Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.508
Filter
1.
Front Immunol ; 15: 1412800, 2024.
Article in English | MEDLINE | ID: mdl-39170608

ABSTRACT

Like TNFα, IL-6 is upregulated in Crohn's disease (CD) especially in patients associated with Mycobacterium avium paratuberculosis (MAP) infection, and both cytokines have been targeted as a therapeutic option for the treatment of the disease despite the accepted partial response in some patients. Limited response to anti-IL-6 receptor-neutralizing antibodies therapy may be related to the homeostatic dual role of IL-6. In this study, we investigated the effects and the signaling mechanism of IL-6 involved in intestinal epithelial integrity and function during MAP infection using an in vitro model that consists of THP-1, HT-29 and Caco-2 cell lines. Clinically, we determined that plasma samples from MAP-infected CD patients have higher IL-6 levels compared to controls (P-value < 0.001). In CD-like macrophages, MAP infection has significantly upregulated the secretion of IL-6 and the shedding of (IL-6R) from THP-1 macrophages, P-value < 0.05. Intestinal cell lines (Caco-2 and HT-29) were treated with the supernatant of MAP-infected THP-1 macrophages with or without a neutralizing anti-IL-6R antibody. Treating intestinal Caco-2 cells with supernatant of MAP-infected macrophages resulted in significant upregulation of intestinal damage markers including claudin-2 and SERPINE1/PAI-1. Interestingly, blocking IL-6 signaling exacerbated that damage and further increased the levels of the damage markers. In HT-29 cells, MAP infection upregulated MUC2 expression, a protective response that was reversed when IL-6R was neutralized. More importantly, blocking IL-6 signaling during MAP infection rescued damaged Caco-2 cells from MAP-induced apoptosis. The data clearly supports a protective role of IL-6 in intestinal epithelia integrity and function especially in CD patients associated with MAP infection. The findings may explain the ineffective response to anti-IL6 based therapy and strongly support a therapeutic option that restores the physiologic level of IL-6 in patient's plasma. A new treatment strategy based on attenuation of IL-6 expression and secretion in inflammatory diseases should be considered.


Subject(s)
Interleukin-6 , Intestinal Mucosa , Mycobacterium avium subsp. paratuberculosis , Paratuberculosis , Receptors, Interleukin-6 , Humans , Receptors, Interleukin-6/metabolism , Receptors, Interleukin-6/antagonists & inhibitors , Receptors, Interleukin-6/immunology , Mycobacterium avium subsp. paratuberculosis/immunology , Caco-2 Cells , Interleukin-6/metabolism , Interleukin-6/immunology , HT29 Cells , Intestinal Mucosa/microbiology , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Paratuberculosis/immunology , Paratuberculosis/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Crohn Disease/immunology , Crohn Disease/microbiology , THP-1 Cells , Male , Antibodies, Neutralizing/pharmacology , Female , Adult , Epithelial Cells/metabolism , Epithelial Cells/immunology , Epithelial Cells/microbiology , Middle Aged , Signal Transduction
2.
Front Immunol ; 15: 1400722, 2024.
Article in English | MEDLINE | ID: mdl-39170612

ABSTRACT

Background: Although neoadjuvant therapy has brought numerous benefits to patients, not all patients can benefit from it. Chemokines play a crucial role in the tumor microenvironment and are closely associated with the prognosis and treatment of colorectal cancer. Therefore, constructing a prognostic model based on chemokines will help risk stratification and providing a reference for the personalized treatment. Methods: Employing LASSO-Cox predictive modeling, a chemokine-based prognostic model was formulated, harnessing the data from TCGA and GEO databases. Then, our exploration focused on the correlation between the chemokine signature and elements such as the immune landscape, somatic mutations, copy number variations, and drug sensitivity. CXCL10+M1 macrophages identified via scRNA-seq. Monocle2 showed cell pseudotime trajectories, CellChat characterized intercellular communication. CytoTRACE analyzed neoadjuvant therapy stemness, SCENIC detected cell type-specific regulation. Lastly, validation was performed through multiplex immunofluorescence experiments. Results: A model based on 15 chemokines was constructed and validated. High-risk scores correlated with poorer prognosis and advanced TNM and clinical stages. Individuals presenting elevated risk scores demonstrated an increased propensity towards the development of chemotherapy resistance. Subsequent scRNA-seq data analysis indicated that patients with higher presence of CXCL10+ M1 macrophages in tumor tissues are more likely to benefit from neoadjuvant therapy. Conclusion: We developed a chemokine-based prognostic model by integrating both single-cell and bulk RNA-seq data. Furthermore, we revealed epithelial cell heterogeneity in neoadjuvant outcomes and identified CXCL10+ M1 macrophages as potential therapy response predictors. These findings could significantly contribute to risk stratification and serve as a key guide for the advancement of personalized therapeutic approaches.


