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

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (610)

Search Parameters:
Keywords = leukocyte receptors

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 638 KB  
Review
Mast Cells Accumulate in the Stroma of Breast Adenocarcinoma and Secrete Pro-Inflammatory Cytokines and Tumor-Damaging Mediators: Could IL-37 and IL-38 Play an Anti-Tumor Role?
by Pio Conti, Carla E. Gallenga, Ciro Annicchiarico, Armando Coppola, Raffaello Pellegrino, Michelangelo J. Conti and Filiberto Mastrangelo
Int. J. Mol. Sci. 2026, 27(2), 824; https://doi.org/10.3390/ijms27020824 - 14 Jan 2026
Viewed by 75
Abstract
Tumor tissue is surrounded by mast cells (MCs), which participate in the inflammatory immune response by producing cytokines, proteases, and other molecules. MCs are involved in both innate and acquired immunity and are associated with the IgE response through the FcεRI receptor. MCs [...] Read more.
Tumor tissue is surrounded by mast cells (MCs), which participate in the inflammatory immune response by producing cytokines, proteases, and other molecules. MCs are involved in both innate and acquired immunity and are associated with the IgE response through the FcεRI receptor. MCs mediate inflammation in several immune reactions, including acute hyperreactivity, leukocyte recruitment, acute tissue swelling, anaphylaxis, and pro-inflammatory cytokine production. They not only function as pro-inflammatory effector cells but may also contribute to the regulation of the acquired immune response in tumor tissue. Therefore, MCs may mediate immunity in breast cancer by promoting remodelling and counteracting cancer growth. They also produce anti-inflammatory substances, such as histamine, transforming growth factor-β (TGF-β)1, IL-10, and IL-4, which inhibit the acquired immune response and reduce the inflammatory state. IL-37 and IL-38 are novel natural regulators of inflammation and are anti-inflammatory members of the IL-1 family. IL-1, generated by immune cells such as macrophages and lymphocytes, is released downstream of oncogenes in breast cancer, triggering an inflammatory response by stimulating other pro-inflammatory cytokines such as IL-6, tumor necrosis factor (TNF), and IL-33 (an early warning cytokine). Therefore, blocking IL-1 with IL-37 or IL-38 could represent a novel therapeutic strategy that, when combined with other treatments, could be beneficial in breast cancer. This review focuses on the new discoveries and insights into the role of MCs in breast cancer. We also analyzed molecules that can promote tumor growth and those that can inhibit cancer development and metastasis. This review aims to study the role of MCs accumulated in the stroma of breast adenocarcinoma in relation to secreted anti-inflammatory cytokines, such as IL-37 and IL-38. Full article
(This article belongs to the Special Issue Cytokines and Inflammatory Diseases)
Show Figures

Figure 1

15 pages, 20677 KB  
Article
Immune Profiling the Axilla with Fine Needle Aspiration Is Feasible to Risk-Stratify Breast Cancer
by Jasmine A. Gore, Amy M. Llewellyn, Chuen Y. R. Lam, Jacqueline D. Shields and Kalnisha Naidoo
Cancers 2026, 18(2), 251; https://doi.org/10.3390/cancers18020251 - 14 Jan 2026
Viewed by 131
Abstract
Background: Axillary lymph node (ALN) metastasis is a critical prognostic determinant in breast cancer (BC) that informs surgical management. However, surgically clearing the axilla carries morbidity, so less invasive methods of risk-stratifying patients are needed. ALN fine needle aspiration (FNA) is currently [...] Read more.
Background: Axillary lymph node (ALN) metastasis is a critical prognostic determinant in breast cancer (BC) that informs surgical management. However, surgically clearing the axilla carries morbidity, so less invasive methods of risk-stratifying patients are needed. ALN fine needle aspiration (FNA) is currently used to detect BC metastases, but these samples also contain immune cells. Methods: Cells obtained via FNA from BC-patient-derived ALNs were analysed using flow cytometry. Results: FNA acquires sufficient leukocytes for comprehensive immunophenotyping of reactive, patient-derived ALNs. All CD4+ and CD8+ T-cell subsets (naïve, terminal effector, central memory, and effector memory) and rarer (<2%) natural killer (NK) and plasmacytoid dendritic cell (pDC) populations are represented. Importantly, the immune-cell profile of one reactive ALN appears to reflect the immune status of the patient’s axilla. Furthermore, FNA captures immune differences between patients with ≤1 or ≥2 metastatic ALNs. Increased numbers of naïve CD4+ T cells, but fewer terminal effector, central memory, and effector memory subpopulations, were obtained from patients with ≥2 metastatic ALNs. Moreover, despite their sparse distribution pattern on whole-section immunohistochemistry (WSI), FNA revealed that CD56+ NK cell activation receptors were decreased in patients with ≥2 metastatic ALNs. Finally, FNA captured a decrease in pDCs in patients with ≤1 metastatic ALNs, despite their clustered distribution pattern on WSI. Conclusions: FNA is not only feasible for sampling leukocytes from reactive, patient-derived ALNs, but also identifies immune-cell profiles that reflect axillary tumour burden in BC. Thus, this technique could be used to risk-stratify BC patients in the future. Full article
Show Figures

