Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.072
Filter
1.
Mol Biol Rep ; 51(1): 946, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39215899

ABSTRACT

BACKGROUND: Increased case reports have shown that patients with NF1 have an increased risk of extensive vascular vasculopathy. Previous studies demonstrated the presence of macrophages and smooth muscle cells in the neoplastic intima of carotid arteries after injury in Nf1+/- mice. However, whether NF1 gene mutations affect macrophage polarization and macrophage-smooth muscle cell interactions remains to be elucidated. METHODS: Scratch assay and transwell assay were utilized to detect cell migration ability. The dye 2',7'dichlorofluorescin diacetate and neutral red stain were used to assess intracellular ROS production and cell phagocytosis function, respectively. Proteins and mRNA expression were determined by western blot, RT-qPCR, and immunofluorescence. Finally, the macrophage (MAC) and vascular smooth muscle cell (VSMC) co-culture system was used to detect cellular crosstalk. RESULTS: Cell function assays confirmed that the Nf1-Q181X point mutation attenuated the phagocytosis of bone marrow-derived macrophages (BMDMs) and promoted the migration and ROS production of BMDMs. Moreover, we found that the Nf1-Q181X point mutation inhibited M1 but promoted M2 macrophage polarization by down-regulating p38, ERK, and JNK and up-regulating the Akt/STAT3 signaling pathway, respectively. Furthermore, in the MAC-VSMC co-culture system, we demonstrated that Nf1-Q181X point mutation-activated M2 BMDMs promoted proliferation and migration of VSMCs and induced the transformation of VSMCs from contractile phenotype to synthetic phenotype. CONCLUSION: The findings suggest that the Nf1-Q181X point mutation can mediate macrophage polarization and promote smooth muscle cell proliferation and migration, providing clinical clues for the treatment of NF1-complicated vasculopathy.


Subject(s)
Cell Movement , Cell Proliferation , Macrophages , Myocytes, Smooth Muscle , Neurofibromin 1 , Point Mutation , Proto-Oncogene Proteins c-akt , Signal Transduction , Cell Movement/genetics , Animals , Cell Proliferation/genetics , Macrophages/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Myocytes, Smooth Muscle/metabolism , Mice , Neurofibromin 1/genetics , Neurofibromin 1/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Phagocytosis/genetics , Reactive Oxygen Species/metabolism , Mice, Inbred C57BL , Coculture Techniques
2.
JCI Insight ; 9(17)2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39042472

ABSTRACT

Alveolar macrophages (AMs) act as gatekeepers of the lung's immune responses, serving essential roles in recognizing and eliminating pathogens. The transcription factor (TF) early growth response 2 (EGR2) has been recently described as required for mature AMs in mice; however, its mechanisms of action have not been explored. Here, we identified EGR2 as an epigenomic regulator and likely direct proximal transcriptional activator in AMs using epigenomic approaches (RNA sequencing, ATAC sequencing, and CUT&RUN). The predicted direct proximal targets of EGR2 included a subset of AM identity genes and ones related to pathogen recognition, phagosome maturation, and adhesion, such as Clec7a, Atp6v0d2, Itgb2, Rhoc, and Tmsb10. We provided evidence that EGR2 deficiency led to impaired zymosan internalization and reduced the capacity to respond to Aspergillus fumigatus. Mechanistically, the lack of EGR2 altered the transcriptional response, secreted cytokines (i.e., CXCL11), and inflammation-resolving lipid mediators (i.e., RvE1) of AMs during in vivo zymosan-induced inflammation, which manifested in impaired resolution. Our findings demonstrated that EGR2 is a key proximal transcriptional activator and epigenomic bookmark in AMs responsible for select, distinct components of cell identity and a protective transcriptional and epigenomic program against fungi.


