbers in the blood were examined. Plasma levels of 10 immunoregulatory cytokines IL-4, IL-5, IL-9, IL-10, IL-13, IL-27, IL-35, TGF-β1, sTNF-R1, and sTNF-R2 and monocyte chemotactic protein-1 (MCP-1) were assessed using ELISA assay. The correlations of cytokines and cells with different DAS28 scores and ultrasound grades were investigated, and cytokines and cells were compared between different categories of DAS28 scores and ultrasound grades. Plasma TGF-β1 levels were higher in the DAS28-ESR < 2.6 (remission) subgroup than in the DAS28-ESR ≥ 2.6 (nonremission) subgroup (p = 0.037). However, plasma TGF-β1 levels were higher in the high ultrasound grade subgroup than those in the low ultrasound grade subgroup (p = 0.007). The number of M2 macrophages was lower in the DAS28-MCP-1 < 2.2 subgroup than in the DAS28-MCP-1 ≥ 2.2 subgroup (p = 0.036). The levels of TGF-β1, sTNF-R2, IL-10, and IL-27 were higher in patients with high ultrasound grades than in those with low ultrasound grades. IL-27 was also higher in the nonremission DAS28-ESR subgroup than the remission one (p = 0.025). Moreover, sTNF-R1 levels in the 2011 American College of Rheumatology/European League Against Rheumatism (ACR/EULAR) remission subgroup were significantly lower than in the 2011 ACR/EULAR nonremission subgroup (p = 0.007). This trend was reflected in that lower sTNF-R1 levels correlated with low DAS28-MCP-1 scores (rho = 0.222, p = 0.007). We conclude that high plasma TGF-β1 levels indicate the DAS28-ESR remission (<2.6) subgroup and the high ultrasound grade subgroup. IL-27 probably connects the nonremission DAS28-ESR to high ultrasound grades. Low sTNF-R1 levels probably link low DAS28-MCP-1 scores with the 2011 ACR/EULAR remission subgroup. It suggests that incongruent immuno-inflammatory abnormalities exist between DAS28 scores and ultrasound grades, and are also dissimilar among various DAS28-formula categories. Therefore, this study may provide a basis for further research into individual cytokines and immunoregulatory cells behind each DAS28 formula and ultrasound grades/scores. Full article
(This article belongs to the Special Issue Molecular Insights in Rheumatoid Arthritis)
Show Figures

Figure 1

Figure 1
<p>Different cytokine levels or M2 macrophage numbers across DAS28-based remission statuses: (<b>A</b>–<b>C</b>) Plasma TGF-β1, IL-27, and IL-5 levels significantly differentiated DAS28-ESR &lt; 2.6 (n = 98) from DAS28-ESR ≥ 2.6 (n = 49, having higher values for IL-27 and IL-5) at <span class="html-italic">p</span> = 0.037, 0.025, and 0.028, respectively. (<b>D</b>) The number of M2 macrophages in the blood discriminated DAS28-MCP-1 &lt; 2.2 (n = 79) from DAS28-MCP-1 ≥ 2.2 (n = 68, having higher M2 macrophage numbers) at <span class="html-italic">p</span> = 0.036. All comparisons were conducted using the Mann–Whitney U test. * Asterisks indicate the subgroup had significantly higher cytokine levels or M2 macrophage numbers than another subgroup.</p>
Full article ">Figure 2