Figure 1

24 pages, 742 KB  
Review
NR4A Receptors in Immunity: Bridging Neuroendocrine and Inflammatory Pathways
by Simone Lemes Ferreira and Natalia Santucci
Receptors 2026, 5(1), 3; https://doi.org/10.3390/receptors5010003 - 25 Dec 2025
Viewed by 382
Abstract
Nuclear receptors (NRs) are ligand-activated transcription factors that mediate diverse cellular processes, including signalling, survival, proliferation, immune response and metabolism, through both genomic and non-genomic mechanisms in response to hormones and metabolic ligands. Given their central role in inter-organ, tissue, and cellular communication, [...] Read more.
Nuclear receptors (NRs) are ligand-activated transcription factors that mediate diverse cellular processes, including signalling, survival, proliferation, immune response and metabolism, through both genomic and non-genomic mechanisms in response to hormones and metabolic ligands. Given their central role in inter-organ, tissue, and cellular communication, NRs are critical for maintaining homeostasis and have become a major focus in biomedical research and drug discovery due to their association with numerous diseases. Among NRs, the NR4A subfamily (NR4A1/Nur77, NR4A2/Nurr1, and NR4A3/Nor1) responds to various stimuli—such as insulin, growth factors, inflammatory cytokines, and β-adrenergic signals—though their endogenous ligands remain unidentified. Their expression is tissue-dependent, particularly in energy-demanding tissues, where they modulate leukocyte function and promote an anti-inflammatory profile. Like other NRs, NR4As regulate acute and chronic inflammation by suppressing pro-inflammatory transcription factors (e.g., NF-κB) or enhancing their inhibitors, thereby polarising macrophages toward an anti-inflammatory phenotype. This review summarises current knowledge on the role of NR4A receptors in immune responses. Given their well-documented involvement in autoimmune diseases, inflammatory conditions, and cancer, elucidating their contributions to neuro–immune–endocrine crosstalk may uncover their therapeutic potential for immunopathological disorders. Full article
Show Figures

Figure 1

13 pages, 440 KB  
Article
Blood Immunophenotyping in Prediction of Gestational Hypertensive Conditions
by Almagul Kurmanova, Altynay Nurmakova, Damilya Salimbayeva, Gulfiruz Urazbayeva, Gaukhar Kurmanova, Natalya Kravtsova, Zhanar Kypshakbayeva and Madina Khalmirzaeva
Biomedicines 2025, 13(12), 3122; https://doi.org/10.3390/biomedicines13123122 - 18 Dec 2025
Viewed by 357
Abstract
Background: Hypertensive conditions during pregnancy, such as preeclampsia (PE), are multisystem obstetric complications, accompanied by changes in the immunological status. Although several types of immune cells are involved in pathogenesis of preeclampsia, such as regulatory T cells, macrophages, natural killer cells, and [...] Read more.
Background: Hypertensive conditions during pregnancy, such as preeclampsia (PE), are multisystem obstetric complications, accompanied by changes in the immunological status. Although several types of immune cells are involved in pathogenesis of preeclampsia, such as regulatory T cells, macrophages, natural killer cells, and neutrophils, most studies have focused on the concentration of circulating cytokines. Much less is known about intracellular cytokine production at the level of individual groups of peripheral blood immune cells. This gap limits our understanding of the early immunological changes that precede the clinical manifestation of the disease. Thus, the study of intracellular cytokine production in various leukocyte populations may provide new biomarkers for predicting preeclampsia. Objectives: To test the hypothesis that women with preeclampsia exhibit distinct intracellular cytokine profiles in specific peripheral blood immune cell subsets compared with normotensive pregnant women, and to assess whether these differences could serve as potential biomarkers for disease prediction. Methods: The study included a total of 78 pregnant women admitted to labor with physiological pregnancy (n = 32) and with gestational hypertension (GH) (n = 39) and PE (n = 7). The multicolor immunophenotyping with intracellular cytokine production of TNF, GM-CSF, IGF and receptor VEGFR-2 by different immunocompetent cell types was evaluated on a BD FACS CALIBUR flow cytometer. Results: Flow cytometry revealed a marked increase in the proportion of CD8+ GM-CSF+, CD56+VEGFR2+, CD14+IL-10+, and CD19+IGF+ cells in both hypertensive groups versus controls (p < 0.001). In contrast, CD56+TNF+ levels were significantly reduced (p < 0.001). For differentiating PE from GH, CD56+VEGFR2+ and CD19+IGF+ should be prioritized (AUC~0.66–0.78) with good specificity and moderate sensitivity. Conclusions: These data will not only expand existing knowledge about the role of intracellular cytokines in the pathogenesis of preeclampsia, but will also help to obtain new markers for predicting preeclampsia. Full article
(This article belongs to the Special Issue The Role of Cytokines in Health and Disease: 3rd Edition)
Show Figures