Subject(s)
Aspergillus fumigatus , Early Growth Response Protein 2 , Macrophages, Alveolar , Phagocytosis , Animals , Macrophages, Alveolar/immunology , Macrophages, Alveolar/metabolism , Early Growth Response Protein 2/genetics , Early Growth Response Protein 2/metabolism , Early Growth Response Protein 2/immunology , Mice , Phagocytosis/immunology , Phagocytosis/genetics , Aspergillus fumigatus/immunology , Mice, Knockout , Mice, Inbred C57BL , Epigenomics , Epigenesis, Genetic/immunology , Zymosan
3.
PLoS Biol ; 22(7): e3002724, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39052688

ABSTRACT

Alternative transcription start site (TSS) usage regulation has been identified as a major means of gene expression regulation in metazoans. However, in fungi, its impact remains elusive as its study has thus far been restricted to model yeasts. Here, we first re-analyzed TSS-seq data to define genuine TSS clusters in 2 species of pathogenic Cryptococcus. We identified 2 types of TSS clusters associated with specific DNA sequence motifs. Our analysis also revealed that alternative TSS usage regulation in response to environmental cues is widespread in Cryptococcus, altering gene expression and protein targeting. Importantly, we performed a forward genetic screen to identify a unique transcription factor (TF) named Tur1, which regulates alternative TSS (altTSS) usage genome-wide when cells switch from exponential phase to stationary phase. ChiP-Seq and DamID-Seq analyses suggest that at some loci, the role of Tur1 might be direct. Tur1 has been previously shown to be essential for virulence in C. neoformans. We demonstrated here that a tur1Δ mutant strain is more sensitive to superoxide stress and phagocytosed more efficiently by macrophages than the wild-type (WT) strain.


Subject(s)
Fungal Proteins , Gene Expression Regulation, Fungal , Genome, Fungal , Transcription Factors , Transcription Initiation Site , Fungal Proteins/genetics , Fungal Proteins/metabolism , Transcription Factors/metabolism , Transcription Factors/genetics , Cryptococcus/genetics , Cryptococcus/pathogenicity , Cryptococcus/metabolism , Cryptococcus neoformans/genetics , Cryptococcus neoformans/pathogenicity , Cryptococcus neoformans/metabolism , Macrophages/microbiology , Macrophages/metabolism , Animals , Mice , Virulence/genetics , Phagocytosis/genetics
4.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39025525

ABSTRACT

Macrophages have important roles in mammary gland development and tissue homeostasis, but the specific mechanisms that regulate macrophage function need further elucidation. We have identified C/EBPß as an important transcription factor expressed by multiple macrophage populations in the normal mammary gland. Mammary glands from mice with C/EBPß-deficient macrophages (Cebpb ΔM) show a significant decrease in alveolar budding during the diestrus stage of the reproductive cycle, whereas branching morphogenesis remains unchanged. Defects in alveolar budding were found to be the result of both systemic hormones and local macrophage-directed signals. RNA sequencing shows significant changes in PR-responsive genes and alterations in the Wnt landscape of mammary epithelial cells of Cebpb ΔM mice, which regulate stem cell expansion during diestrus. Cebpb ΔM macrophages demonstrate a shift from a pro-inflammatory to a tissue-reparative phenotype, and exhibit increased phagocytic capacity as compared to WT. Finally, Cebpb ΔM macrophages down-regulate Notch2 and Notch3, which normally promote stem cell expansion during alveolar budding. These results suggest that C/EBPß is an important macrophage factor that facilitates macrophage-epithelial crosstalk during a key stage of mammary gland tissue homeostasis.


Subject(s)
CCAAT-Enhancer-Binding Protein-beta , Estrous Cycle , Macrophages , Mammary Glands, Animal , Animals , CCAAT-Enhancer-Binding Protein-beta/metabolism , CCAAT-Enhancer-Binding Protein-beta/genetics , Female , Mice , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Macrophages/metabolism , Estrous Cycle/genetics , Mice, Knockout , Receptors, Notch/metabolism , Receptors, Notch/genetics , Epithelial Cells/metabolism , Phagocytosis/genetics , Mice, Inbred C57BL , Gene Deletion
5.
Cell Mol Life Sci ; 81(1): 302, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-39008111

