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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (416)

Search Parameters:
Keywords = human macrophage cell line

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
24 pages, 12136 KB  
Article
BGN Secreted by Cancer-Associated Fibroblasts Promotes Esophageal Squamous Cell Carcinoma Progression via Activation of TLR4-Mediated Erk and NF-κB Signaling Pathways
by Hiroki Yokoo, Yu-ichiro Koma, Naozane Nomura, Rikuya Torigoe, Masaki Omori, Takashi Nakanishi, Shoji Miyako, Takaaki Nakanishi, Takayuki Kodama, Manabu Shigeoka, Yoshihiro Kakeji and Masafumi Horie
Int. J. Mol. Sci. 2025, 26(24), 12024; https://doi.org/10.3390/ijms262412024 - 13 Dec 2025
Viewed by 511
Abstract
Esophageal squamous cell carcinoma (ESCC) is associated with poor prognosis due to aggressive invasion and therapy resistance. Cancer-associated fibroblasts (CAFs) are key stromal components that promote tumor progression; however, their specific roles in ESCC remain unclear. Using a direct co-culture model of ESCC [...] Read more.
Esophageal squamous cell carcinoma (ESCC) is associated with poor prognosis due to aggressive invasion and therapy resistance. Cancer-associated fibroblasts (CAFs) are key stromal components that promote tumor progression; however, their specific roles in ESCC remain unclear. Using a direct co-culture model of ESCC cell lines (TE-9, -10, and -15) and mesenchymal stem cells (MSCs) to generate CAF-like cells, we identified biglycan (BGN) as a significantly upregulated gene in CAF-like cells via cDNA microarray analysis. Public single-cell RNA sequencing data also demonstrated elevated BGN expression in CAF clusters. We confirmed that CAF-like cells exhibited elevated BGN expression and secretion at both the mRNA and protein levels. Recombinant human BGN enhanced ESCC cell proliferation and migration by activating Erk and NF-κB signaling pathways, effects abrogated by TLR4 blockade. Furthermore, BGN promoted CAF marker expression in MSCs, M2-like macrophage polarization, and enhanced proliferation and migration abilities in both cell types. Immunohistochemical analysis of 66 ESCC tissues revealed that high stromal BGN expression correlated with greater tumor invasion, lymphatic invasion, and shorter disease-free survival. These findings indicate that CAF-derived BGN promotes ESCC progression via TLR4-mediated signaling and modulates stromal cell behavior, highlighting its potential as a prognostic biomarker and therapeutic target. Full article
Show Figures

Figure 1

15 pages, 1952 KB  
Article
Epithelial–Macrophage Crosstalk in Host Responses to Campylobacter jejuni Infection in Humans
by Khaled Abdelaziz, Shreeya Sharma, Mostafa Naguib and Alexis Stamatikos
Microorganisms 2025, 13(12), 2808; https://doi.org/10.3390/microorganisms13122808 - 10 Dec 2025
Viewed by 380
Abstract
Interactions between Campylobacter jejuni and host immune cells have been studied using various single-cell line models, such as macrophages and intestinal epithelial cells; however, these single-cell approaches do not fully capture the complexity of the host response. Investigating the interactions between these cell [...] Read more.
Interactions between Campylobacter jejuni and host immune cells have been studied using various single-cell line models, such as macrophages and intestinal epithelial cells; however, these single-cell approaches do not fully capture the complexity of the host response. Investigating the interactions between these cell types offers a more comprehensive model for understanding Campylobacter–host dynamics. Therefore, this study aimed to investigate these interactions, specifically between intestinal epithelial cells and macrophages, using an in vitro model of C. jejuni infection. We examined whether soluble factors secreted from C. jejuni-infected HT-29 cells (human colorectal adenocarcinoma cells that express characteristics of mature intestinal cells) at 10 and 50 multiplicities of infection (MOI) influence RAW 264.7 macrophage activity, including nitric oxide (NO) production, migration, phagocytosis, bacterial killing, and the expression of cytokines (IL-6, IL-1β, TNF-α) and the chemokine CCL2. C. jejuni infection of HT-29 cells at 10 MOI induced significant IFN-γ production, a key macrophage activator. The treatment of macrophages with supernatants from HT-29 cells infected with C. jejuni significantly increased NO production, enhanced migration and phagocytic activity, and increased IL-6, TNF-α and CCL2 gene expression. However, no significant killing of phagocytosed C. jejuni was observed. On the other hand, supernatants from HT-29 cells infected with 50 MOI of C. jejuni suppressed NO production and macrophage phagocytosis, which may explain individual variations in the immune system’s ability to contain infection, potentially influenced by the infectious dose. These findings support the notion that Campylobacter can evade macrophage killing even under activated conditions. Further studies are needed to elucidate the molecular mechanisms by which Campylobacter survives within activated macrophages. Full article
(This article belongs to the Special Issue Breaking Barriers: The Breakdown of Epithelial Defenses by Pathogens)
Show Figures