Graphical abstract

16 pages, 6166 KB  
Article
Potential Use of VYN202, a Novel Small Molecular Bromodomain and Extra-Terminal Inhibitor, in Mitigating Secondhand Smoke (SHS)-Induced Pulmonary Inflammation
by Katelyn A. Sturgis, Benjamin D. Davidson, Andrew W. Richardson, Olivia Hiatt, Blake C. Edwards, Ethan P. Evans, Carrleigh Campbell, Jack H. Radford, Juan A. Arroyo, Benjamin T. Bikman and Paul R. Reynolds
Curr. Issues Mol. Biol. 2025, 47(12), 1062; https://doi.org/10.3390/cimb47121062 - 18 Dec 2025
Viewed by 228
Abstract
Inflammation underpins pulmonary disease progression during tobacco smoke exposure, which may culminate in irreversible pulmonary disease. While primary smoke poses a notable risk, nearly half of the US population is also susceptible due to frequent exposure to secondhand smoke (SHS). In the present [...] Read more.
Inflammation underpins pulmonary disease progression during tobacco smoke exposure, which may culminate in irreversible pulmonary disease. While primary smoke poses a notable risk, nearly half of the US population is also susceptible due to frequent exposure to secondhand smoke (SHS). In the present study, we assessed the potential role of VYN202, a novel small molecular bromodomain and extra-terminal inhibitor, as a possible means of attenuating SHS-mediated inflammation. We exposed wild-type mice to an acute time course of room air (RA), SHS via a nose-only delivery system (Scireq Scientific, Montreal, Canada), or to both SHS and 10 mg/kg VYN202 (efficacious dose from prior inflammatory models) via oral gavage three times a week. Specific smoke exposure delivery to mice involved SHS from two cigarettes over 10 min, equilibration in room air for 10 min, followed by exposure to SHS from one cigarette for an additional 10 min, for a total SHS exposure of 20 min per day, five days a week for 30 days. We evaluated leukocyte abundance and the secretion of inflammatory mediators in bronchoalveolar lavage fluid (BALF). We also assessed general morphology via histology staining and the activation of receptor tyrosine kinase (RTK) family members. While standard hematoxylin and eosin (H&E) staining resulted in unchanged morphology, SHS-mediated increases in BALF protein abundance, total cellularity, and percent PMNs were attenuated with concomitant administration of VYN202. We also discovered SHS-induced activation of RTKs that were pro-inflammatory (JAK1, JAK3, ABL1, and ACK1), as well as RTKs related to endothelial and vascular remodeling (VEGFR3, VEGFR2, EphB4, EphB6, and FAK). Furthermore, inflammatory cytokines including GCSF, IFN-γ, IL-12p70, IL-17A, LIX, and TNF-α were all augmented by SHS exposure. Despite SHS exposure, each of these RTKs and cytokines/chemokines was significantly attenuated by VYN202. In summary, inflammatory responses induced by SHS exposure were mitigated by VYN202. These data reveal fascinating potential for the utility of VYN202 in lessening smoke-induced pulmonary exacerbations. Full article
(This article belongs to the Section Molecular Medicine)
Show Figures