ABSTRACT

DNAX-activating protein of 12 kDa (DAP12) is a transmembrane adapter protein expressed in lymphoid and myeloid lineage cells. It interacts with several immunoreceptors forming functional complexes that trigger intracellular signaling pathways. One of the DAP12 associated receptors is the triggering receptor expressed on myeloid cells 2 (TREM2). Mutations in both DAP12 and TREM2 have been linked to neurodegenerative diseases. However, mechanisms involved in the regulation of subcellular trafficking and turnover of these proteins are not well understood. Here, we demonstrate that proteasomal degradation of DAP12 is increased in the absence of TREM2. Interestingly, unassembled DAP12 is also retained in early secretory compartments, including the endoplasmic reticulum (ER) and the ER-Golgi intermediate compartment (ERGIC), thereby preventing its transport to the plasma membrane. We also show that unassembled DAP12 interacts with the retention in ER sorting receptor 1 (RER1). The deletion of endogenous RER1 decreases expression of functional TREM2-DAP12 complexes and membrane proximal signaling, and resulted in almost complete inhibition of phagocytic activity in THP-1 differentiated macrophage-like cells. These results indicate that RER1 acts as an important regulator of DAP12 containing immunoreceptor complexes and immune cell function.


Subject(s)
Adaptor Proteins, Signal Transducing , Endoplasmic Reticulum , Membrane Glycoproteins , Receptors, Immunologic , Secretory Pathway , Humans , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/genetics , Endoplasmic Reticulum/metabolism , Secretory Pathway/genetics , Membrane Proteins/metabolism , Membrane Proteins/genetics , HEK293 Cells , Signal Transduction , Phagocytosis/genetics , Macrophages/metabolism , Protein Transport , Protein Binding , Animals , Golgi Apparatus/metabolism , Vesicular Transport Proteins/metabolism , Vesicular Transport Proteins/genetics , Cell Membrane/metabolism
6.
Life Sci Alliance ; 7(10)2024 Oct.
Article in English | MEDLINE | ID: mdl-39084876

ABSTRACT

Phagocytosis is an important immune response that protects the host from pathogen invasion. Rit1 GTPase is known to be involved in diverse cellular processes. However, its role in FcγR-mediated phagocytosis remains unclear. Our live-cell imaging analysis revealed that Rit1 was localized to the membranes of F-actin-rich phagocytic cups in RAW264 macrophages. Rit1 knockout and expression of the GDP-locked Rit1 mutant suppressed phagosome formation. We also found that TBC1D10B, a GAP for the Rab family GTPases, colocalizes with Rit1 in the membranes of phagocytic cups. Expression and knockout studies have shown that TBC1D10B decreases phagosome formation in both Rab-GAP activity-dependent and -independent manners. Notably, the expression of the GDP-locked Rit1 mutant or Rit1 knockout inhibited the dissociation of TBC1D10B from phagocytic cups. In addition, the expression of the GTP-locked Rit1 mutant promoted the dissociation of TBC1D10B in phagocytic cups and restored the rate of phagosome formation in TBC1D10B-expressing cells. These data suggest that Rit1-TBC1D10B signaling regulates FcγR-mediated phagosome formation in macrophages.


Subject(s)
GTPase-Activating Proteins , Macrophages , Phagocytosis , Phagosomes , Receptors, IgG , Signal Transduction , Animals , Mice , Actins/metabolism , Adaptor Proteins, Signal Transducing/metabolism , GTPase-Activating Proteins/metabolism , GTPase-Activating Proteins/genetics , Macrophages/metabolism , Phagocytosis/genetics , Phagosomes/metabolism , rab GTP-Binding Proteins/metabolism , rab GTP-Binding Proteins/genetics , RAW 264.7 Cells , Receptors, IgG/metabolism , ras Proteins/genetics , ras Proteins/metabolism
7.
Clin Immunol ; 265: 110301, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38944364