Figure 1

11 pages, 7223 KB  
Case Report
Primary Pericardial Well-Differentiated Papillary Mesothelioma in a Spotted Hyena (Crocuta crocuta)
by Louise van der Weyden, Dewald Keet and Nicolize O’Dell
Vet. Sci. 2025, 12(12), 1170; https://doi.org/10.3390/vetsci12121170 - 9 Dec 2025
Viewed by 197
Abstract
There have been few reports of neoplasia in hyenas to date. In this report, we describe a captive adult female spotted hyena (Crocuta crocuta) that developed inappetence, lethargy and marked abdominal distension over a 3-day period. The hyena was chemically immobilised [...] Read more.
There have been few reports of neoplasia in hyenas to date. In this report, we describe a captive adult female spotted hyena (Crocuta crocuta) that developed inappetence, lethargy and marked abdominal distension over a 3-day period. The hyena was chemically immobilised to allow clinical investigation of the severe symptoms; however, she died before any internal examination occurred. At necropsy, severe serosanguinous hydropericardium was evident, as well as pulmonary congestion and oedema, ascites and chronic passive congestion of the liver with mild fibrosis. Histopathological examination of the pericardial surface revealed fibrous proliferations lined by mostly a single layer of large proliferating neoplastic mesothelial cells forming papillary projections into the lumen of the pericardial sac as well as infiltration into the pericardial connective tissue, with innumerable haemosiderin-laden macrophages in places, suggestive of chronic haemorrhage. The liver revealed severe congestion and interstitial fibrosis, and the lung revealed congestion and oedema, with moderate numbers of alveolar macrophages and marked anthracosis. The diagnosis was pericardial well-differentiated papillary mesothelioma, with death under anaesthesia caused by cardiogenic shock due to pericardial mesothelioma-associated cardiac tamponade. As primary pericardial mesothelioma (PPM) is a rare tumour type for both animals and humans, and this is the first report of a PPM in a hyena, we compare the clinical findings with those seen in other species. Full article
(This article belongs to the Section Anatomy, Histology and Pathology)
Show Figures

Figure 1

27 pages, 9480 KB  
Article
The Anti-EMMPRIN Monoclonal Antibody hMR18-mAb Induces Tumor Dormancy and Inhibits the EMT Process in Human Carcinoma Cell Lines Co-Cultured with Macrophages
by Elina Simanovich, Felix Oyelami, Phillipp Brockmeyer and Michal A. Rahat
Biomedicines 2025, 13(12), 2950; https://doi.org/10.3390/biomedicines13122950 - 30 Nov 2025
Viewed by 429
Abstract
Background: The epithelial-to-mesenchymal transition (EMT) process is necessary for metastasis as it enables tumor cells’ migration and invasion. In the remote organ, tumor cells can develop into metastatic lesions or arrest their proliferation and become dormant, thus suspending metastatic development. EMMPRIN is [...] Read more.
Background: The epithelial-to-mesenchymal transition (EMT) process is necessary for metastasis as it enables tumor cells’ migration and invasion. In the remote organ, tumor cells can develop into metastatic lesions or arrest their proliferation and become dormant, thus suspending metastatic development. EMMPRIN is a membrane glycoprotein, implicated in cell–cell interactions, proliferation, angiogenesis, and EMT. We asked whether neutralizing EMMPRIN with the new anti-EMMPRIN monoclonal antibody hMR18-mAb can inhibit EMT. Methods: We co-cultured tumor cell lines (breast carcinoma MCF-7, MDA-MB-231, or oral squamous cell carcinoma SCC-40) together with U937 monocytic-like cells, with or without hMR18-mAb or its negative control rabbit IgG. Results: We demonstrate that depending on the initial state of the cells along the epithelial–mesenchymal axis (E/M axis), co-culture enhanced the EMT process, whereas hMR18-mAb reversed this effect. The co-culture increased EMT-inducer cytokines in all cell lines (by 2.5-fold), while hMR18-mAb reduced them (by ~55–70% in the breast cancer cells and by 81% in the SCC-40 cells). The co-culture reduced E-cadherin (by 2-fold in MCF-7 and SCC-40 cells) and increased vimentin expression (by 2–3-fold in MDA-MB-231 and SCC-40), while hMR18-mAb reverted this effect. Co-culture enhanced proliferation, migration, and angiogenic potential of the tumor cells, while hMR18-mAb reduced these by ~20%, 30–44% and ~60–80%, respectively. Co-culture reduced the standard markers of dormancy (NR2F1, p21, p27) and stemness (SOX2, Nanog) (by 30–60% in MCF-7 and SCC-40), while hMR18-mAb elevated gene expression of these markers (by 1.5–3.5-fold) in all cell lines, pushing the cells towards dormancy. Conclusions: We conclude that EMMPRIN is a gatekeeper that prevents cells from entering dormancy, and that hMR18-mAb disrupts this effect. As it is the first antibody shown to induce dormancy in tumor cells and stop the development of metastases, this could become a new therapeutic strategy to prevent and treat metastasis. Full article
(This article belongs to the Section Cancer Biology and Oncology)
Show Figures