<p>Correlation of cytokines and cells with Total Ultrasound Scores. Ultrasound grades were scored as in ref. [<a href="#B20-ijms-25-08694" class="html-bibr">20</a>]: (<b>A</b>) The percentage of Tr1 cells (n = 89) correlated with Total Ultrasound Scores (the sum of ultrasound grades for grayscale and power Doppler of all examined joints) in the ultrasound grade subgroup (≤1) at rho = 0.242 and <span class="html-italic">p</span> = 0.022. Tr1 is presented as percentages (%). (<b>B</b>) The M2 macrophage numbers (n = 89) correlated with Total Ultrasound Scores in the ultrasound grade subgroup (≤1) at rho = 0.221 and <span class="html-italic">p</span> = 0.038. (<b>C</b>) The IL-10 level (n = 58) correlated with Total Ultrasound Scores in the high ultrasound grade subgroup (&gt;1) at rho = 0.311 and <span class="html-italic">p</span> = 0.017. (<b>D</b>) The TGF-β1 level (n = 147) correlated with Total Ultrasound Scores at rho = 0.203 and <span class="html-italic">p</span> = 0.033. Correlation performed by Spearman’s correlation.</p>
Full article ">Figure 3
<p>Comparison of M2 macrophage numbers and immunoregulatory cytokines between two groups with ultrasound grades for individual joints. Ultrasound grades were scored as described in a previous study [<a href="#B20-ijms-25-08694" class="html-bibr">20</a>]: (<b>A</b>) The number of M2 macrophages in the group with low ultrasound grades (grades 0 and 1) (n = 140) was higher than that in the group with high ultrasound grades (grades 2 and 3) (n = 7) for the ankle joint (<span class="html-italic">p</span> = 0.038). (<b>B</b>) The number of M2 macrophages in the group with low ultrasound grades (n = 137) was higher than that in the group with high ultrasound grades (n = 10) for the 2nd metacarpo-phalangeal (MCP) joint (<span class="html-italic">p</span> = 0.027). (<b>C</b>) IL-10 levels in the group with high ultrasound grades (n = 15) were higher than those in the group with low ultrasound grades (n = 132) in the second MCP joint (<span class="html-italic">p</span> = 0.038). (<b>D</b>) IL-27 levels in the group with high ultrasound grades (n = 15) were higher than those in the group with low ultrasound grades (n = 132) in the second and third MCP joint (<span class="html-italic">p</span> = 0.002). (<b>E</b>) TGF-β1 levels in the group with high ultrasound grades (n = 15) were higher than those in the group with low ultrasound grades (n = 132) for the second and third MCP joints (<span class="html-italic">p</span> = 0.007). (<b>F</b>) sTNF-R2 levels in the group with high ultrasound grades (n = 10) were higher than those in the group with low ultrasound grades (n = 137) in the second MCP joint (<span class="html-italic">p</span> = 0.010). All comparisons were conducted using the Mann–Whitney U test. * Asterisks indicate the subgroup had significantly higher cytokine levels or M2 macrophage numbers than another subgroup.</p>
Full article ">
20 pages, 3776 KiB  
Review
Surface Markers and Chemokines/Cytokines of Tumor-Associated Macrophages in Osteosarcoma and Other Carcinoma Microenviornments—Contradictions and Comparisons
by Rikito Tatsuno, Yoshihiro Komohara, Cheng Pan, Tomonori Kawasaki, Atsushi Enomoto, Takahiro Jubashi, Hiroyuki Kono, Masanori Wako, Tomoyuki Ashizawa, Hirotaka Haro and Jiro Ichikawa
Cancers 2024, 16(16), 2801; https://doi.org/10.3390/cancers16162801 - 8 Aug 2024
Viewed by 396
Abstract
Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents. Prognosis is improving with advances in multidisciplinary treatment strategies, but the development of new anticancer agents has not, and improvement in prognosis for patients with pulmonary metastases has stalled. In [...] Read more.