ABSTRACT

Septic cardiomyopathy (SCM) is characterized by an abnormal inflammatory response and increased mortality. The role of efferocytosis in SCM is not well understood. We used integrated multi-omics analysis to explore the clinical and genetic roles of efferocytosis in SCM. We identified six module genes (ATP11C, CD36, CEBPB, MAPK3, MAPKAPK2, PECAM1) strongly associated with SCM, leading to an accurate predictive model. Subgroups defined by EFFscore exhibited distinct clinical features and immune infiltration levels. Survival analysis showed that the C1 subtype with a lower EFFscore had better survival outcomes. scRNA-seq analysis of peripheral blood mononuclear cells (PBMCs) from sepsis patients identified four genes (CEBPB, CD36, PECAM1, MAPKAPK2) associated with high EFFscores, highlighting their role in SCM. Molecular docking confirmed interactions between diagnostic genes and tamibarotene. Experimental validation supported our computational results. In conclusion, our study identifies a novel efferocytosis-related SCM subtype and diagnostic biomarkers, offering new insights for clinical diagnosis and therapy.


Subject(s)
Biomarkers , Cardiomyopathies , Machine Learning , Phagocytosis , Sepsis , Humans , Cardiomyopathies/genetics , Cardiomyopathies/diagnosis , Prognosis , Male , Sepsis/genetics , Sepsis/diagnosis , Phagocytosis/genetics , Female , Middle Aged , Leukocytes, Mononuclear/metabolism , Aged , Molecular Docking Simulation , Efferocytosis , Multiomics
8.
Cell Death Dis ; 15(5): 313, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702326

ABSTRACT

CD24 is overexpressed in various tumours and considered a regulator of cell migration, invasion, and proliferation. Recent studies have found that CD24 on ovarian cancer (OC) and triple-negative breast cancer cells interacts with the inhibitory receptor sialic-acid-binding Ig-like lectin 10 (Siglec-10) on tumour-associated macrophages (TAMs) to inhibit phagocytosis by macrophages. Because of its multiple roles in regulating the immune response and tumorigenesis, CD24 is a very promising therapeutic target. However, the regulatory mechanism of CD24 in OC remains unclear. Here, we found that the long noncoding RNA (lncRNA) IL21-AS1, which was upregulated in OC, inhibited macrophage-mediated phagocytosis and promoted OC cell proliferation and apoptosis inhibition. More importantly, after IL21-AS1 knockdown, a significant survival advantage was observed in mice engrafted with tumours. Mechanistically, we identified IL21-AS1 as a hypoxia-induced lncRNA. Moreover, IL21-AS1 increased HIF1α-induced CD24 expression under hypoxic conditions. In parallel, we found that IL21-AS1 acted as a competing endogenous RNA (ceRNA) for miR-561-5p to regulate CD24 expression. Finally, IL21-AS1 increased CD24 expression in OC and facilitated OC progression. Our findings provide a molecular basis for the regulation of CD24, thus highlighting a potential strategy for targeted treatment of OC.


Subject(s)
CD24 Antigen , Carcinogenesis , Ovarian Neoplasms , Phagocytosis , RNA, Long Noncoding , CD24 Antigen/metabolism , CD24 Antigen/genetics , Female , Humans , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Phagocytosis/genetics , Animals , Mice , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Disease Progression , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , MicroRNAs/genetics , Mice, Nude , Apoptosis/genetics , Mice, Inbred BALB C , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics
9.
Microb Pathog ; 192: 106681, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38754565