Graphical abstract

23 pages, 4665 KB  
Article
Preclinical Mechanistic Evaluation of Hyaluronan/Niacinamide (Vitamin B3) Hydrogels: Toward an Enhanced Viscosupplement System with Ancillary Anti-Arthritic Attributes
by Farid Hadjab, Stivens Antoine, Béatrice Hamel, Mohamed Benderdour, Hassan Fahmi, Alexandre Porcello, Virginie Philippe, Robin Martin, Cíntia Marques, Kelly Lourenço, Corinne Scaletta, Nathalie Hirt-Burri, Philippe Abdel-Sayed, Lee Ann Applegate and Alexis E. Laurent
Bioengineering 2025, 12(11), 1246; https://doi.org/10.3390/bioengineering12111246 - 14 Nov 2025
Viewed by 855
Abstract
Osteoarthritis (OA), a degenerative joint disease primarily affecting the hips and knees, is characterized by multifactorial dysregulation of chondrocyte homeostasis and currently lacks curative treatment options. Intra-articular hyaluronic acid (HA) injections have clinically provided symptomatic relief for three decades; however, HA’s rapid in [...] Read more.
Osteoarthritis (OA), a degenerative joint disease primarily affecting the hips and knees, is characterized by multifactorial dysregulation of chondrocyte homeostasis and currently lacks curative treatment options. Intra-articular hyaluronic acid (HA) injections have clinically provided symptomatic relief for three decades; however, HA’s rapid in vivo degradation by free radicals and hyaluronidases limits its efficacy. We hypothesized that adding niacinamide (vitamin B3) to linear HA hydrogels would provide ancillary anti-inflammatory and anti-catabolic properties, thereby improving HA-based viscosupplementation therapy. This preliminary preclinical mechanistic study investigated the functional effects of incorporating niacinamide into linear HA-based hydrogels using in vitro cellular models. Initially, Raw 264.7 macrophages and C28/I2 or SW1353 human chondrocytes were pre-treated with varying concentrations of HA/B3, with or without lipopolysaccharide (LPS) or interleukin-1β (IL-1β), respectively. Subsequently, pro-inflammatory and pro-catabolic markers were quantified biochemically. Results demonstrated that HA/B3 hydrogels exhibited enhanced functional stability compared to HA alone and possessed significant anti-inflammatory and anti-catabolic properties, without inducing cytotoxicity in either cell line. In Raw 264.7 macrophages, HA/B3 inhibited LPS-induced tumor necrosis factor-α (TNF-α) release and suppressed cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) protein expression. In vitro, HA/B3 hydrogels reduced IL-1β-induced IL-6 production in primary chondrocytes by 16% and suppressed PGE2 concentration in both macrophages and chondrocytes by 60%, effects superior to HA alone. Finally, a rat primary articular chondrocyte model suggested slight anti-hypertrophic effects of HA/B3 in vitro. Collectively, these findings suggest that HA/B3 hydrogels possess anti-arthritic potential, highlighting a novel strategy for next-generation viscosupplement systems. Full article
Show Figures