Subject(s)
Chemokine CXCL10 , Colorectal Neoplasms , Neoadjuvant Therapy , Tumor Microenvironment , Humans , Colorectal Neoplasms/therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Neoadjuvant Therapy/methods , Prognosis , Tumor Microenvironment/immunology , Biomarkers, Tumor , Macrophages/immunology , Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Female , Male , Treatment Outcome , Chemokines/metabolism , Chemokines/genetics
3.
Proc Natl Acad Sci U S A ; 121(35): e2405746121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39172787

ABSTRACT

While macrophage heterogeneity during metabolic dysfunction-associated steatohepatitis (MASH) has been described, the fate of these macrophages during MASH regression is poorly understood. Comparing macrophage heterogeneity during MASH progression vs regression, we identified specific macrophage subpopulations that are critical for MASH/fibrosis resolution. We elucidated the restorative pathways and gene signatures that define regression-associated macrophages and establish the importance of TREM2+ macrophages during MASH regression. Liver-resident Kupffer cells are lost during MASH and are replaced by four distinct monocyte-derived macrophage subpopulations. Trem2 is expressed in two macrophage subpopulations: i) monocyte-derived macrophages occupying the Kupffer cell niche (MoKC) and ii) lipid-associated macrophages (LAM). In regression livers, no new transcriptionally distinct macrophage subpopulation emerged. However, the relative macrophage composition changed during regression compared to MASH. While MoKC was the major macrophage subpopulation during MASH, they decreased during regression. LAM was the dominant macrophage subtype during MASH regression and maintained Trem2 expression. Both MoKC and LAM were enriched in disease-resolving pathways. Absence of TREM2 restricted the emergence of LAMs and formation of hepatic crown-like structures. TREM2+ macrophages are functionally important not only for restricting MASH-fibrosis progression but also for effective regression of inflammation and fibrosis. TREM2+ macrophages are superior collagen degraders. Lack of TREM2+ macrophages also prevented elimination of hepatic steatosis and inactivation of HSC during regression, indicating their significance in metabolic coordination with other cell types in the liver. TREM2 imparts this protective effect through multifactorial mechanisms, including improved phagocytosis, lipid handling, and collagen degradation.


Subject(s)
Kupffer Cells , Liver Cirrhosis , Macrophages , Membrane Glycoproteins , Receptors, Immunologic , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Animals , Mice , Macrophages/metabolism , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Liver Cirrhosis/genetics , Kupffer Cells/metabolism , Liver/metabolism , Liver/pathology , Lipid Metabolism , Mice, Inbred C57BL , Male , Lipids , Fatty Liver/metabolism , Fatty Liver/pathology , Fatty Liver/genetics , Mice, Knockout
4.
Proc Natl Acad Sci U S A ; 121(35): e2400194121, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39172792

ABSTRACT

Size-dependent phagocytosis is a well-characterized phenomenon in monocytes and macrophages. However, this size effect for preferential gene delivery to these important cell targets has not been fully exploited because commonly adopted stabilization methods for electrostatically complexed nucleic acid nanoparticles, such as PEGylation and charge repulsion, typically arrest the vehicle size below 200 nm. Here, we bridge the technical gap in scalable synthesis of larger submicron gene delivery vehicles by electrostatic self-assembly of charged nanoparticles, facilitated by a polymer structurally designed to modulate internanoparticle Coulombic and van der Waals forces. Specifically, our strategy permits controlled assembly of small poly(ß-amino ester)/messenger ribonucleic acid (mRNA) nanoparticles into particles with a size that is kinetically tunable between 200 and 1,000 nm with high colloidal stability in physiological media. We found that assembled particles with an average size of 400 nm safely and most efficiently transfect monocytes following intravenous administration and mediate their differentiation into macrophages in the periphery. When a CpG adjuvant is co-loaded into the particles with an antigen mRNA, the monocytes differentiate into inflammatory dendritic cells and prime adaptive anticancer immunity in the tumor-draining lymph node. This platform technology offers a unique ligand-independent, particle-size-mediated strategy for preferential mRNA delivery and enables therapeutic paradigms via monocyte programming.


Subject(s)
Monocytes , Nanoparticles , RNA, Messenger , Monocytes/metabolism , Nanoparticles/chemistry , RNA, Messenger/genetics , RNA, Messenger/metabolism , Animals , Mice , Humans , Polyelectrolytes/chemistry , Macrophages/metabolism , Polyamines/chemistry , Particle Size , Cell Differentiation , Gene Transfer Techniques , Dendritic Cells/metabolism , Static Electricity , Polymers
5.
Clin Exp Med ; 24(1): 187, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39136841

ABSTRACT

Glioblastoma (GBM) is a highly heterogeneous disease with poor clinical outcomes. To comprehensively dissect the molecular landscape of GBM and heterogeneous macrophage clusters in the progression of GBM, this study integrates single-cell and bulk transcriptome data to recognize a distinct pro-tumor macrophage cluster significantly associated with the prognosis of GBM and develop a GBM prognostic signature to facilitate prior subtypes. Leveraging glioma single-cell sequencing data, we identified a novel pro-tumor macrophage subgroup, marked by S100A9, which might interact with endothelial cells to facilitate tumor progression via angiogenesis. To further benefit clinical application, a prognostic signature was established with the genes associated with pro-tumor macrophages. Patients classified within the high-risk group characterized with enrichment in functions related to tumor progression, including epithelial-mesenchymal transition and hypoxia, displays elevated mutations in the TERT promoter region, reduced methylation in the MGMT promoter region, poorer prognoses, and diminished responses to temozolomide therapy, thus effectively discriminating between the prognostic outcomes of GBM patients. Our research sheds light on the intricate microenvironment of gliomas and identifies potential molecular targets for the development of novel therapeutic approaches.