Figure 1

16 pages, 16608 KB  
Article
HPV-Driven Immune Evasion in Cervical Cancer: Transcriptomic Identification of Downregulated Hub Genes and Suppressed Leukocyte Migration Pathways
by Sana Ismael Ameen, Mahla Masoudi, Hossein Azizi and Thomas Skutella
Int. J. Mol. Sci. 2025, 26(24), 12121; https://doi.org/10.3390/ijms262412121 - 17 Dec 2025
Viewed by 314
Abstract
Cervical cancer progression, particularly in the context of HPV infection, is driven by complex transcriptional alterations within the tumor microenvironment. Understanding the molecular mechanisms underlying HPV-induced immune evasion is crucial for developing effective therapeutic strategies. Transcriptomic analyses were performed using three independent datasets [...] Read more.
Cervical cancer progression, particularly in the context of HPV infection, is driven by complex transcriptional alterations within the tumor microenvironment. Understanding the molecular mechanisms underlying HPV-induced immune evasion is crucial for developing effective therapeutic strategies. Transcriptomic analyses were performed using three independent datasets (GSE127265, GSE166466, and GSE218460) to identify differentially expressed genes (DEGs) between HPV-positive and HPV-negative cervical cancer samples. Protein–protein interaction networks were constructed using Cytoscape and STRING, and immune infiltration was assessed via the TIMER database. A total of 572 DEGs were commonly identified between tumor and normal tissues, with HPV-positive samples showing distinct transcriptional profiles. Several downregulated hub genes were associated with immune regulation and receptor tyrosine kinase signaling. Immune infiltration analysis revealed altered dendritic cell and T cell patterns, indicating HPV-mediated immune modulation. Pathway enrichment identified the leukocyte transendothelial migration pathway as a key mechanism impaired by HPV infection. These findings highlight the critical role of immune-related hub genes in HPV-driven cervical cancer progression and suggest potential therapeutic targets to counteract HPV-induced immune suppression. Full article
Show Figures

Figure 1

24 pages, 1966 KB  
Review
The Expanding Role of HLA-E in Host Defense: A Target for Broadly Applicable Vaccines and Immunotherapies
by Mahsa Rafieiyan, Marco Pio La Manna, Francesco Dieli, Nadia Caccamo and Giusto Davide Badami
Cells 2025, 14(24), 1983; https://doi.org/10.3390/cells14241983 - 14 Dec 2025
Viewed by 507
Abstract
Human leukocyte antigen (HLA)-E, a non-classical class I molecule with limited polymorphism, bridges innate and adaptive immunity. Traditionally, the role of HLA-E had been associated with regulating natural killer (NK) cell activity via CD94/NKG2 receptors, by presenting self-peptides derived from the leader sequence [...] Read more.
Human leukocyte antigen (HLA)-E, a non-classical class I molecule with limited polymorphism, bridges innate and adaptive immunity. Traditionally, the role of HLA-E had been associated with regulating natural killer (NK) cell activity via CD94/NKG2 receptors, by presenting self-peptides derived from the leader sequence of HLA-I. Recent findings reveal its ability to present pathogen-derived peptides to CD8+ T cells, eliciting unconventional cytotoxic responses. This review examines the expanding role of HLA-E-restricted T cells in viral and bacterial infections and their capacity to recognize diverse microbial peptides and enhance immune response when classical HLA pathways are impaired. We also highlight key advances in immunotherapy and vaccine development, including CMV-vectored platforms, donor-unrestricted TCR-based strategies, and peptide prediction algorithms. The minimal polymorphism of HLA-E, its resistance to viral immune evasion, and its ability to present conserved pathogen peptides position it as a promising target for universal vaccines and next-generation immunotherapies. Understanding these unconventional roles may pave the way for broadly applicable immunotherapies and vaccines against infectious diseases. Full article
(This article belongs to the Section Cellular Immunology)
Show Figures