Figure 1

16 pages, 4973 KB  
Article
The Apoptosis Induction and Immunomodulating Activities of Nga-Kee-Mon (Perilla frutescens) Seed Extract
by Pongsathorn Dhumtanom, Anurak Wongta and Wantida Chaiyana
Foods 2025, 14(21), 3685; https://doi.org/10.3390/foods14213685 - 29 Oct 2025
Cited by 1 | Viewed by 695
Abstract
Perilla frutescens, “Nga-Kee-Mon” in Thai, is a high-nutritional-value plant. This study aims to identify the phytochemicals, apoptosis induction and immunomodulating activities of the perilla seed extract (PSE) and highlight the high pharmaceutical value of perilla. The phytochemical profile of PSE was characterized [...] Read more.
Perilla frutescens, “Nga-Kee-Mon” in Thai, is a high-nutritional-value plant. This study aims to identify the phytochemicals, apoptosis induction and immunomodulating activities of the perilla seed extract (PSE) and highlight the high pharmaceutical value of perilla. The phytochemical profile of PSE was characterized using HPLC. Antioxidant capacity was studied using DPPH assay. Apoptosis was confirmed by morphological changes and DNA fragmentation of the human colon adenocarcinoma cell line (HT-29). Immunomodulating activity was studied in an LPS-stimulated murine macrophage cell line (RAW 264.7). PSE had high levels of TPC (375.04 ± 11.45 mg GAE/g) and TFC (223.45 ± 16.02 mg QE/g) with strong radical scavenging capacity (312.87 ± 12.98 mg TE/100 g). Rosmarinic acid (0.116 g%) and luteolin (0.010 g%) were the major phytochemicals. PSE at 50 µg/mL, equivalent to 0.85 and 0.08 µg/mL of rosmarinic acid and luteolin, respectively, caused morphological alterations and DNA fragmentation within 24 h. PSE at 200 µg/mL, equivalent to 3.38 and 0.30 µg/mL of rosmarinic acid and luteolin, respectively, had significant inhibitory activity on IL-1β, IL-6, and TNF-α secretion. These results demonstrate that PSE has high antioxidant capacity, with rosmarinic acid and luteolin as the major phytochemicals. It can trigger apoptosis in HT-29 cells and has immunomodulatory effects. These findings highlight the potential of perilla seed extract as a promising natural source for therapeutic applications related to oxidative stress, cancer prevention, and immune modulation. Full article
(This article belongs to the Special Issue Polyphenols and Health Benefits: 2nd Edition)
Show Figures

Figure 1

16 pages, 2928 KB  
Article
Immunomodulatory Potential of a Composite Amniotic Membrane Hydrogel for Wound Healing: Effects on Macrophage Cytokine Secretion
by Tao Wang, Zhiyuan Zhu, Wei Hua and Siliang Xue
Biomedicines 2025, 13(10), 2574; https://doi.org/10.3390/biomedicines13102574 - 21 Oct 2025
Cited by 1 | Viewed by 804
Abstract
Background: The human acellular amniotic membrane (HAAM) is widely used as a decellularized bioscaffold in tissue engineering to promote wound healing, but its clinical application is limited by poor mechanical properties, rapid degradation, and handling difficulties. This study aimed to develop a modified [...] Read more.
Background: The human acellular amniotic membrane (HAAM) is widely used as a decellularized bioscaffold in tissue engineering to promote wound healing, but its clinical application is limited by poor mechanical properties, rapid degradation, and handling difficulties. This study aimed to develop a modified amniotic membrane-based composite material loaded with vascular endothelial growth factor (VEGF) and the Notch signaling inhibitor N-[N-(3,5-difluorophenacetyl)-Lalanylhydrazide]-Sphenylglycine t-butyl ester (DAPT) to enhance wound healing by modulating macrophage polarization and cytokine secretion. Methods: VEGF-loaded gellan gum-hyaluronic acid (GG-HA) hydrogels (VEGF-GG-HA) and DAPT-loaded HAAM (DAPT-HAAM) were prepared and combined to form a novel composite material (VEGF-GG-HA & DAPT-HAAM). The morphology and microstructure of the materials were characterized using scanning electron microscopy. In vitro studies were conducted using the human monocytic cell line (Tohoku Hospital Pediatrics-1, THP-1) to evaluate the effects of the materials on cell viability, cytokine secretion, and protein expression. Assessments included CCK-8 assays, ELISA, quantitative real-time PCR, Western blot analysis, and immunohistochemical staining. Results: The composite material VEGF-GG-HA & DAPT-HAAM exhibited good biocompatibility and significantly promoted THP-1 cell proliferation compared to control and single-component groups. It enhanced the secretion of IL-10, TNF-α, TGF-β, MMP1, and MMP3, while suppressing excessive TGF-β overexpression. The material also modulated macrophage polarization, showing a trend toward anti-inflammatory M2 phenotypes while maintaining pro-inflammatory signals (e.g., TNF-α) for a balanced immune response. Conclusions: The modified amniotic membrane hydrogel composite promotes wound healing through a phased immune response: it modulates macrophage polarization (balancing M1 and M2 phenotypes), enhances cytokine and matrix metalloproteinase secretion, and controls TGF-β levels. These effects contribute to improved vascular remodeling, reduced fibrosis, and prevention of scar formation, demonstrating the potential for enhanced wound management. Full article
(This article belongs to the Special Issue New Advances in Wound Healing and Skin Regeneration)
Show Figures