ABSTRACT

Tuberculosis (TB) is a major fatal infectious disease globally, exhibiting high morbidity rates and impacting public health and other socio-economic factors. However, some individuals are resistant to TB infection and are referred to as "Resisters". Resisters remain uninfected even after exposure to high load of Mycobacterium tuberculosis (Mtb). To delineate this further, this study aimed to investigate the factors and mechanisms influencing the Mtb resistance phenotype. We assayed the phagocytic capacity of peripheral blood mononuclear cells (PBMCs) collected from Resisters, patients with latent TB infection (LTBI), and patients with active TB (ATB), following infection with fluorescent Mycobacterium bovis Bacillus Calmette-Guérin (BCG). Phagocytosis was stronger in PBMCs from ATB patients, and comparable in LTBI patients and Resisters. Subsequently, phagocytes were isolated and subjected to whole transcriptome sequencing and small RNA sequencing to analyze transcriptional expression profiles and identify potential targets associated with the resistance phenotype. The results revealed that a total of 277 mRNAs, 589 long non-coding RNAs, 523 circular RNAs, and 35 microRNAs were differentially expressed in Resisters and LTBI patients. Further, the endogenous competitive RNA (ceRNA) network was constructed from differentially expressed genes after screening. Bioinformatics, statistical analysis, and quantitative real-time polymerase chain reaction were used for the identification and validation of potential crucial targets in the ceRNA network. As a result, we obtained a ceRNA network that contributes to the resistance phenotype. TCONS_00034796-F3, ENST00000629441-DDX43, hsa-ATAD3A_0003-CYP17A1, and XR_932996.2-CERS1 may be crucial association pairs for resistance to TB infection. Overall, this study demonstrated that the phagocytic capacity of PBMCs was not a determinant of the resistance phenotype and that some non-coding RNAs could be involved in the natural resistance to TB infection through a ceRNA mechanism.


Subject(s)
Leukocytes, Mononuclear , MicroRNAs , Mycobacterium tuberculosis , Phagocytes , Phagocytosis , Tuberculosis , Humans , Phagocytes/metabolism , Phagocytes/immunology , Mycobacterium tuberculosis/genetics , Mycobacterium tuberculosis/immunology , Tuberculosis/genetics , Tuberculosis/microbiology , Tuberculosis/immunology , Phagocytosis/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Male , Adult , Gene Expression Profiling , Gene Regulatory Networks , Female , Transcriptome/genetics , Latent Tuberculosis/genetics , Latent Tuberculosis/immunology , Latent Tuberculosis/microbiology , Disease Resistance/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Mycobacterium bovis/immunology , Middle Aged , Computational Biology/methods , Young Adult , RNA, Competitive Endogenous
10.
Front Immunol ; 15: 1397541, 2024.
Article in English | MEDLINE | ID: mdl-38774870

ABSTRACT

Aim: Despite the significant therapeutic outcomes achieved in systemic treatments for liver hepatocellular carcinoma (LIHC), it is an objective reality that only a low proportion of patients exhibit an improved objective response rate (ORR) to current immunotherapies. Antibody-dependent cellular phagocytosis (ADCP) immunotherapy is considered the new engine for precision immunotherapy. Based on this, we aim to develop an ADCP-based LIHC risk stratification system and screen for relevant targets. Method: Utilizing a combination of single-cell RNA sequencing (scRNA-seq) and bulk RNA-seq data, we screened for ADCP modulating factors in LIHC and identified differentially expressed genes along with their involved functional pathways. A risk scoring model was established by identifying ADCP-related genes with prognostic value through LASSO Cox regression analysis. The risk scoring model was then subjected to evaluations of immune infiltration and immunotherapy relevance, with pan-cancer analysis and in vitro experimental studies conducted on key targets. Results: Building on the research by Kamber RA et al., we identified GYPA, CLDN18, and IRX5 as potential key target genes regulating ADCP in LIHC. These genes demonstrated significant correlations with immune infiltration cells, such as M1-type macrophages, and the effectiveness of immunotherapy in LIHC, as well as a close association with clinical pathological staging and patient prognosis. Pan-cancer analysis revealed that CLDN18 was prognostically and immunologically relevant across multiple types of cancer. Validation through tissue and cell samples confirmed that GYPA and CLDN18 were upregulated in liver cancer tissues and cells. Furthermore, in vitro knockdown of CLDN18 inhibited the malignancy capabilities of liver cancer cells. Conclusion: We have identified an ADCP signature in LIHC comprising three genes. Analysis based on a risk scoring model derived from these three genes, coupled with subsequent experimental validation, confirmed the pivotal role of M1-type macrophages in ADCP within LIHC, establishing CLDN18 as a critical ADCP regulatory target in LIHC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , RNA-Seq , Female , Humans , Male , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/therapy , Claudins/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Immunotherapy/methods , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Liver Neoplasms/therapy , Phagocytosis/genetics , Prognosis , Single-Cell Gene Expression Analysis , Tumor Microenvironment/immunology , Tumor Microenvironment/genetics
11.
Aging (Albany NY) ; 16(8): 6883-6897, 2024 04 10.
Article in English | MEDLINE | ID: mdl-38613800