Figure 1

11 pages, 1318 KB  
Article
Antibody-Dependent Cytotoxicity of Monocytes in Preeclampsia Is Associated with Soluble Forms of HLA
by Aleksey M. Krasnyi, Leya E. Sorokina, Anastacia Maria Argentova-Stevens, Diana N. Kokoeva, Aleksey A. Alekseev, Tatjana Jankevic, Natalia E. Kan, Victor L. Tyutyunnik and Gennady T. Sukhikh
Int. J. Mol. Sci. 2025, 26(23), 11638; https://doi.org/10.3390/ijms262311638 - 1 Dec 2025
Viewed by 380
Abstract
Preeclampsia (PE) is a serious gestational complication that affects the lives of the mother and the child. Women with PE showed higher levels of pro-inflammatory cytokines secreted by leukocytes compared with women with normal pregnancies. The differences are most noticeable in the percentage [...] Read more.
Preeclampsia (PE) is a serious gestational complication that affects the lives of the mother and the child. Women with PE showed higher levels of pro-inflammatory cytokines secreted by leukocytes compared with women with normal pregnancies. The differences are most noticeable in the percentage of CD16+ monocytes, although the mechanism underlying this increase remains unclear. The CD16 receptor is critical for antibody-dependent cellular cytotoxicity, and by binding to antibodies on the surface of target cells, it activates their death. In this study, we examined the effect of soluble placental factors on the expression of CD16 monocytes and the potential role the soluble form of human leukocyte antigen (HLA) has on CD16 monocyte expression. At the first stage of our study, we collected samples of placental villi fragments from 58 pregnant women (38 women with PE and 20 with a healthy pregnancy). Then we studied the effect of placental villus-conditioned culture medium on CD16 expression by monocytes derived from the same women. It was shown that the content of CD16+ monocytes increased significantly in women with PE within 3 h and to a lesser extent in women with a healthy pregnancy (p = 0.009). Also, the addition of the recombinant histocompatibility HLA-B to the placental villus-conditioned culture medium blocks the induction of CD16 expression on monocytes. At the second stage of our study, we typed HLA class I and class II alleles in the umbilical cord blood samples and the venous blood samples taken from 38 women with PE and 40 women with a normal pregnancy. It was found that certain HLA class II alleles predominate in women with preeclampsia. The DRB1*01:01:01G allele showed the greatest difference (p < 0.001). Analyzing five alleles simultaneously makes it possible to predict the PE with AUC = 0.76. Evaluation of unique children’s alleles also showed that class II alleles have greater differences among them than class I alleles. The DQB1*06:03:01G allele had the greatest differences with p = 0.03 (the number was higher in the control group). Performing an analysis of four alleles of children simultaneously allowed us to predict PE with an AUC of 0.64. This work suggests that the activation of CD16+ monocyte expression occurs due to the interaction of soluble placental antigens with monocytes. The most likely way to activate CD16 expression on monocytes is by HLA class II (both maternal and fetal) interaction with CD4 receptors on the surface of monocytes, whereas HLA class I is capable of blocking this process. Evaluation of maternal HLA alleles may be a significant marker for predicting PE. Full article
(This article belongs to the Special Issue Pathogenesis of Preeclampsia: From a Molecular Perspective)
Show Figures

Figure 1

22 pages, 701 KB  
Review
The Potential of β-Synuclein-Specific Regulatory T Cell Therapy as a Treatment for Progressive Multiple Sclerosis
by Grace E. Osmond, Nevin A. John, Yi Tian Ting and Joshua D. Ooi
Int. J. Mol. Sci. 2025, 26(23), 11534; https://doi.org/10.3390/ijms262311534 - 28 Nov 2025
Viewed by 576
Abstract
Disease progression in multiple sclerosis (MS) is now known to affect many patients, even those not diagnosed with progressive subtypes. Progressive and neurodegenerative aspects of MS are poorly treated by currently available therapies. Research on new therapeutic options is needed to improve health [...] Read more.
Disease progression in multiple sclerosis (MS) is now known to affect many patients, even those not diagnosed with progressive subtypes. Progressive and neurodegenerative aspects of MS are poorly treated by currently available therapies. Research on new therapeutic options is needed to improve health outcomes in people with MS. This review highlights the potential for treatment using an engineered T cell receptor–regulatory T cell (TCR-Treg) therapy targeting the presynaptic protein beta-synuclein. Tregs respond to self-antigens presented on human leukocyte antigen (HLA) class II with anti-inflammatory and pro-neural healing effects, but this response is impaired in MS patients. Since the HLA-DRB1*15:01 allele is known to contribute to MS pathogenesis, a TCR specific to a known antigen presented on DRB1*15:01 can be transduced into Tregs to direct them to activate within the inflamed brain tissue. Beta-synuclein is released from neurons at a high level after neural damage, may be presented on HLA, enables homing of specific T cells to the grey matter, and is immunogenic in progressive MS patients. This review presents beta-synuclein as a disease-relevant antigen to target for therapeutic development. Full article
(This article belongs to the Special Issue Insights in Multiple Sclerosis (MS) and Neuroimmunology: 2nd Edition)
Show Figures