Figure 1

19 pages, 4150 KB  
Article
Cisplatin-Loaded M1 Macrophage-Derived Vesicles Have Anti-Cancer Activity in Osteosarcoma
by Namrata Anand, Joseph Robert McCorkle, David S. Schweer, Lan Li, Kristen S. Hill, Melissa A. Fath, Derek B. Allison, Christopher L. Richards and Jill M. Kolesar
Cells 2025, 14(20), 1616; https://doi.org/10.3390/cells14201616 - 17 Oct 2025
Cited by 1 | Viewed by 2902
Abstract
Osteosarcoma (OS) is a relatively rare bone malignancy that primarily affects children and young adults and is associated with significant morbidity and mortality. Cisplatin is a mainstay of treatment, but its efficacy is limited by off-target toxicities. Immunotherapy is not effective due to [...] Read more.
Osteosarcoma (OS) is a relatively rare bone malignancy that primarily affects children and young adults and is associated with significant morbidity and mortality. Cisplatin is a mainstay of treatment, but its efficacy is limited by off-target toxicities. Immunotherapy is not effective due to a poor antigenic tumor microenvironment. Here, we address these challenges by using manufactured M1 macrophage-derived vesicles (MVs) loaded with cisplatin. Human blood and mouse RAW 264.7 M1 macrophages were used to prepare empty (E-MVs) and cisplatin-loaded MVs (C-MVs). Human OS cell lines were used in vitro and in a tibia xenograft mouse model to evaluate the anti-cancer and immune-stimulating abilities of MVs. C-MVs had lower IC50s but equivalent DNA damage in OS cell lines when compared with free cisplatin. E-MVs and C-MVs were observed to accumulate in the tumor in OS tumor-bearing mice. C-MVs significantly reduced tumor burden and prolonged survival in a mouse model of OS. Animals dosed with free cisplatin experienced weight loss and renal and hepatic toxicity, while equivalent doses of C-MVs did not cause these effects. In addition, both E-MVs and C-MVs showed immunomodulation of the tumor microenvironment with a significant increase in the M1/M2 macrophages ratio (7-fold and 22-fold, respectively) and increased levels of TNF-α in serum (1.8-fold and 2.1-fold, respectively) compared to control mice. Collectively, these experiments support further development of C-MVs for the treatment of OS. Full article
Show Figures

Graphical abstract

16 pages, 8320 KB  
Article
Bactericidal and Anti-Inflammatory Effects of Ashitaba-Extract Ameliorate the Gingivitis and Halitosis in Dogs with Porphyromonas gulae-Infected Periodontal Disease
by Takayoshi Miyamoto, So Shirahata, Mariko Komuro, Mao Kaneki, Chiharu Ohira and Tomoki Fukuyama
Vet. Sci. 2025, 12(10), 981; https://doi.org/10.3390/vetsci12100981 - 13 Oct 2025
Viewed by 924
Abstract
Ashitaba (Angelica keiskei) is a perennial herb native to Japan, traditionally consumed as a health-promoting food and herbal medicine. This study evaluated the antimicrobial, anti-halitosis, and anti-inflammatory effects of Ashitaba extract on canine periodontal disease (PD) caused by Porphyromonas gulae ( [...] Read more.
Ashitaba (Angelica keiskei) is a perennial herb native to Japan, traditionally consumed as a health-promoting food and herbal medicine. This study evaluated the antimicrobial, anti-halitosis, and anti-inflammatory effects of Ashitaba extract on canine periodontal disease (PD) caused by Porphyromonas gulae (P. gulae). In vitro, Ashitaba extract (0.006–0.1%) significantly inhibited P. gulae viability by up to 80% and reduced biofilm formation by approximately 10% at 0.1%. The extract also suppressed the production of volatile sulfur compounds—hydrogen sulfide and methyl mercaptan—by over 80% and 40%, respectively, within 10 min. Furthermore, Ashitaba extract markedly decreased P. gulae-induced pro-inflammatory cytokine secretion (IL-1β, IL-6, TNF-α) by up to 90% in murine, canine, and human macrophage and gingival cell lines. In vivo, daily oral application of 0.05% Ashitaba-extract gel for four weeks, with or without tooth brushing, significantly improved gingivitis scores (by 40–60%), reduced halitosis levels, and decreased P. gulae DNA detection and enzymatic activity in dogs with PD. These findings demonstrate that Ashitaba extract possesses potent bactericidal, anti-halitosis, and anti-inflammatory properties, supporting its potential as a natural adjunctive therapy for the prevention and management of canine periodontal disease. Full article
(This article belongs to the Section Veterinary Microbiology, Parasitology and Immunology)
Show Figures