Subject(s)
Gene Expression Profiling , Glioblastoma , Single-Cell Analysis , Humans , Glioblastoma/genetics , Glioblastoma/pathology , Prognosis , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Tumor Microenvironment/genetics , Temozolomide/therapeutic use , Macrophages/metabolism , Transcriptome , Telomerase/genetics , Tumor Suppressor Proteins/genetics , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , DNA Modification Methylases/genetics , DNA Repair Enzymes
6.
BMC Pulm Med ; 24(1): 392, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39138424

ABSTRACT

BACKGROUND: The immunologic features of nontuberculous mycobacterial pulmonary disease (NTM-PD) are largely unclear. This study investigated the immunologic features of NTM-PD using digital spatial profiling techniques. METHODS: Lung tissues obtained from six patients with NTM-PD between January 1, 2006, and December 31, 2020, at Seoul National University Hospital were subjected to RNA sequencing. Cores from the peribronchial areas were stained with CD3, CD68, and DNASyto13, and gene expression at the whole-transcriptome level was quantified using PCR amplification and Illumina sequencing. Lung tissues from six patients with bronchiectasis collected during the same period were used as controls. The RNA sequencing results were validated using immunohistochemistry (IHC) in another cohort (30 patients with NTM-PD and 15 patients with bronchiectasis). RESULTS: NTM-PD exhibited distinct gene expression patterns in T cells and macrophages. Gene set enrichment analysis revealed that pathways related to antigen presentation and processing were upregulated in NTM-PD, particularly in macrophages. Macrophages were more prevalent and the expression of genes associated with the M1 phenotype (CD40 and CD80) was significantly elevated. Although macrophages were activated in the NTM-PD group T cell activity was unaltered. Notably, expression of the costimulatory molecule CD28 was decreased in NTM-PD. IHC analysis showed that T cells expressing Foxp3 or TIM-3, which facilitate the regulatory functions of T cells, were increased. CONCLUSIONS: NTM-PD exhibits distinct immunologic signatures characterized by the activation of macrophages without T cell activation.


Subject(s)
Mycobacterium Infections, Nontuberculous , Humans , Male , Mycobacterium Infections, Nontuberculous/immunology , Mycobacterium Infections, Nontuberculous/genetics , Female , Middle Aged , Aged , Transcriptome , Macrophages/immunology , Macrophages/metabolism , Lung/microbiology , Lung/immunology , Lung/pathology , Nontuberculous Mycobacteria/genetics , Nontuberculous Mycobacteria/immunology , Lung Diseases/genetics , Lung Diseases/microbiology , Lung Diseases/immunology , T-Lymphocytes/immunology , Gene Expression Profiling , Adult , Bronchiectasis/immunology , Bronchiectasis/genetics , Bronchiectasis/microbiology
7.
Methods Mol Biol ; 2828: 1-9, 2024.
Article in English | MEDLINE | ID: mdl-39147965

ABSTRACT

Immune responses rely on efficient and coordinated migration of immune cells to the site of infection or injury. To reach the site of immunological threat often requires long-range navigation of immune cells through complex tissue and vascular networks. Chemotaxis, cell migration steered by gradients of cell-attractive chemicals that bind sensory receptors, is central to this response. Chemoattractant receptors mostly belong to the G-protein-coupled receptor (GPCR) family, but the way attractant-receptor signaling directs cell migration is not fully understood. Direct-viewing chemotaxis chambers combined with time-lapse microscopy give a powerful tool to study the dynamic details of cells' responses to different attractant landscapes. Here, we describe the application of one such chamber (the Dunn chamber) to study bone marrow-derived macrophage chemotaxis to gradients of complement C5a.


Subject(s)
Chemotaxis , Macrophages , Chemotaxis/drug effects , Macrophages/metabolism , Macrophages/cytology , Macrophages/drug effects , Animals , Mice , Complement C5a/metabolism , Complement C5a/pharmacology , Time-Lapse Imaging/methods , Cell Movement , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Bone Marrow Cells/drug effects , Receptors, G-Protein-Coupled/metabolism
8.
Sci Rep ; 14(1): 19538, 2024 08 22.
Article in English | MEDLINE | ID: mdl-39174693

ABSTRACT

Macrophages played an important role in the progression and treatment of head and neck squamous cell carcinoma (HNSCC). We employed weighted gene co-expression network analysis (WGCNA) to identify macrophage-related genes (MRGs) and classify patients with HNSCC into two distinct subtypes. A macrophage-related risk signature (MRS) model, comprising nine genes: IGF2BP2, PPP1R14C, SLC7A5, KRT9, RAC2, NTN4, CTLA4, APOC1, and CYP27A1, was formulated by integrating 101 machine learning algorithm combinations. We observed lower overall survival (OS) in the high-risk group and the high-risk group showed elevated expression levels in most of the immune checkpoint and human leukocyte antigen (HLA) genes, suggesting a strong immune evasion capacity. Correspondingly, TIDE score positively correlated with risk score, implying that high-risk tumors may resist immunotherapy more effectively. At the single-cell level, we noted macrophages in the tumor microenvironment (TME) predominantly stalled in the G2/M phase, potentially hindering epithelial-mesenchymal transition and playing a crucial role in the inhibition of tumor progression. Finally, the proliferation and migration abilities of HNSCC cells significantly decreased after the expression of IGF2BP2 and SLC7A5 reduced. It also decreased migration ability of macrophages and facilitated their polarization towards the M1 direction. Our study constructed a novel MRS for HNSCC, which could serve as an indicator for predicting the prognosis, immune infiltration and immunotherapy for HNSCC patients.