Figure 1

14 pages, 652 KB  
Review
The Inflammatory Role of Serum Amyloid A in the Pathogenesis and Progression of Diabetic Nephropathy
by Antigoni Stavrou, Christina A. Kousparou and Argyrios Tsakalis
J. Clin. Med. 2025, 14(23), 8427; https://doi.org/10.3390/jcm14238427 - 27 Nov 2025
Viewed by 1091
Abstract
Diabetic nephropathy (DN) remains the leading cause of end-stage renal disease (ESRD) worldwide, primarily affecting individuals with Type 2 Diabetes Mellitus (T2DM). While traditional risk factors—such as hypertension, poor glycemic control, and dyslipidemia—are well known, recent research has illuminated the pivotal role of [...] Read more.
Diabetic nephropathy (DN) remains the leading cause of end-stage renal disease (ESRD) worldwide, primarily affecting individuals with Type 2 Diabetes Mellitus (T2DM). While traditional risk factors—such as hypertension, poor glycemic control, and dyslipidemia—are well known, recent research has illuminated the pivotal role of inflammation in DN pathogenesis. Inflammatory processes involving chemokines, cytokines, immune cell infiltration, and pro-fibrotic signaling pathways (e.g., NFκB, JAK/STAT) contribute significantly to glomerular and tubulointerstitial damage. Key immune players include macrophages and T lymphocytes, particularly CD4+ T cells, which correlate with disease severity and progression. Serum Amyloid A (SAA), an acute-phase reactant traditionally associated with Serum Amyloid A Amyloidosis (AA amyloidosis), has emerged as both a biomarker and active mediator of renal inflammation in DN. SAA promotes cytokine release, leukocyte recruitment, and extracellular matrix remodeling, contributing to glomerular and tubular injury. Elevated Saa3 expression in experimental models correlates with DN progression, while activation of the advanced glycation end products and the receptors for advanced glycation end products (AGE–RAGE) axis in podocytes enhances SAA upregulation and inflammatory signaling. Increasing evidence now indicates that SAA functions, not only as a marker of systemic inflammation, but also as a mechanistically significant driver of intrarenal injury, bridging metabolic dysregulation with sustained inflammatory and fibrotic signaling. Emerging therapeutic approaches—including interleukin 6 (IL-6) blockade, inhibition of AGE formation, targeted anti-fibrotic agents, and recently developed SAA-directed RNA or peptide therapeutics—underscore the therapeutic potential of modulating SAA activity in DN. Preclinical evidence further supports the efficacy of monoclonal antibodies, signaling inhibitors, and dietary anti-inflammatory compounds in mitigating renal injury. Collectively, these developments position SAA as a central mediator at the intersection of metabolic, inflammatory, and fibrotic pathways, highlighting its promise as both a diagnostic biomarker and a therapeutic target for early intervention in diabetic kidney disease. Full article
(This article belongs to the Section Nephrology & Urology)
Show Figures

Figure 1

12 pages, 1559 KB  
Article
Modulation of Master Transcription Factor Expression of Nile Tilapia Leukocytes via Cholinergic Pathways
by Manuel Ivan Girón-Pérez, Kenia María Ramírez-Ibarra, Carlos Eduardo Covantes-Rosales, Daniel Alberto Girón-Pérez, Francisco Fabián Razura-Carmona, Arturo Contis-Montes de Oca, Jorge Morales-Montor, Lenin Pavón and Gladys Alejandra Toledo-Ibarra
Int. J. Mol. Sci. 2025, 26(22), 11206; https://doi.org/10.3390/ijms262211206 - 20 Nov 2025
Viewed by 329
Abstract
Teleost fish are the first evolutionary group to exhibit an innate and adaptive immune system. Within the mechanisms of adaptive immunity, fish possess, among others, T-helper cells (CD4-like) and their differentiation machinery, regulated by the master transcription factors T-bet, GATA3, Foxp3, and RORγ. [...] Read more.
Teleost fish are the first evolutionary group to exhibit an innate and adaptive immune system. Within the mechanisms of adaptive immunity, fish possess, among others, T-helper cells (CD4-like) and their differentiation machinery, regulated by the master transcription factors T-bet, GATA3, Foxp3, and RORγ. Many studies support the existence of a non-neuronal cholinergic system involved in the immune response, named after the ability of leukocytes to synthesize de novo acetylcholine (ACh). Organophosphorus pesticides (OPs), such as diazoxon (DXN), are examples of compounds that act as cholinergic disruptors with immunotoxic effects. The present study aimed to evaluate the expression of transcription factors in leukocytes (spleen mononuclear cells, SMNCs) of Nile tilapia by modulating cholinergic pathways in immune cells using agonists, antagonists, and diazoxon (DXN), an anticholinesterase substance. The obtained data showed a significant increase in RORγ mRNA expression upon stimulation with the nicotinic agonist, whereas activation of the muscarinic receptor with its agonist increased T-bet mRNA expression. An alteration in RORγ expression levels induced by DXN exposure was also observed. The results suggest a probable directing of the immune response towards a pro-inflammatory profile orchestrated mainly by RORγ and T-bet transcription factors in response to cholinergic stimuli. Full article
(This article belongs to the Special Issue Molecular Mechanisms of Toxicity Caused by Environmental Pollutants)
Show Figures