Graphical abstract

11 pages, 3360 KB  
Article
Loss of Type 1 Pili and Flagella in Uropathogenic Escherichia coli Leads to Reduced Phagocytosis by Human and Murine Monocytes
by William R. Schwan
Pathogens 2025, 14(10), 968; https://doi.org/10.3390/pathogens14100968 - 25 Sep 2025
Viewed by 982
Abstract
Background: Uropathogenic Escherichia coli (UPEC) is the number one cause of urinary tract infections (UTIs) in humans. The ability to bind to uroepithelial cells through type 1 pili and ascend the urinary tract via flagella is important in the early stages of a [...] Read more.
Background: Uropathogenic Escherichia coli (UPEC) is the number one cause of urinary tract infections (UTIs) in humans. The ability to bind to uroepithelial cells through type 1 pili and ascend the urinary tract via flagella is important in the early stages of a UTI. However, both type 1 pili and flagella can also target the bacteria for elimination via monocytes/macrophages later in a UTI. We hypothesized that the loss of both type 1 pili and flagella on the UPEC cells would make them less likely to be phagocytized by phagocytic cells. Methods: In this study, ΔfimA, ΔfliC, and ΔfimA ΔfliC mutants were compared to the wild type UPEC strain NU149 in phagocytosis assays using human and murine monocytic cell lines. Results: A ΔfimA ΔfliC double mutant was phagocytized significantly less than the wild type strain. Conclusion: The data show that the loss of both type 1 pili and flagella expression on the UPEC cells reduces phagocytosis of the bacteria by human and murine monocytes. Although type 1 pili and flagella are important for establishing a UTI and ascension into the kidneys, the loss of these proteinaceous structures may allow the UPEC cells to evade the innate immune defenses in certain environments within the human body. Full article
Show Figures

Figure 1

22 pages, 9932 KB  
Article
A Novel Serum-Free Triculture Model of Glioblastoma, Astrocytes, and Macrophages
by Hasan Alrefai, Lauren C. Nassour-Caswell, Manoj Kumar, Benjamin Lin, Taylor L. Schanel, Nicholas J. Eustace, Jianqing Zhang, Christian T. Stackhouse, Nayonika Mukherjee, Patricia H. Hicks, Joshua C. Anderson, Christopher Ryan Miller and Christopher D. Willey
Int. J. Mol. Sci. 2025, 26(19), 9335; https://doi.org/10.3390/ijms26199335 - 24 Sep 2025
Viewed by 1029
Abstract
Glioblastoma (GBM) is the most common and deadly primary brain tumor in adults. While in vitro patient-derived xenografts (PDX) lines are useful for studying GBM, they often exclude astrocytes and macrophages, which contribute significantly to tumor growth, invasion, and chemoradioresistance. Integrating these cells [...] Read more.
Glioblastoma (GBM) is the most common and deadly primary brain tumor in adults. While in vitro patient-derived xenografts (PDX) lines are useful for studying GBM, they often exclude astrocytes and macrophages, which contribute significantly to tumor growth, invasion, and chemoradioresistance. Integrating these cells into tumor models is difficult due to their need for serum, which triggers GBM-PDX lines to lose their stem-like properties. The aim of this study was to develop a serum-free triculture model of GBM-PDX lines, normal human astrocytes (NHAs), and macrophages. Serum-free media alternatives were formulated for NHAs and identified for THP-1 macrophages, then combined with GBM PDX media to establish “PSX,” an experimental maintenance media. Cells were transitioned to serum-free media alternatives and functionally assessed through several parameters unique to each cell type. In addition to assessing GBM “stemness,” a custom 350-gene NanoString chip was used to assess differential gene expression in monocultured PDX cells versus PDX cells exposed to NHAs and macrophages. PSX maintained canonical function in astrocytes and macrophages while preserving the stem-like properties of GBM-PDX cells. Tri-culturing all three cells increased the expression of stemness-associated transcription factors and increased the expression of genes related to stemness and hypoxia in GBM cells. GBM PDX cells exposed to NHAs and macrophages in direct triculture exhibit increases in markers of stemness and hypoxia. These findings suggest that the serum-free triculture model presented herein may better recapitulate the tumoral heterogeneity of GBM in vitro, providing a novel model to utilize in current research. Full article
(This article belongs to the Special Issue Molecular Insights into Glioblastoma Pathogenesis and Therapeutics)
Show Figures