ABSTRACT

BACKGROUND: Diabetic foot ulcers (DFUs) pose a serious long-term threat because of elevated mortality and disability risks. Research on its biomarkers is still, however, very limited. In this paper, we have effectively identified biomarkers linked with macrophage excretion in diabetic foot ulcers through the application of bioinformatics and machine learning methodologies. These findings were subsequently validated using external datasets and animal experiments. Such discoveries are anticipated to offer novel insights and approaches for the early diagnosis and treatment of DFU. METHODS: In this work, we used the Gene Expression Omnibus (GEO) database's datasets GSE68183 and GSE80178 as the training dataset to build a gene model using machine learning methods. After that, we used the training and validation sets to validate the model (GSE134431). On the model genes, we performed enrichment analysis using both gene set variant analysis (GSVA) and gene set enrichment analysis (GSEA). Additionally, the model genes were subjected to immunological association and immune function analyses. RESULTS: In this study, PROS1 was identified as a potential key target associated with macrophage efflux in DFU by machine learning and bioinformatics approaches. Subsequently, the key biomarker status of PROS1 in DFU was also confirmed by external datasets. In addition, PROS1 also plays a key role in macrophage exudation in DFU. This gene may be associated with macrophage M1, CD4 memory T cells, naïve B cells, and macrophage M2, and affects IL-17, Rap1, hedgehog, and JAK-STAT signaling pathways. CONCLUSIONS: PROS1 was identified and validated as a biomarker for DFU. This finding has the potential to provide a target for macrophage clearance of DFU.


Subject(s)
Diabetic Foot , Machine Learning , Macrophages , Protein S , Humans , Biomarkers/metabolism , Computational Biology , Diabetic Foot/genetics , Diabetic Foot/metabolism , Efferocytosis , Macrophages/metabolism , Phagocytosis/genetics , Protein S/genetics
12.
Front Immunol ; 15: 1355315, 2024.
Article in English | MEDLINE | ID: mdl-38558807

ABSTRACT

Macrophage activation syndrome (MAS) is a life-threatening complication of systemic juvenile arthritis, accompanied by cytokine storm and hemophagocytosis. In addition, COVID-19-related hyperinflammation shares clinical features of MAS. Mechanisms that activate macrophages in MAS remain unclear. Here, we identify the role of miRNA in increased phagocytosis and interleukin-12 (IL-12) production by macrophages in a murine model of MAS. MAS significantly increased F4/80+ macrophages and phagocytosis in the mouse liver. Gene expression profile revealed the induction of Fcγ receptor-mediated phagocytosis (FGRP) and IL-12 production in the liver. Phagocytosis pathways such as High-affinity IgE receptor is known as Fc epsilon RI -signaling and pattern recognition receptors involved in the recognition of bacteria and viruses and phagosome formation were also significantly upregulated. In MAS, miR-136-5p and miR-501-3p targeted and caused increased expression of Fcgr3, Fcgr4, and Fcgr1 genes in FGRP pathway and consequent increase in phagocytosis by macrophages, whereas miR-129-1-3p and miR-150-3p targeted and induced Il-12. Transcriptome analysis of patients with MAS revealed the upregulation of FGRP and FCGR gene expression. A target analysis of gene expression data from a patient with MAS discovered that miR-136-5p targets FCGR2A and FCGR3A/3B, the human orthologs of mouse Fcgr3 and Fcgr4, and miR-501-3p targets FCGR1A, the human ortholog of mouse Fcgr1. Together, we demonstrate the novel role of miRNAs during MAS pathogenesis, thereby suggesting miRNA mimic-based therapy to control the hyperactivation of macrophages in patients with MAS as well as use overexpression of FCGR genes as a marker for MAS classification.