Figure 1

15 pages, 1592 KB  
Article
Genetic Aberrations in Normal Tissues Adjacent to Biliary Tract Cancers
by Chae Hwa Kwon, Sung Hee Park, Hye Ji Lee, Jong Hyun Lee, Sung Yong Han, Yong Mok Park, Hyung Il Seo, Dong Uk Kim and Byeong Gwan Noh
Biomedicines 2025, 13(11), 2812; https://doi.org/10.3390/biomedicines13112812 - 18 Nov 2025
Viewed by 569
Abstract
Background: The role of normal tissues adjacent to tumors (NATs) in biliary tract cancer (BTC) remains unclear, despite their potential contributions to field cancerization. Methods: Targeted genomic profiling of tumor tissues, patient-matched NATs, and peripheral blood leukocytes from 13 patients with [...] Read more.
Background: The role of normal tissues adjacent to tumors (NATs) in biliary tract cancer (BTC) remains unclear, despite their potential contributions to field cancerization. Methods: Targeted genomic profiling of tumor tissues, patient-matched NATs, and peripheral blood leukocytes from 13 patients with BTCs was performed. Clinicopathological data, including inflammatory conditions and precursor lesions (biliary intraepithelial neoplasia [BilIN] and intraductal papillary neoplasm of the bile duct), were integrated with genomic findings. Results: Tumor tissues exhibited recurrent alterations in genes regulating DNA damage response, cell cycle control, and oncogenic signaling. Importantly, rather than being genetically silent, NATs harbor early somatic variants distinct from those in both tumor and germline DNA. These alterations were not directly associated with cancer-related pathways, but rather with extracellular matrix-receptor interactions, suggesting that NATs may represent an intermediate step in carcinogenesis. All patients with extrahepatic cholangiocarcinoma presented with BilIN in adjacent tissues, providing histological evidence of field cancerization linked to chronic inflammation. Conclusions: This systematic comparison of tumors, NATs, and germline DNAs in BTCs revealed that NATs contain biologically relevant somatic mutations. The concordance between the inflammatory background, precursor lesions, and genomic alterations supports a multistep carcinogenic model and highlights opportunities for early BTC detection and risk stratification. Full article
Show Figures

Graphical abstract

27 pages, 8542 KB  
Article
The Receptor for Advanced Glycation End-Products (RAGE) Regulates Cell Adhesion Through Upregulation of ITGA8
by Swetha Thiyagarajan, Estelle Leclerc and Stefan W. Vetter
Cells 2025, 14(22), 1805; https://doi.org/10.3390/cells14221805 - 17 Nov 2025
Viewed by 772
Abstract
The Receptor for Advanced Glycation End-Products (RAGE) is a cell surface receptor of the immunoglobulin-like receptor superfamily. RAGE is a pattern-recognition, multi-ligand receptor that binds glycated proteins, specific non-glycated proteins, and nucleic acids. RAGE ligands are typically part of the group of damage-associated [...] Read more.
The Receptor for Advanced Glycation End-Products (RAGE) is a cell surface receptor of the immunoglobulin-like receptor superfamily. RAGE is a pattern-recognition, multi-ligand receptor that binds glycated proteins, specific non-glycated proteins, and nucleic acids. RAGE ligands are typically part of the group of damage-associated molecular patterns (DAMPs) or alarmins. As such, RAGE is a receptor for molecular products of cellular stress, abnormal metabolism, and inflammation. Activation of RAGE by its ligands leads to pro-inflammatory signaling, often resulting in persistent RAGE activation in various disease states. Consequently, RAGE has been investigated as a potential drug target in the treatment of diabetic complications, vascular disease, Alzheimer’s disease, and multiple types of cancer. An underexplored aspect of RAGE is its role in cell adhesion. Structural comparison of the extracellular domain of RAGE has revealed structural similarity to the activated leukocyte cell adhesion molecule (ALCAM). The present study reveals the role and mechanism of RAGE in regulating cell adhesion. We investigated the role of individual RAGE domains in cell adhesion to extracellular matrix proteins and the changes in protein expression resulting from RAGE upregulation. Key findings include that RAGE displays substrate-specific adhesion to extracellular matrix proteins, that the intracellular domain of RAGE is required for modulating cell spreading, and that regulation of ITGA8 depends on the cytoplasmic domain of RAGE. Full article
(This article belongs to the Section Cell Motility and Adhesion)
Show Figures