Subject(s)
Head and Neck Neoplasms , Immunotherapy , Machine Learning , Macrophages , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment , Humans , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/therapy , Squamous Cell Carcinoma of Head and Neck/pathology , Immunotherapy/methods , Prognosis , Macrophages/immunology , Macrophages/metabolism , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/therapy , Head and Neck Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/genetics , Male , Cell Line, Tumor
9.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 49(5): 655-666, 2024 May 28.
Article in English, Chinese | MEDLINE | ID: mdl-39174879

ABSTRACT

OBJECTIVES: Progressive bone resorption and destruction is one of the most critical clinical features of middle ear cholesteatoma, potentially leading to various intracranial and extracranial complications. However, the mechanisms underlying bone destruction in middle ear cholesteatoma remain unclear. This study aims to explore the role of parathyroid hormone-related protein (PTHrP) in bone destruction associated with middle ear cholesteatoma. METHODS: A total of 25 cholesteatoma specimens and 13 normal external auditory canal skin specimens were collected from patients with acquired middle ear cholesteatoma. Immunohistochemical staining was used to detect the expressions of PTHrP, receptor activator for nuclear factor-kappa B ligand (RANKL), and osteoprotegerin (OPG) in cholesteatoma and normal tissues. Tartrate-resistant acid phosphatase (TRAP) staining was used to detect the presence of TRAP positive multi-nucleated macrophages in cholesteatoma and normal tissues. Mono-nuclear macrophage RAW264.7 cells were subjected to interventions, divided into a RANKL intervention group and a PTHrP+ RANKL co-intervention group. TRAP staining was used to detect osteoclast formation in the 2 groups. The mRNA expression levels of osteoclast-related genes, including TRAP, cathepsin K (CTSK), and nuclear factor of activated T cell cytoplasmic 1 (NFATc1), were measured using real-time polymerase chain reaction (real-time PCR) after the interventions. Bone resorption function of osteoclasts was assessed using a bone resorption pit analysis. RESULTS: Immunohistochemical staining showed significantly increased expression of PTHrP and RANKL and decreased expression of OPG in cholesteatoma tissues (all P<0.05). PTHrP expression was significantly positively correlated with RANKL, the RANKL/OPG ratio, and negatively correlated with OPG expression (r=0.385, r=0.417, r=-0.316, all P<0.05). Additionally, the expression levels of PTHrP and RANKL were significantly positively correlated with the degree of bone destruction in cholesteatoma (r=0.413, r=0.505, both P<0.05). TRAP staining revealed a large number of TRAP-positive cells, including multi-nucleated osteoclasts with three or more nuclei, in the stroma surrounding the cholesteatoma epithelium. After 5 days of RANKL or PTHrP+RANKL co-intervention, the number of osteoclasts was significantly greater in the PTHrP+RANKL co-intervention group than that in the RANKL group (P<0.05), with increased mRNA expression levels of TRAP, CTSK, and NFATc1 (all P<0.05). Scanning electron microscopy of bone resorption pits showed that the number (P<0.05) and size of bone resorption pits on bone slices were significantly greater in the PTHrP+RANKL co-intervention group compared with the RANKL group. CONCLUSIONS: PTHrP may promote the differentiation of macrophages in the surrounding stroma of cholesteatoma into osteoclasts through RANKL induction, contributing to bone destruction in middle ear cholesteatoma.


Subject(s)
Bone Resorption , Cell Differentiation , Cholesteatoma, Middle Ear , Macrophages , Osteoclasts , Osteoprotegerin , Parathyroid Hormone-Related Protein , RANK Ligand , Osteoclasts/metabolism , RANK Ligand/metabolism , RANK Ligand/genetics , Parathyroid Hormone-Related Protein/metabolism , Humans , Animals , Mice , Osteoprotegerin/metabolism , Cholesteatoma, Middle Ear/metabolism , Cholesteatoma, Middle Ear/pathology , Bone Resorption/metabolism , Macrophages/metabolism , NFATC Transcription Factors/metabolism , NFATC Transcription Factors/genetics , RAW 264.7 Cells , Male
10.
Zh Nevrol Psikhiatr Im S S Korsakova ; 124(7. Vyp. 2): 79-84, 2024.
Article in Russian | MEDLINE | ID: mdl-39175244

ABSTRACT

OBJECTIVE: To investigate the role of dopamine receptor D1DR and D2DR in the production of cytokines interleukin-6 (IL-6) and IL-1ß by monocytes and macrophages in patients with relapsing-remitting multiple sclerosis (MS). MATERIAL AND METHODS: Ten patients with relapsing-remitting MS and 10 healthy subjects were examined. The level of IL-6 and IL-1ß production was assessed in culture supernatants obtained from CD14+ monocytes or macrophages stimulated with interferon-γ (IFN-γ) and lipopolysaccharide (LPS). To study the role of dopamine receptors in the regulation of CD14+ monocytes or macrophages, samples of cells were incubated in the presence of specific D1DR or D2DR antagonists, after which IFN-γ/LPS were added to the cultures. Levels of cytokines in culture supernatants were measured by enzyme-linked immunosorbent assay. RESULTS: The production of IL-6 and IL-1ß by CD14+ monocytes and macrophages was comparable between the groups. Blockade of D1DR suppressed cytokine production by CD14+ monocytes and macrophages in both groups. In contrast, blockade of D2DR increased the production of cytokines by CD14+ monocytes and did not affect cytokine production by macrophages in both groups. CONCLUSIONS: Targeting of dopaminergic receptors could be considered as an additional mechanism of immunomodulation in MS with both pro- and anti-inflammatory effects on cells of the innate immune system.