Subject(s)
Macrophage Activation Syndrome , MicroRNAs , Humans , Animals , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Receptors, IgG/genetics , Macrophage Activation Syndrome/genetics , Phagocytosis/genetics , Interleukin-12
13.
Cell Rep ; 43(4): 114073, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38578825

ABSTRACT

Macrophages are central innate immune cells whose function declines with age. The molecular mechanisms underlying age-related changes remain poorly understood, particularly in human macrophages. We report a substantial reduction in phagocytosis, migration, and chemotaxis in human monocyte-derived macrophages (MDMs) from older (>50 years old) compared with younger (18-30 years old) donors, alongside downregulation of transcription factors MYC and USF1. In MDMs from young donors, knockdown of MYC or USF1 decreases phagocytosis and chemotaxis and alters the expression of associated genes, alongside adhesion and extracellular matrix remodeling. A concordant dysregulation of MYC and USF1 target genes is also seen in MDMs from older donors. Furthermore, older age and loss of either MYC or USF1 in MDMs leads to an increased cell size, altered morphology, and reduced actin content. Together, these results define MYC and USF1 as key drivers of MDM age-related functional decline and identify downstream targets to improve macrophage function in aging.


Subject(s)
Aging , Macrophages , Phagocytosis , Proto-Oncogene Proteins c-myc , Upstream Stimulatory Factors , Humans , Macrophages/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Proto-Oncogene Proteins c-myc/genetics , Adult , Upstream Stimulatory Factors/metabolism , Upstream Stimulatory Factors/genetics , Middle Aged , Adolescent , Phagocytosis/genetics , Young Adult , Transcription, Genetic , Aged , Chemotaxis/genetics
14.
Mol Immunol ; 170: 35-45, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38613944

ABSTRACT

Microglia play a pivotal role in the pathology of Alzheimer's Disease (AD), with the Triggering Receptor Expressed on Myeloid cells 2 (TREM2) central to their neuroprotective functions. The R47H variant of TREM2 has emerged as a significant genetic risk factor for AD, leading to a loss-of-function phenotype in mouse AD models. This study elucidates the roles of TREM2 in human microglia-like HMC3 cells and the regulation of these functions by SH2-containing inositol-5'-phosphatase 1 (SHIP1). Using stable cell lines expressing wild-type TREM2, the R47H variant, and TREM2-deficient lines, we found that functional TREM2 is essential for the phagocytosis of Aß, lysosomal capacity, and mitochondrial activity. Notably, the R47H variant displayed increased phagocytic activity towards apoptotic neurons. Introducing SHIP1, known to modulate TREM2 signaling in other cells, revealed its role as a negative regulator of these TREM2-mediated functions. Moreover, pharmacological inhibition of both SHIP1 and its isoform SHIP2 amplified Aß phagocytosis and lysosomal capacity, independently of TREM2 or SHIP1 expression, suggesting a potential regulatory role for SHIP2 in these functions. The absence of TREM2, combined with the presence of both SHIP isoforms, suppressed mitochondrial activity. However, pan-SHIP1/2 inhibition enhanced mitochondrial function in these cells. In summary, our findings offer a deeper understanding of the relationship between TREM2 variants and SHIP1 in microglial functions, and emphasize the therapeutic potential of targeting the TREM2 and SHIP1 pathways in microglia for neurodegenerative diseases.


Subject(s)
Membrane Glycoproteins , Microglia , Phagocytosis , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases , Receptors, Immunologic , Animals , Humans , Alzheimer Disease/metabolism , Alzheimer Disease/genetics , Amyloid beta-Peptides/metabolism , Apoptosis/genetics , Cell Line , Lysosomes/metabolism , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Microglia/metabolism , Mitochondria/metabolism , Phagocytosis/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Signal Transduction
15.
Biochem Biophys Res Commun ; 715: 150007, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38678783