Figure 1

21 pages, 7427 KB  
Article
Effect of Oral Peritumoral Tissue on Infiltration and Differentiation of Tumor-Associated Macrophages in Oral Squamous Cell Carcinoma
by Tianyan Piao, Kiyofumi Takabatake, Takuma Arashima, Yulu Zhao, Hotaka Kawai, Htoo Shwe Eain, Yamin Soe, Zin Zin Min, Keisuke Nakano and Hitoshi Nagatsuka
Cells 2025, 14(18), 1481; https://doi.org/10.3390/cells14181481 - 22 Sep 2025
Viewed by 772
Abstract
The recruitment of tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) of oral squamous carcinoma (OSCC) affects significant cancer invasion; however, in the normal host tissue that is located in the cancer’s surrounding area, this is poorly investigated. In this study, we examined [...] Read more.
The recruitment of tumor-associated macrophages (TAMs) in the tumor microenvironment (TME) of oral squamous carcinoma (OSCC) affects significant cancer invasion; however, in the normal host tissue that is located in the cancer’s surrounding area, this is poorly investigated. In this study, we examined the impact of gingival connective tissue cells (GCTCs) and periodontal ligament cells (PDLCs), which are involved in the invasive pathway of OSCC, on oral cancer invasion via TAMs recruitment. Transwell (migration) assays were used to examine the effects of GCTCs and PDLCs on the migration of macrophages, which indicated that the interaction between GCTCs and HSC-2/HSC-3 (human oral squamous cell carcinoma cell line) promoted the recruitment of macrophages, whereas the interaction between PDLCs was inhibited. An indirect co-culture was then used to examine the effects of GCTCs and PDLCs on the differentiation of macrophages, which indicated that the interaction between GCTCs enhanced their ability to transform into M2-type macrophages. Furthermore, the effects of GCTCs and PDLCs on the recruitment of CD45(+) monocytes, F4/80(+) M0 macrophages, iNOS(+) M1 macrophages, and CD163(+) M2 TAMs were assayed by immunohistochemistry. The results revealed that the interaction between GCTCs and HSC-2/HSC-3 promoted the infiltration of CD45(+) monocytes, F4/80(+) M0 macrophages, and CD163(+) M2 TAMs, whereas the PDLCs inhibited it, while their effect on iNOS(+) M1 macrophages was limited. Collectively, the GCTCs contributed to the infiltration of TAMs into the TME of OSCC cells, whereas the PDLCs exerted an inhibitory effect. These findings suggest a potential regulatory mechanism underlying the progression of OSCC. Full article
Show Figures

Figure 1

25 pages, 3830 KB  
Article
Lipoprotein(a)/CD36 Interaction Drives IL-6/RhoA-GTP Signaling and miRNA Epigenetic Regulation in Coronary Artery Spasm
by Yen-Kuang Lin, Tsung-Han Hsieh, Chi-Tai Yeh, Vijesh Kumar Yadav, Iat-Hang Fong, Kuang-Tai Kuo, Nicholas G. Kounis, Patrick Hu and Ming-Yow Hung
Pharmaceuticals 2025, 18(9), 1384; https://doi.org/10.3390/ph18091384 - 16 Sep 2025
Viewed by 899
Abstract
Background: Lipoprotein(a) [Lp(a)]-induced inflammation contributes to coronary artery spasm (CAS) by the contraction of vascular smooth muscle cells. However, the interaction between Lp(a) and soluble CD36 (sCD36)/interleukin (IL)-6/RAS Homolog Family Member A (RhoA)-GTP signaling pathway has not been evaluated. Methods: We investigated the [...] Read more.
Background: Lipoprotein(a) [Lp(a)]-induced inflammation contributes to coronary artery spasm (CAS) by the contraction of vascular smooth muscle cells. However, the interaction between Lp(a) and soluble CD36 (sCD36)/interleukin (IL)-6/RAS Homolog Family Member A (RhoA)-GTP signaling pathway has not been evaluated. Methods: We investigated the relevance of Lp(a)/CD36 signaling in CAS patient monocyte-derived macrophages (PMDMs) and a human coronary artery smooth muscle cell (HCASMC) line using expression profile correlation analyses, molecular docking, RNA sequencing, flow cytometry, immunoblotting, and quantitative reverse transcription polymerase chain reaction. Results: Plasma Lp(a) and sCD36 levels in 41 CAS patients were significantly higher (p = 0.001) and positively correlated (r2 = 0.3145, p < 0.001), a trend not observed in 36 non-CAS controls. RNA sequencing indicated a significant co-overexpression of CD36 and RhoA in Lp(a)-treated CAS PMDMs and HCASMCs, of which the mRNA and protein expression of CD36 and RhoA were significantly enhanced (p < 0.001) dose-dependently. Lp(a) rather than LDL preferentially induced CD80+ PMDM (M1) polarization. In HCASMCs, the CD36 knockdown using either short hairpin RNA or natural biflavonoid amentoflavone suppressed Lp(a)-upregulated protein expression of CD36, RhoA-GTP, IL-6, tumor necrosis factor (TNF)-α, nuclear factor (NF)-κB, and CD80; however, overexpressed CD36 increased their levels. Lp(a) decreased and amentoflavone increased the epigenetic expression of CD36 inhibitors, miR-335-5p, and miR-448, respectively. Reciprocally, an miRNA inhibitor or mimic could magnify or diminish Lp(a)-induced CD36, TNF-α, NF-κB and IL-6 expressions in HCASMCs, respectively. Conclusions: Elevated Lp(a) levels upregulate the CD36-dependent TNF-α/NF-κB/IL-6/RhoA-GTP signaling pathway in CAS PMDMs and HCASMCs, indicating that Lp(a)/CD36 inflammatory signaling, HCASMC activation, and macrophage M1 polarization mediate CAS development. Full article
(This article belongs to the Section Biopharmaceuticals)
Show Figures