Subject(s)
Interleukin-1beta , Interleukin-6 , Macrophages , Multiple Sclerosis, Relapsing-Remitting , Receptors, Dopamine D1 , Humans , Adult , Female , Interleukin-6/metabolism , Interleukin-1beta/metabolism , Male , Multiple Sclerosis, Relapsing-Remitting/metabolism , Multiple Sclerosis, Relapsing-Remitting/immunology , Receptors, Dopamine D1/metabolism , Macrophages/metabolism , Macrophages/immunology , Monocytes/metabolism , Monocytes/immunology , Receptors, Dopamine D2/metabolism , Lipopolysaccharides/pharmacology , Cells, Cultured , Lipopolysaccharide Receptors/metabolism , Middle Aged , Interferon-gamma/metabolism
11.
Oncoimmunology ; 13(1): 2393442, 2024.
Article in English | MEDLINE | ID: mdl-39175947

ABSTRACT

The inflammatory tumor microenvironment (TME) is a key driver for tumor-promoting processes. Tumor-associated macrophages are one of the main immune cell types in the TME and their increased density is related to poor prognosis in prostate cancer. Here, we investigated the influence of pro-inflammatory (M1) and immunosuppressive (M2) macrophages on prostate cancer lineage plasticity. Our findings reveal that M1 macrophage secreted factors upregulate genes related to stemness while downregulating genes associated with androgen response in prostate cancer cells. The expression of cancer stem cell (CSC) plasticity markers NANOG, KLF4, SOX2, OCT4, and CD44 was stimulated by the secreted factors from M1 macrophages. Moreover, AR and its target gene PSA were observed to be suppressed in LNCaP cells treated with secreted factors from M1 macrophages. Inhibition of NFκB signaling using the IKK16 inhibitor resulted in downregulation of NANOG, SOX2, and CD44 and CSC plasticity. Our study highlights that the secreted factors from M1 macrophages drive prostate cancer cell plasticity by upregulating the expression of CSC plasticity markers through NFκB signaling pathway.


Subject(s)
Hyaluronan Receptors , Kruppel-Like Factor 4 , Macrophages , NF-kappa B , Nanog Homeobox Protein , Neoplastic Stem Cells , Prostatic Neoplasms , SOXB1 Transcription Factors , Signal Transduction , Male , Humans , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/genetics , Nanog Homeobox Protein/metabolism , Nanog Homeobox Protein/genetics , SOXB1 Transcription Factors/metabolism , SOXB1 Transcription Factors/genetics , Hyaluronan Receptors/metabolism , Hyaluronan Receptors/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Kruppel-Like Factor 4/metabolism , NF-kappa B/metabolism , Cell Line, Tumor , Macrophages/metabolism , Up-Regulation , Tumor Microenvironment/immunology , Cell Plasticity/genetics , Gene Expression Regulation, Neoplastic , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Animals , Mice
12.
Front Immunol ; 15: 1447190, 2024.
Article in English | MEDLINE | ID: mdl-39176096

ABSTRACT

F. nucleatum, involved in carcinogenesis of colon carcinomas, has been described as part of the commensal flora of the female upper reproductive tract. Although its contribution to destructive inflammatory processes is well described, its role as commensal uterine bacteria has not been thoroughly investigated. Since carcinogenesis shares similar mechanisms with early pregnancy development (including proliferation, invasion, blood supply and the induction of tolerance), these mechanisms induced by F. nucleatum could play a role in early pregnancy. Additionally, implantation and placentation require a well-balanced immune activation, which might be suitably managed by the presence of a limited amount of bacteria or bacterial residues. We assessed the effect of inactivated F. nucleatum on macrophage-trophoblast interactions. Monocytic cells (THP-1) were polarized into M1, M2a or M2c macrophages by IFN-γ, IL-4 or TGF-ß, respectively, and subsequently treated with inactivated fusobacteria (bacteria:macrophage ratio of 0.1 and 1). Direct effects on macrophages were assessed by viability assay, flow cytometry (antigen presentation molecules and cytokines), qPCR (cytokine expression), in-cell Western (HIF and P-NF-κB) and ELISA (VEGF secretion). The function of first trimester extravillous trophoblast cells (HTR-8/SVneo) in response to macrophage-conditioned medium was microscopically assessed by migration (scratch assay), invasion (sprouting assay) and tube formation. Underlying molecular changes were investigated by ELISA (VEGF secretion) and qPCR (matrix-degrading factors and regulators). Inflammation-primed macrophages (M1) as well as high bacterial amounts increased pro-inflammatory NF-κB expression and inflammatory responses. Subsequently, trophoblast functions were impaired. In contrast, low bacterial stimulation caused an increased HIF activation and subsequent VEGF-A secretion in M2c macrophages. Accordingly, there was an increase of trophoblast tube formation. Our results suggest that a low-mass endometrial/decidual microbiome can be tolerated and while it supports implantation and further pregnancy processes.


Subject(s)
Fusobacterium nucleatum , Macrophages , Trophoblasts , Humans , Trophoblasts/immunology , Trophoblasts/microbiology , Trophoblasts/metabolism , Fusobacterium nucleatum/immunology , Fusobacterium nucleatum/physiology , Macrophages/immunology , Macrophages/microbiology , Macrophages/metabolism , Female , Pregnancy , Cytokines/metabolism , THP-1 Cells , NF-kappa B/metabolism , Fusobacterium Infections/immunology , Fusobacterium Infections/microbiology , Vascular Endothelial Growth Factor A/metabolism
13.
Semin Immunopathol ; 46(3-4): 11, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-39134914