Osteosarcoma (OS) is the most common primary bone tumor in children and adolescents. Prognosis is improving with advances in multidisciplinary treatment strategies, but the development of new anticancer agents has not, and improvement in prognosis for patients with pulmonary metastases has stalled. In recent years, the tumor microenvironment (TME) has gained attention as a therapeutic target for cancer. The immune component of OS TME consists mainly of tumor-associated macrophages (TAMs). They exhibit remarkable plasticity, and their phenotype is influenced by the TME. In general, surface markers such as CD68 and CD80 show anti-tumor effects, while CD163 and CD204 show tumor-promoting effects. Surface markers have potential value as diagnostic and prognostic biomarkers. The cytokines and chemokines produced by TAMs promote tumor growth and metastasis. However, the role of TAMs in OS remains unclear to date. In this review, we describe the role of TAMs in OS by focusing on TAM surface markers and the TAM-produced cytokines and chemokines in the TME, and by comparing their behaviors in other carcinomas. We found contrary results from different studies. These findings highlight the urgency for further research in this field to improve the stalled OS prognosis percentages. Full article
(This article belongs to the Special Issue Clinical Treatment of Osteosarcoma)
Show Figures

Figure 1

Figure 1
<p>Cell-cell interaction between TAMs and OS cells. IHC analysis indicated that TAMs are predominant stromal cells in OS tissues, especially in metastatic lesions. Picture figures were IHC of Iba-1 (a pan-macrophage marker) from paired primary lesion and lung metastatic lesion of three cases. Scale bar: 50 mm. Picture figures of Iba1 IHC of three cases shown in our previous study were presented [<a href="#B34-cancers-16-02801" class="html-bibr">34</a>]. IHC was performed in newly prepared sections as described in a previous study [<a href="#B52-cancers-16-02801" class="html-bibr">52</a>]. The study was approved by the ethics committees of each institution (No. 2456).</p>
Full article ">Figure 2
<p>Biomarkers related to osteosarcoma and tumor-associated macrophage. COX-2: Cyclooxygenase-2, EMT; Epithelial-mesenchymal transition, OS: Osteosarcoma, PITX1: Paired-like homeodomain transcription factor 1, TAM: Tumor-associated macrophage.</p>
Full article ">Figure 3
<p>Myeloid cell subgroups suggested by single cell-RNA sequence. (<b>A</b>); Detailed methods and results were described and shown in <a href="#app1-cancers-16-02801" class="html-app">Supplementary Materials</a> UMAP plot of <span class="html-italic">AIF1</span> (<span class="html-italic">Iba-1</span>) gene expression across all the cells (GSE152048) is presented. (<b>B</b>); UMAP plot of the selected <span class="html-italic">AIF1 (Iba-1)</span> gene and <span class="html-italic">CD74</span> gene are presented. (<b>C</b>); UMAP plot of selected genes indicated the myeloid cells were composed of their subpopulations (dendritic cells, resident TAMs, and monocyte-derived TAMs). (<b>D</b>); Gene expression scheme in each subpopulation was shown.</p>
Full article ">
26 pages, 3187 KiB  
Review
Aggregatibacter actinomycetemcomitans Dispersin B: The Quintessential Antibiofilm Enzyme
by Jeffrey B. Kaplan, Svetlana A. Sukhishvili, Miloslav Sailer, Khalaf Kridin and Narayanan Ramasubbu
Pathogens 2024, 13(8), 668; https://doi.org/10.3390/pathogens13080668 - 7 Aug 2024
Viewed by 363
Abstract
The extracellular matrix of most bacterial biofilms contains polysaccharides, proteins, and nucleic acids. These biopolymers have been shown to mediate fundamental biofilm-related phenotypes including surface attachment, intercellular adhesion, and biocide resistance. Enzymes that degrade polymeric biofilm matrix components, including glycoside hydrolases, proteases, and [...] Read more.