Graphical abstract

21 pages, 2688 KB  
Article
Single- vs. Multi-Walled Carbon Nanotubes: Differential Cellular Stress and Lipid Metabolism Effects in Macrophage Models
by Sara Nahle, Hilary Cassidy, David Matallanas, Bertrand H. Rihn, Olivier Joubert and Luc Ferrari
Nanomaterials 2025, 15(18), 1401; https://doi.org/10.3390/nano15181401 - 11 Sep 2025
Cited by 1 | Viewed by 679
Abstract
This study examines the toxicological effects of carbon nanotubes (CNTs) of different diameters—single-walled CNTs (SWCNT, 2 nm) and multi-walled CNTs (MWCNT, 74 nm)—on two macrophage cell lines, rat alveolar NR8383 cells and human differentiated THP-1. Using standardized exposure conditions and employing an integrated [...] Read more.
This study examines the toxicological effects of carbon nanotubes (CNTs) of different diameters—single-walled CNTs (SWCNT, 2 nm) and multi-walled CNTs (MWCNT, 74 nm)—on two macrophage cell lines, rat alveolar NR8383 cells and human differentiated THP-1. Using standardized exposure conditions and employing an integrated omics approach (transcriptomic and proteomic analyses), both CNT types were found to induce cellular stress responses and inflammation, especially in NR8383 cells, with notable involvement of the Sirtuin signaling pathway. After 24 h, MWCNTs uniquely disrupted lipid metabolism in NR8383 cells, resulting in foam cell formation and syncytia. While SWCNTs were less disruptive to metabolic pathways, they significantly altered gene regulation, particularly RNA splicing mechanisms. The dispersion medium—fetal bovine serum (FBS) versus human surfactant—also modulated the observed toxicological responses, highlighting the critical role of the protein corona in influencing CNT-cell interactions. These findings demonstrate that CNT diameter significantly affects cytotoxicity and cellular response pathways in a cell-type-specific manner. Full article
(This article belongs to the Section Biology and Medicines)
Show Figures

Figure 1

28 pages, 3218 KB  
Systematic Review
Antioxidant Potential of Opuntia dillenii: A Systematic Review of Influencing Factors and Biological Efficacy
by Ruymán Santana-Farré, Nisa Buset-Ríos and Mussa Makran
Nutraceuticals 2025, 5(3), 22; https://doi.org/10.3390/nutraceuticals5030022 - 13 Aug 2025
Cited by 1 | Viewed by 1128
Abstract
Opuntia dillenii has gained considerable scientific attention as a potential natural source of antioxidants. This systematic review compiles and evaluates current evidence regarding its antioxidant activity. A PRISMA-guided literature search was conducted using PubMed, Scopus, and Web of Science, identifying 37 eligible studies. [...] Read more.
Opuntia dillenii has gained considerable scientific attention as a potential natural source of antioxidants. This systematic review compiles and evaluates current evidence regarding its antioxidant activity. A PRISMA-guided literature search was conducted using PubMed, Scopus, and Web of Science, identifying 37 eligible studies. These studies employed two main methodological approaches: chemical-based assays and biological models. Chemical assays, including radical scavenging and reducing power assays, demonstrated a broad range of antioxidant activity influenced by factors such as the extraction method, plant part, plant maturity, and geographic origin. Polysaccharides, betalains, and polyphenols were consistently identified as primary contributors to these effects. Biological models further supported the antioxidant properties of O. dillenii extracts. In animal studies, administration of the extracts significantly improved oxidative stress biomarkers, increasing glutathione levels, reducing malondialdehyde concentrations, and enhancing the activity of antioxidant enzymes, particularly in the liver and other digestive tissues like the colon, stomach, and pancreas. Cellular studies using hepatocyte, macrophage, enterocyte, and neuronal cell lines produced comparable results, confirming the antioxidant effects. In conclusion, O. dillenii exhibits promising antioxidant potential across various experimental models. However, the absence of human clinical trials highlights the need for further research to establish its efficacy and safety as a nutraceutical product. Full article
Show Figures

Figure 1

Back to TopTop