ABSTRACT

Throughout the individual's reproductive period of life the ovary undergoes continues changes, including cyclic processes of cell death, tissue regeneration, proliferation, and vascularization. Tissue-resident leucocytes particularly macrophages, play a crucial role in shaping ovarian function and maintaining homeostasis. Macrophages crucially promote angiogenesis in the follicles and corpora lutea, thereby supporting steroidogenesis. Recent research on macrophage origins and early tissue seeding has unveiled significant insights into their role in early organogenesis, e.g. in the testis. Here, we review evidence about the prenatal ovarian seeding of leucocytes, primarily macrophages with angiogenic profiles, and its connection to gametogenesis. In the prenatal ovary, germ cells proliferate, form cysts, and undergo changes that, following waves of apoptosis, give rice to the oocytes contained in primordial follicles. These follicles constitute the ovarian reserve that lasts throughout the female's reproductive life. Simultaneously, yolk-sac-derived primitive macrophages colonizing the early ovary are gradually replaced or outnumbered by monocyte-derived fetal macrophages. However, the cues indicating how macrophage colonization and follicle assembly are related are elusive. Macrophages may contribute to organogenesis by promoting early vasculogenesis. Whether macrophages contribute to ovarian lymphangiogenesis or innervation is still unknown. Ovarian organogenesis and gametogenesis are vulnerable to prenatal insults, potentially programming dysfunction in later life, as observed in polycystic ovary syndrome. Experimental and, more sparsely, epidemiological evidence suggest that adverse stimuli during pregnancy can program defective folliculogenesis or a diminished follicle reserve in the offspring. While the ovary is highly sensitive to inflammation, the involvement of local immune responses in programming ovarian health and disease remains to be thoroughly investigated.


Subject(s)
Macrophages , Ovary , Humans , Female , Ovary/immunology , Ovary/metabolism , Animals , Macrophages/immunology , Macrophages/metabolism , Ovarian Diseases/metabolism , Ovarian Diseases/pathology , Pregnancy , Ovarian Follicle/metabolism
14.
Nat Commun ; 15(1): 7097, 2024 Aug 17.
Article in English | MEDLINE | ID: mdl-39154007

ABSTRACT

Converging evidence indicates that extra-embryonic yolk sac is the source of both macrophages and endothelial cells in adult mouse tissues. Prevailing views are that these embryonically derived cells are maintained after birth by proliferative self-renewal in their differentiated states. Here we identify clonogenic endothelial-macrophage (EndoMac) progenitor cells in the adventitia of embryonic and postnatal mouse aorta, that are independent of Flt3-mediated bone marrow hematopoiesis and derive from an early embryonic CX3CR1+ and CSF1R+ source. These bipotent progenitors are proliferative and vasculogenic, contributing to adventitial neovascularization and formation of perfused blood vessels after transfer into ischemic tissue. We establish a regulatory role for angiotensin II, which enhances their clonogenic and differentiation properties and rapidly stimulates their proliferative expansion in vivo. Our findings demonstrate that embryonically derived EndoMac progenitors participate in local vasculogenic responses in the aortic wall by contributing to the expansion of endothelial cells and macrophages postnatally.


Subject(s)
Aorta , Macrophages , Animals , Macrophages/cytology , Macrophages/metabolism , Aorta/cytology , Mice , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Endothelial Cells/cytology , Endothelial Cells/metabolism , Cell Differentiation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Angiotensin II , Cell Proliferation , Stem Cells/cytology , Stem Cells/metabolism , Mice, Inbred C57BL , Female , Neovascularization, Physiologic , Receptors, Chemokine/metabolism , Receptors, Chemokine/genetics , Male , Hematopoiesis/physiology , fms-Like Tyrosine Kinase 3
15.
Front Immunol ; 15: 1438935, 2024.
Article in English | MEDLINE | ID: mdl-39156890

ABSTRACT

Background: pancreatic ductal adenocarcinoma (PDAC) is a malignant tumor with a very poor prognosis and a complex tumor microenvironment, which plays a key role in tumor progression and treatment resistance. Glycosylation plays an important role in processes such as cell signaling, immune response and protein stability. Materials and methods: single-cell RNA sequencing data and spatial transcriptome data were obtained from GSE197177 and GSE224411, respectively, and RNA-seq data and survival information were obtained from UCSC Xena and TCGA. Multiple transcriptomic data were comprehensively analyzed to explore the role of glycosylation processes in tumor progression, and functional experiments were performed to assess the effects of MGAT1 overexpression on PDAC cell proliferation and migration. Results: In PDAC tumor samples, the glycosylation level of macrophages was significantly higher than that of normal samples. MGAT1 was identified as a key glycosylation-related gene, and its high expression was associated with better patient prognosis. Overexpression of MGAT1 significantly inhibited the proliferation and migration of PDAC cells and affected intercellular interactions in the tumor microenvironment. Conclusion: MGAT1 plays an important role in PDAC by regulating glycosylation levels in macrophages, influencing tumor progression and improving prognosis.MGAT1 is a potential therapeutic target for PDAC and further studies are needed to develop targeted therapeutic strategies against MGAT1 to improve clinical outcomes.


Subject(s)
Carcinoma, Pancreatic Ductal , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Pancreatic Neoplasms , Tumor Microenvironment , Humans , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/mortality , Glycosylation , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/mortality , Cell Proliferation/genetics , Tumor Microenvironment/genetics , Cell Line, Tumor , Cell Movement/genetics , Prognosis , Macrophages/metabolism , Macrophages/immunology , Biomarkers, Tumor/genetics
16.
Front Immunol ; 15: 1410082, 2024.
Article in English | MEDLINE | ID: mdl-39156889