The extracellular matrix of most bacterial biofilms contains polysaccharides, proteins, and nucleic acids. These biopolymers have been shown to mediate fundamental biofilm-related phenotypes including surface attachment, intercellular adhesion, and biocide resistance. Enzymes that degrade polymeric biofilm matrix components, including glycoside hydrolases, proteases, and nucleases, are useful tools for studying the structure and function of biofilm matrix components and are also being investigated as potential antibiofilm agents for clinical use. Dispersin B is a well-studied, broad-spectrum antibiofilm glycoside hydrolase produced by Aggregatibacter actinomycetemcomitans. Dispersin B degrades poly-N-acetylglucosamine, a biofilm matrix polysaccharide that mediates biofilm formation, stress tolerance, and biocide resistance in numerous Gram-negative and Gram-positive pathogens. Dispersin B has been shown to inhibit biofilm and pellicle formation; detach preformed biofilms; disaggregate bacterial flocs; sensitize preformed biofilms to detachment by enzymes, detergents, and metal chelators; and sensitize preformed biofilms to killing by antiseptics, antibiotics, bacteriophages, macrophages, and predatory bacteria. This review summarizes the results of nearly 100 in vitro and in vivo studies that have been carried out on dispersin B since its discovery 20 years ago. These include investigations into the biological function of the enzyme, its structure and mechanism of action, and its in vitro and in vivo antibiofilm activities against numerous bacterial species. Also discussed are potential clinical applications of dispersin B. Full article
(This article belongs to the Special Issue Aggregatibacter actinomycetemcomitans)
Show Figures

Figure 1

Figure 1
<p>Dispersal of isolated <span class="html-italic">Aggregatibacter actinomycetemcomitans</span> biofilm colonies growing on the surface of polystyrene Petri dishes: (<b>left panel</b>) wild-type strain CU1000; (<b>right panel</b>) Δ<span class="html-italic">dspB</span> mutant strain JK1023. Satellite colonies surrounding the dispersed CU1000 biofilm colony were absent in the JK1023 culture. Photos were taken 3 d after inoculation. Scale bar = 1 mm. Image from [<a href="#B12-pathogens-13-00668" class="html-bibr">12</a>].</p>
Full article ">Figure 2
<p>Phylogenetic relatedness of dispersin B homologues based on pairwise alignments of the amino acid sequences listed in <a href="#pathogens-13-00668-t001" class="html-table">Table 1</a>. The alignment was generated using ClustalW, and the phylogenetic tree was generated using FastTree software. Lacto-<span class="html-italic">N</span>-biosidase from <span class="html-italic">Lactococcus lactis</span> (GenBank accession number AGY45663.1) was used as an outgroup to locate the root of the tree. Horizontal branch lengths are proportional to the number of amino acid differences in the pairwise alignments. Bacterial families are indicated on the right.</p>
Full article ">Figure 3
<p>Ribbon diagram of <span class="html-italic">A. actinomycetemcomitans</span> dispersin B; α-helices are colored red and green; β-strands are colored blue. Image source: Wikimedia Commons.</p>
Full article ">Figure 4
<p>Dispersin B’s active site and mechanism of action: (<b>A</b>) Electrostatic surface potential at the active site showing the negatively charged amino acids (Asp56, Asp183, Glu184, Glu332), which create a shallow anionic region in the catalytic pocket. The size of the pocket is approximately 12 Å. GOL, glycerol; ACY, acetate. Figure generated using ChimeraX [<a href="#B29-pathogens-13-00668" class="html-bibr">29</a>]. (<b>B</b>) Substrate hydrolysis mechanism proposed for dispersin B and other glycoside hydrolase family 20 hexosaminidases. In this substrate-assisted mechanism, Glu184 acts as the acid/base. The nucleophile is the <span class="html-italic">N</span>-acetyl group of the substrate, which is assisted by Asp183. Both exo- (dPNAG) and endoglycosidic (PNAG) cleavage are shown, where the leaving group is either deacetylated or acetylated, respectively. A suitably positioned Asp183 helps stabilize the oxazolium ion in the transition state. Figure generated using ChemDraw (PerkinElmer).</p>
Full article ">Figure 5
<p>Confocal microscopic analysis of PNAG expression by <span class="html-italic">Y. pestis</span> strain KIM6+ grown at 28 °C overnight on Congo red agar. After treatment of bacterial cells with either chitinase (<b>top panels</b>) or dispersin B (<b>bottom panels</b>), cells were stained with Syto 83 to visualize DNA (red) and Alexa Fluor 488-conjugated mAb F598 to detect PNAG (green). Bars = 10 µm. Figure from Yoong et al. [<a href="#B40-pathogens-13-00668" class="html-bibr">40</a>].</p>
Full article ">Figure 6