Figure 1

17 pages, 4706 KB  
Article
P2Y12-Inhibitor Clopidogrel Promotes Collateral Artery Growth in a Murine Hindlimb Model of Arteriogenesis
by Katharina Elbs, Lisa Bobrowski, Christoph Arnholdt, Matthias Kübler, Philipp Götz, Michael R. Rohrmoser, Daphne Merkus, Manuel Lasch and Elisabeth Deindl
Biomedicines 2025, 13(11), 2790; https://doi.org/10.3390/biomedicines13112790 - 16 Nov 2025
Viewed by 2438
Abstract
Background/Objectives: Clopidogrel is a P2Y12 receptor inhibitor commonly used as antiplatelet therapy for patients with cardiovascular occlusive diseases. However, its role in vascular remodeling remains poorly understood. Platelets orchestrate the sterile inflammation in arteriogenesis, an endogenous process to bypass an occluded [...] Read more.
Background/Objectives: Clopidogrel is a P2Y12 receptor inhibitor commonly used as antiplatelet therapy for patients with cardiovascular occlusive diseases. However, its role in vascular remodeling remains poorly understood. Platelets orchestrate the sterile inflammation in arteriogenesis, an endogenous process to bypass an occluded artery. Therefore, we investigated the impact of P2Y12-inhibition by Clopidogrel on arteriogenesis. Methods: In this study, we utilized a well-established murine hindlimb model of arteriogenesis. To quantify the growth of collateral arteries, we employed laser-Doppler perfusion measurements and immunohistological analysis of growing compared to resting collateral arteries. Additional immunofluorescence and histological stains were conducted to assess immune cell recruitment and activation. Whole-blood flow cytometry was performed to analyze platelet–leukocyte interactions, and complete blood counts were obtained to quantify leukocyte and platelet numbers. Results: The findings of this study demonstrate that Clopidogrel promotes perfusion recovery following the induction of arteriogenesis compared to controls, attributed to elevated levels of proliferating vascular cells. Furthermore, compared to controls, Clopidogrel treatment significantly enhanced platelet-leukocyte interactions, increasing perivascular mast cell recruitment and degranulation, finally resulting in regenerative macrophage accumulation required for collateral artery growth. Conclusions: Clopidogrel treatment boosts arteriogenesis by enhancing the local regenerative inflammation relevant for vascular cell proliferation. Therefore, P2Y12 inhibition may represent a therapeutic option to effectively promote natural bypass growth in patients with cardiovascular occlusive diseases. Full article
(This article belongs to the Section Cell Biology and Pathology)
Show Figures

Graphical abstract

20 pages, 3608 KB  
Article
Toll-like Receptor 7 Deficiency Attenuates Platelet Dysfunction in Sepsis
by Rashida Mohamed-Hinds, Arijit Dutta, Chanhee Park, Xiaomei Yang, Lin Zou, Wei Chao and Brittney Williams
Biomolecules 2025, 15(11), 1604; https://doi.org/10.3390/biom15111604 - 15 Nov 2025
Viewed by 644
Abstract
Sepsis is a clinical syndrome caused by abnormal host response to infection. Thrombocytopenia and platelet dysfunction are common findings in sepsis and associated with worse outcomes. The innate immune single-stranded RNA sensor, Toll-like Receptor-7 (TLR7), plays a key role in thrombocytopenia in sepsis. [...] Read more.
Sepsis is a clinical syndrome caused by abnormal host response to infection. Thrombocytopenia and platelet dysfunction are common findings in sepsis and associated with worse outcomes. The innate immune single-stranded RNA sensor, Toll-like Receptor-7 (TLR7), plays a key role in thrombocytopenia in sepsis. This study investigated whether TLR7 signaling also contributes to platelet dysfunction in sepsis, and whether the bioactivity of downstream inflammatory mediators, specifically extracellular vesicles (EVs), is impacted by the TLR7 signaling pathway. Sepsis was induced in wild-type (WT) and TLR7-deficient (TLR7−/−) mice by cecal ligation and puncture. Blood was collected at twenty-four hours for platelet and plasma isolation, and platelet function was assessed using aggregation, adhesion, and calcium flux assays. EVs were isolated from plasma and used in vitro to evaluate their impact on platelet–leukocyte aggregate (PLA) formation. We found that septic platelets are highly activated and more adhesive, yet show markedly impaired aggregation and reduced calcium signaling, indicating functional exhaustion despite activation. Notably, mice lacking TLR7 maintained stronger platelet aggregation, enhanced adhesion, and preserved calcium release in the septic state compared to wild-type controls, suggesting a protective effect of TLR7 deficiency. Plasma EVs increased in abundance and size during sepsis and promoted clot and PLA formation in vitro. Notably, EV-mediated platelet activation was reduced with EVs derived from TLR7-deficient mice. Our results demonstrate that while sepsis drives persistent platelet activation and dysfunction, TLR7 deficiency preserves platelet function and modulates the pathogenic activity of EV-mediated platelet activation, highlighting TLR7 as a key regulator and potential therapeutic target in sepsis-induced platelet dysfunction. Full article
Show Figures

Figure 1

Back to TopTop