ABSTRACT

The immune system requires a high energy expenditure to resist pathogen invasion. Macrophages undergo metabolic reprogramming to meet these energy requirements and immunologic activity and polarize to M1-type macrophages. Understanding the metabolic pathway switching in large yellow croaker (Larimichthys crocea) macrophages in response to lipopolysaccharide (LPS) stimulation and whether this switching affects immunity is helpful in explaining the stronger immunity of hypoxia-tolerant L. crocea. In this study, transcript levels of glycolytic pathway genes (Glut1 and Pdk1), mRNA levels or enzyme activities of glycolytic enzymes [hexokinase (HK), phosphofructokinase (PFK), pyruvate kinase (PK), and lactate dehydrogenase A (LDHA)], aerobic respiratory enzymes [pyruvate dehydrogenase (PDH), isocitrate dehydrogenase (IDH), and succinate dehydrogenase (SDH)], metabolites [lactic acid (LA) and adenosine triphosphate (ATP)], levels of bactericidal products [reactive oxygen species (ROS) and nitric oxide (NO)], and transcripts and level changes of inflammatory factors [IL1ß, TNFα, and interferon (IFN) γ] were detected in LPS-stimulated L. crocea head kidney macrophages. We showed that glycolysis was significantly induced, the tricarboxylic acid (TCA) cycle was inhibited, and metabolic reprogramming occurred, showing the Warburg effect when immune cells were activated. To determine the potential regulatory mechanism behind these changes, LcHIF-1α was detected and found to be significantly induced and transferred to the nucleus after LPS stimulation. LcHif-1α interference led to a significant reduction in glycolytic pathway gene transcript expression, enzyme activity, metabolites, bactericidal substances, and inflammatory factor levels; a significant increase in the aerobic respiration enzymes; and decreased migration, invasion, and phagocytosis. Further ultrastructural observation by electron microscopy showed that fewer microspheres contained phagocytes and that more cells were damaged after LcHif-1α interference. LcHif-1α overexpression L. crocea head kidney macrophages showed the opposite trend, and promoter activities of Ldha and Il1ß were significantly enhanced after LcHif-1α overexpression in HEK293T cells. Our data showed that LcHIF-1α acted as a metabolic switch in L. crocea macrophages and was important in polarization. Hypoxia-tolerant L. crocea head kidney showed a stronger Warburg effect and inhibited the TCA cycle, higher metabolites, and bactericidal substance levels. These results collectively revealed that LcHif-1α may promote the functional activities of head kidney macrophages in protecting hypoxia-tolerant L. crocea from Aeromonas hydrophila infection.


Subject(s)
Aeromonas hydrophila , Fish Diseases , Gram-Negative Bacterial Infections , Hypoxia-Inducible Factor 1, alpha Subunit , Macrophages , Perciformes , Animals , Perciformes/immunology , Perciformes/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/microbiology , Fish Diseases/immunology , Fish Diseases/microbiology , Fish Diseases/metabolism , Gram-Negative Bacterial Infections/immunology , Gram-Negative Bacterial Infections/veterinary , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Aeromonas hydrophila/physiology , Aeromonas hydrophila/immunology , Lipopolysaccharides/immunology , Glycolysis , Fish Proteins/genetics , Fish Proteins/metabolism , Macrophage Activation/immunology , Hypoxia/immunology , Hypoxia/metabolism , Head Kidney/immunology , Head Kidney/metabolism
17.
Front Immunol ; 15: 1427457, 2024.
Article in English | MEDLINE | ID: mdl-39156902

ABSTRACT

Aconitate decarboxylase-1 (ACOD1) is expressed by activated macrophages and generates itaconate that exerts anti-microbial and immunoregulatory effects. ACOD1-itaconate is essential for macrophage-mediated control of the intracellular pathogen Coxiella (C.) burnetii, which causes Q fever. Two isomers of itaconate, mesaconate and citraconate, have overlapping yet distinct activity on macrophage metabolism and inflammatory gene expression. Here, we found that all three isomers inhibited the growth of C. burnetii in axenic culture in ACCM-2 medium. However, only itaconate reduced C. burnetii replication efficiently in Acod1-/- macrophages. In contrast, addition of citraconate strongly increased C. burnetii replication in Acod1+/- macrophages, whereas mesaconate weakly enhanced bacterial burden in Acod1-/- macrophages. Analysis of intracellular isomers showed that exogenous citraconate and mesaconate inhibited the generation of itaconate by infected Acod1+/- macrophages. Uptake of added isomers into Acod1-/- macrophages was increased after infection for itaconate and mesaconate, but not for citraconate. Mesaconate, but not citraconate, competed with itaconate for uptake into macrophages. Taken together, inhibition of itaconate generation by macrophages and interference with the uptake of extracellular itaconate could be identified as potential mechanisms behind the divergent effects of citraconate and mesaconate on C. burnetii replication in macrophages or in axenic culture.


Subject(s)
Axenic Culture , Carboxy-Lyases , Coxiella burnetii , Macrophages , Succinates , Coxiella burnetii/drug effects , Coxiella burnetii/growth & development , Succinates/pharmacology , Animals , Macrophages/microbiology , Macrophages/immunology , Macrophages/metabolism , Macrophages/drug effects , Mice , Carboxy-Lyases/metabolism , Mice, Knockout , Q Fever/immunology , Q Fever/microbiology , Mice, Inbred C57BL , Hydro-Lyases
18.
Biomed Mater ; 19(5)2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39121890