<p>Abiotic surfaces coated with dispersin B resist <span class="html-italic">S. epidermidis</span> biofilm formation and surface attachment: (<b>A</b>) Biofilm formation by <span class="html-italic">S. epidermidis</span> strain NJ9709 on glass slides containing an ultrathin layered poly(allylamine hydrochloride) (PAH) hydrogel coating (<b>left panel</b>) or a PAH coating loaded with dispersin B (<b>right panel</b>). Bacteria were cultured inside plastic cloning cylinders (5 mm internal diameter) that were attached to the slide with high-vacuum grease. After 12 h, the biofilms were rinsed, the cloning cylinders were removed, and the slides were photographed. The rings correspond to the footprints of the cloning cylinders. The biofilm appeared as a white film on the unloaded PAH layer, which was absent on the dispersin-B-loaded PAH layer. (<b>B</b>) Attachment of <span class="html-italic">S. epidermidis</span> strain ATCC35984 to uncoated stainless steel disks, or to disks coated with polydopamine- or plasma-based coatings with or without grafted dispersin B. Source: (<b>A</b>) [<a href="#B65-pathogens-13-00668" class="html-bibr">65</a>]; (<b>B</b>) redrawn from [<a href="#B66-pathogens-13-00668" class="html-bibr">66</a>].</p>
Full article ">Figure 7
<p>Effects of dispersin B on plant-associated bacteria: (<b>A</b>) <span class="html-italic">Xanthomonas citri</span> subsp. <span class="html-italic">citri</span> strain 306 forms aggregates when cultured in broth (<b>left panel</b>). These aggregates were rapidly dissolved upon dispersin B treatment (<b>right panel</b>). (<b>B</b>) Biofilm formation by <span class="html-italic">Ralstonia solanacearum</span> strain Molk2 in polystyrene microtiter plates in the absence or presence of 20 µg/mL dispersin B. Biofilms were stained with crystal violet. (<b>C</b>) Binding of <span class="html-italic">Pseudomonas fluorescens</span> strain WCS365 to tomato roots in the absence or presence of 20 µg/mL dispersin B. Bacteria were mixed with 6-day-old tomato roots for 90 min. The roots were then crushed, mixed by vortex agitation, diluted, and plated on agar for CFU enumeration. Each data point represents one individual root. (<b>D</b>) Tobacco leaves infected with <span class="html-italic">Pectobacterium carotovorum</span> subsp. <span class="html-italic">carotovorum</span> strain ATCC 15713. Leaves were photographed 24 h after inoculation: (<b>left</b>) wild-type tobacco leaf; (<b>right</b>) leaf from a transgenic tobacco plant expressing dispersin B. Source: (<b>A</b>–<b>C</b>) J.B. Kaplan, unpublished data; (<b>D</b>) Ragunath et al. [<a href="#B104-pathogens-13-00668" class="html-bibr">104</a>], N. Ramasubbu, unpublished data.</p>
Full article ">
17 pages, 5976 KiB  
Article
GRB7 Plays a Vital Role in Promoting the Progression and Mediating Immune Evasion of Ovarian Cancer
by Liang Wen, Wei Hu, Sen Hou, Ce Luo, Yiteng Jin, Zexian Zeng, Zhe Zhang and Yuanguang Meng
Pharmaceuticals 2024, 17(8), 1043; https://doi.org/10.3390/ph17081043 - 7 Aug 2024
Viewed by 271
Abstract
Background: Despite breakthroughs in treatment, ovarian cancer (OC) remains one of the most lethal gynecological malignancies, with an increasing age-standardized mortality rate. This underscores an urgent need for novel biomarkers and therapeutic targets. Although growth factor receptor-bound protein 7 (GRB7) is implicated in [...] Read more.
Background: Despite breakthroughs in treatment, ovarian cancer (OC) remains one of the most lethal gynecological malignancies, with an increasing age-standardized mortality rate. This underscores an urgent need for novel biomarkers and therapeutic targets. Although growth factor receptor-bound protein 7 (GRB7) is implicated in cell signaling and tumorigenesis, its expression pattern and clinical implications in OC remain poorly characterized. Methods: To systematically investigate GRB7’s expression in OC, our study utilized extensive datasets from TCGA, GTEx, CCLE, and GEO. The prognostic significance of GRB7 was evaluated by means of Kaplan–Meier and Cox regression analyses. Using a correlation analysis and gene set enrichment analysis, relationships between GRB7’s expression and gene networks, immune cell infiltration and immunotherapy response were investigated. In vitro experiments were conducted to confirm GRB7’s function in the biology of OC. Results: Compared to normal tissues, OC tissues exhibited a substantial upregulation of GRB7. Reduced overall survival, disease-specific survival, and disease-free interval were all connected with high GRB7 mRNA levels. The network study demonstrated that GRB7 is involved in pathways relevant to the course of OC and has a positive connection with several key driver genes. Notably, GRB7’s expression was linked to the infiltration of M2 macrophage and altered response to immunotherapy. Data from single-cell RNA sequencing data across multiple cancer types indicated GRB7’s predominant expression in malignant cells. Moreover, OC cells with GRB7 deletion showed decreased proliferation and migration, as well as increased susceptibility to T cell-mediated cytotoxicity. Conclusion: With respect to OC, our results validated GRB7 as a viable prognostic biomarker and a promising therapeutic target, providing information about its function in tumorigenesis and immune modulation. GRB7’s preferential expression in malignant cells highlights its significance in the biology of cancer and bolsters the possibility that it could be useful in enhancing the effectiveness of immunotherapy. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

Figure 1
<p>The expression of GRB7. (<b>A</b>,<b>B</b>) GRB7 mRNA levels in pan-cancer (<b>A</b>), OC (<b>B</b>), and the corresponding normal tissues in TCGA and GTEx databases. (<b>C</b>–<b>E</b>) GRB7’s expression in OC and normal tissues in GEO databases, GSE6008 (<b>C</b>), GSE36668 (<b>D</b>), and GSE66957 (<b>E</b>). (<b>F</b>) GRB7 protein levels in OC and paired adjacent normal tissues from cProCite database. (<b>G</b>) Representative results of immunochemically stained GRB7 proteins in OC and normal ovarian tissues from Human Protein Atlas. * <span class="html-italic">p</span> &lt; 0.05; *** <span class="html-italic">p</span> &lt; 0.001 by unpaired Student’s <span class="html-italic">t</span> test (<b>A</b>–<b>F</b>). ns, not significant.</p>
Full article ">Figure 2
<p>The prognosis and diagnosis value of GRB7 in OC. (<b>A</b>–<b>D</b>) OS, DSS, DFI, and PFI curves of lowly and highly expressed GRB7 in OC. (<b>E</b>) Univariate and multivariate regression analyses of GRB7 and clinicopathologic factors with OS in OC patients from TCGA. (<b>F</b>) A nomogram to predict OS probability at 1-year, 2-year, and 3-year overall survival probabilities for OC. * <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 by unpaired Student’s t test (E, F). # Events represents the number of death cases.</p>
Full article ">Figure 3
<p>Gene expression and enrichment of GRB7-associated gene in OC from TCGA. (<b>A</b>) Heatmap of top 20 genes positively correlated with GRB7 and top 20 negatively correlated genes in OC. (<b>B</b>) KEGG enrichment results of all 225 different expressed genes. (<b>C</b>) Biological processes, cellular components, and molecular functions from GO enrichment results. (<b>D</b>) Chord diagrams of biological processes.</p>
Full article ">Figure 4
<p>The immune infiltration and association with immunotherapy response of GRB7 in OC. (<b>A</b>) Immune cell enrichment in low and high expression levels of GRB7 in OC from CIBERSORT. (<b>B</b>) GRB7’s expression levels in responders and non-responders of ICB in syngeneic mouse models. (<b>C</b>) Protein–protein interaction signature of GRB7 from string database. (<b>D</b>,<b>E</b>) The GRB7 signature expression level in responders and non-responders of ICB-treated clinical cohorts, PD1 + CTLA4 in melanoma (<b>D</b>), and PDL1 in metastatic urothelial cancer (<b>E</b>). (<b>F</b>) GRB7 knockout in cancer cells cocultured with T cells from several CRISPR-Cas9 screens. Box plots indicate median (middle line), 25th and 75th percentile (box), and 5th and 95th percentile (whiskers) (<b>A</b>,<b>B</b>,<b>D</b>,<b>E</b>), and each dot in the scatter represents an individual patient sample (<b>D</b>,<b>E</b>). * <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 by unpaired Student’s <span class="html-italic">t</span> test (<b>A</b>,<b>B</b>,<b>D</b>,<b>E</b>). ns, not significant.</p>
Full article ">Figure 5
<p>GRB7’s expression among different cell types and datasets. The cohorts highlighted in red are the ovarian cancer single-cell datasets.</p>
Full article ">Figure 6
<p>Knockout GRB7 inhibits the proliferation of OVCAR3. (<b>A</b>) Western blot analysis of GRB7 knockout efficiency. (<b>B</b>) Colony formation capacity of GRB7 knockout and control. (<b>C</b>) CCK-8 assay of GRB7 knockout and control. Data are represented as mean ± standard deviation (SD) (<b>A</b>–<b>C</b>). The Shapiro–Wilk test confirmed normality, and Brown–Forsythe test confirmed homogeneity of variance (<b>B</b>,<b>C</b>). * <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 by one-way ANOVA (<b>B</b>) and two-way ANOVA (<b>C</b>). Data are representative of three independent experiments (<b>B</b>,<b>C</b>).</p>