ABSTRACT

This study delves into the potential of amorphous titanium oxide (aTiO2) nano-coating to enhance various critical aspects of non-Ti-based metallic orthopedic implants. These implants, such as medical-grade stainless steel (SS), are widely used for orthopedic devices that demand high strength and durability. The aTiO2nano-coating, deposited via magnetron sputtering, is a unique attempt to improve the osteogenesis, the inflammatory response, and to reduce bacterial colonization on SS substrates. The study characterized the nanocoated surfaces (SS-a TiO2) in topography, roughness, wettability, and chemical composition. Comparative samples included uncoated SS and sandblasted/acid-etched Ti substrates (Ti). The biological effects were assessed using human mesenchymal stem cells (MSCs) and primary murine macrophages. Bacterial tests were carried out with two aerobic pathogens (S. aureusandS. epidermidis) and an anaerobic bacterial consortium representing an oral dental biofilm. Results from this study provide strong evidence of the positive effects of the aTiO2nano-coating on SS surfaces. The coating enhanced MSC osteoblastic differentiation and exhibited a response similar to that observed on Ti surfaces. Macrophages cultured on aTiO2nano-coating and Ti surfaces showed comparable anti-inflammatory phenotypes. Most significantly, a reduction in bacterial colonization across tested species was observed compared to uncoated SS substrates, further supporting the potential of aTiO2nano-coating in biomedical applications. The findings underscore the potential of magnetron-sputtering deposition of aTiO2nano-coating on non-Ti metallic surfaces such as medical-grade SS as a viable strategy to enhance osteoinductive factors and decrease pathogenic bacterial adhesion. This could significantly improve the performance of metallic-based biomedical devices beyond titanium.


Subject(s)
Coated Materials, Biocompatible , Macrophages , Materials Testing , Mesenchymal Stem Cells , Osteogenesis , Stainless Steel , Surface Properties , Titanium , Titanium/chemistry , Stainless Steel/chemistry , Animals , Humans , Mesenchymal Stem Cells/cytology , Mice , Coated Materials, Biocompatible/chemistry , Coated Materials, Biocompatible/pharmacology , Macrophages/metabolism , Osteogenesis/drug effects , Cell Differentiation , Prostheses and Implants , Osteoblasts/cytology , Staphylococcus aureus/drug effects , Biofilms , Staphylococcus epidermidis/drug effects , Bacterial Adhesion , Wettability
19.
J Exp Clin Cancer Res ; 43(1): 224, 2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39135069

ABSTRACT

BACKGROUND: High infiltration of tumor-associated macrophages (TAMs) is associated with tumor promotion and immunosuppression. The triggering receptor expressed on myeloid cells 2 (TREM2) is emerged as a key immunosuppressive regulator for TAMs, however, how TREM2-expressing TAMs are recruited and what ligands TREM2 interacts with to mediate immunosuppression is unknown. METHODS: Flow cytometry and single-cell RNA sequencing were used to analyze TREM2 expression. Mechanistically, mass spectrometry and immunoprecipitation were employed to identify proteins binding to TREM2. Phagocytosis and co-culture experiments were used to explore the in vitro functions of galectin3-TREM2 pair. Establishment of TREM2f/f-Lyz2-cre mice to validate the role of TREM2 signaling pathway in lung carcinogenesis. GB1107 were further supplemented to validate the therapeutic effect of Galectin3 based on TREM2 signaling regulation. RESULTS: This study identified that abundant TREM2+ macrophages were recruited at the intra-tumor site through the CCL2-CCR2 chemotactic axis. Galectin-3 impaired TREM2-mediated phagocytosis and promoted the conversion of TREM2+ macrophages to immunosuppressive TAMs with attenuated antigen presentation and co-stimulatory functions both in vitro both in vivo, and galectin-3 is a potential ligand for TREM2. Genetic and pharmacological blockade of TREM2 and galectin-3 significantly inhibited lung cancer progression in subcutaneous and orthotopic cancer models by remodeling the tumor immune microenvironment. CONCLUSION: Our findings revealed a previously unknown association between galectin-3 and TREM2 in TAMs of lung cancer, and suggested simultaneous inhibition of galectin3 and TREM2 as potent therapeutic approach for lung cancer therapy.


Subject(s)
Galectin 3 , Lung Neoplasms , Macrophages , Membrane Glycoproteins , Receptors, Immunologic , Animals , Lung Neoplasms/metabolism , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Mice , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Membrane Glycoproteins/metabolism , Humans , Galectin 3/metabolism , Galectin 3/genetics , Macrophages/metabolism , Macrophages/immunology , Disease Models, Animal
20.
Biofabrication ; 16(4)2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39142325

ABSTRACT

Hyalocytes, which are considered to originate from the monocyte/macrophage lineage, play active roles in vitreous collagen and hyaluronic acid synthesis. Obtaining a hyalocyte-compatible bioink during the 3D bioprinting of eye models is challenging. In this study, we investigated the suitability of a cartilage-decellularized extracellular matrix (dECM)-based bioink for printing a vitreous body model. Given that achieving a 3D structure and environment identical to those of the vitreous body necessitates good printability and biocompatibility, we examined the mechanical and biological properties of the developed dECM-based bioink. Furthermore, we proposed a 3D bioprinting strategy for volumetric vitreous body fabrication that supports cell viability, transparency, and self-sustainability. The construction of a 3D structure composed of bioink microfibers resulted in improved transparency and hyalocyte-like macrophage activity in volumetric vitreous mimetics, mimicking real vitreous bodies. The results indicate that our 3D structure could serve as a platform for drug testing in disease models and demonstrate that the proposed printing technology, utilizing a dECM-based bioink and volumetric vitreous body, has the potential to facilitate the development of advanced eye models for future studies on floater formation and visual disorders.


Subject(s)
Bioprinting , Extracellular Matrix , Ink , Printing, Three-Dimensional , Vitreous Body , Vitreous Body/metabolism , Vitreous Body/cytology , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Animals , Bioprinting/methods , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Humans , Cartilage/cytology , Cartilage/chemistry , Cartilage/metabolism , Cell Survival , Macrophages/metabolism , Macrophages/cytology
SELECTION OF CITATIONS
SEARCH DETAIL