Full article ">Figure 7
<p>GRB7 knockout in OVCAR3 inhibits cell migration and sensitizes killing effect of CD8+ T cells. (<b>A</b>) GRB7 knockout in OVCAR3 reduces migrating cell numbers in transwell assay. (<b>B</b>) GRB7 knockout in OVCAR3 slows wound healings. (<b>C</b>) The representative FACS results and summary of the log2 fold change in the ratio of GRB7 KO cells over the control after adding CD8+ T cells. The pseudocolor in the figure represents the variation in cell density. Colors range from red to blue, indicating a gradual decrease in cell density from high to low. Data are represented as mean ± SD (<b>A</b>–<b>C</b>). The Shapiro–Wilk test confirmed normality, and Brown–Forsythe test confirmed homogeneity of variance (<b>A</b>–<b>C</b>). ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001 by one-way ANOVA (<b>A</b>–<b>C</b>). Data are representative of three independent experiments (<b>A</b>–<b>C</b>).</p>
Full article ">
15 pages, 3709 KiB  
Article
Superhydrophobicity Effects on Spheroid Formation and Polarization of Macrophages
by María del Carmen Morán, Francesca Cirisano and Michele Ferrari
Pharmaceuticals 2024, 17(8), 1042; https://doi.org/10.3390/ph17081042 - 7 Aug 2024
Viewed by 246
Abstract
The interaction of biomaterials with the immune system is ruled by the action of macrophages. The surface features of these biomaterials, like wettability, which is an expression of chemical composition, texture, and geometry, can affect macrophages response. Such surface parameters can be then [...] Read more.
The interaction of biomaterials with the immune system is ruled by the action of macrophages. The surface features of these biomaterials, like wettability, which is an expression of chemical composition, texture, and geometry, can affect macrophages response. Such surface parameters can be then efficiently exploited to improve biocompatibility by lowering undesired immunological reactions and at the same time creating the substrate for positive interactions. In this work, the preparation and physicochemical characterization of highly water-repellent surfaces to develop and characterize 3D spheroids derived from monocyte-macrophages (RAW 264.7 cell line) has been carried out. As a measure of cell viability over time, the obtained aggregates have been transferred under standard 2D cell culture conditions. Significant changes on the morphology-associated polarization of the derived cellular entities have been evaluated at the nanoscale through 3D profilometry. The results suggested that the spheroid formation using highly repellent substrates induced the activation of M2-type cells. This simple and cost-effective approach can be used for preparing M2-based macrophages for regenerative purposes. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Figure 1

Figure 1
<p>Three-dimensional detail of the superhydrophobic surface (<b>a</b>) and its roughness acquired by interferometric and confocal profilometry at 20× (<b>b</b>). Average roughness, Sa, 170 nm.</p>
Full article ">Figure 2
<p>Phase contrast images on RAW264.7 cells cultured in TCPS, agarose, and highly water-repellent surfaces (SHS) after 48 h of incubation based on initial cell density (200 or 2000 cel/μL) (<b>a</b>). Circularity and size distribution (<b>b</b>) based on initial cell density (200 or 2000 cel/μL) on agarose and SHS. The results are reported as the average of more than 10 individual spheroids ± standard deviation. * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.001 indicate significant differences between values as a function of the initial cell density for the same substrate. • <span class="html-italic">p</span> &lt; 0.05 indicates significant differences between values as a function of the substrate for the same initial cell density.</p>
Full article ">Figure 3
<p>Phase contrast images of the temporal evolution) of cells derived from SHS-induced spheroids. Representative morphology of migrated cells at 168 h after recovery as a function of the initial cell density (200 or 2000 cel/μL). The scale bar represents 200 μm.</p>
Full article ">Figure 4
<p>Three-dimensional optical profilometry images in confocal mode (100× magnification) for RAW264.7 spheroids formed on SHS surfaces depending on the initial cell density (200 or 2000 cel/μL) for 168 h after recovery and compared to control cells. The scale bar represents 10 μm.</p>
Full article ">
Back to TopTop