ABSTRACT

Smad4, a critical mediator of TGF-ß signaling, plays a pivotal role in regulating various cellular functions, including immune responses. In this study, we investigated the impact of Smad4 knockout specifically in macrophages on anti-tumor immunity, focusing on lung metastasis of B16 melanoma cells. Using a mouse model with Smad4 knockout in macrophages established via Lyz2-cre mice and Smad4 flox/flox mice, we demonstrated a significant inhibition of B16 metastasis in the lungs. Interestingly, the inhibition of tumor growth was found to be independent of adaptive immunity, as no significant changes were observed in the numbers or activities of T cells, B cells, or NK cells. Instead, Smad4 knockout led to the emergence of an MCHIIlow CD206high subset of lung interstitial macrophages, characterized by enhanced phagocytosis function. Our findings highlight the crucial role of Smad4 in modulating the innate immune response against tumors and provide insights into potential therapeutic strategies targeting lung interstitial macrophages to enhance anti-tumor immunity.


Subject(s)
Lung Neoplasms , Melanoma, Experimental , Phagocytosis , Smad4 Protein , Animals , Mice , Cell Line, Tumor , Lung/pathology , Lung/immunology , Lung/metabolism , Lung Neoplasms/secondary , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Lung Neoplasms/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Melanoma, Experimental/pathology , Melanoma, Experimental/immunology , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/genetics , Smad4 Protein/deficiency , Smad4 Protein/genetics , Smad4 Protein/metabolism
16.
Cell Rep Med ; 5(3): 101450, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38508139

ABSTRACT

CD47 is a ligand of SIRPα, an inhibitory receptor expressed by macrophages, dendritic cells, and natural killer (NK) cells, and, therefore, transgenic overexpression of CD47 is considered an effective approach to inhibiting transplant rejection. However, the detrimental effect of CD47 signaling is overlooked when exploring this approach. Here, we construct a mutant CD47 by replacing the transmembrane and intracellular domains with a membrane anchor (CD47-IgV). In both human and mouse cells, CD47-IgV is efficiently expressed on the cell surface and protects against phagocytosis in vitro and in vivo but does not induce cell death or inhibit angiogenesis. Furthermore, hematopoietic stem cells expressing transgenic CD47-IgV show no detectable alterations in engraftment or differentiation. This study provides a potentially effective means of achieving transgenic CD47 expression that may help to produce gene-edited pigs for xenotransplantation and hypoimmunogenic pluripotent stem cells for regenerative medicine.


Subject(s)
Angiogenesis , CD47 Antigen , Animals , Humans , Mice , CD47 Antigen/genetics , CD47 Antigen/metabolism , Cell Death , Phagocytosis/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Swine
17.
J Neuroinflammation ; 21(1): 74, 2024 Mar 25.
Article in English | MEDLINE | ID: mdl-38528525

ABSTRACT

The retinal pigment epithelium (RPE) maintains photoreceptor viability and function, completes the visual cycle, and forms the outer blood-retinal barrier (oBRB). Loss of RPE function gives rise to several monogenic retinal dystrophies and contributes to age-related macular degeneration. Retinal detachment (RD) causes separation of the neurosensory retina from the underlying RPE, disrupting the functional and metabolic relationships between these layers. Although the retinal response to RD is highly studied, little is known about how the RPE responds to loss of this interaction. RNA sequencing (RNA-Seq) was used to compare normal and detached RPE in the C57BL6/J mouse. The naïve mouse RPE transcriptome was compared to previously published RPE signature gene lists and from the union of these 14 genes (Bmp4, Crim1, Degs1, Gja1, Itgav, Mfap3l, Pdpn, Ptgds, Rbp1, Rnf13, Rpe65, Slc4a2, Sulf1 and Ttr) representing a core signature gene set applicable across rodent and human RPE was derived. Gene ontology enrichment analysis (GOEA) of the mouse RPE transcriptome identified expected RPE features and functions, such as pigmentation, phagocytosis, lysosomal and proteasomal degradation of proteins, and barrier function. Differentially expressed genes (DEG) at 1 and 7 days post retinal detachment (dprd) were defined as mRNA with a significant (padj≤0.05) fold change (FC) of 0.67 ≥ FC ≥ 1.5 in detached versus naïve RPE. The RPE transcriptome exhibited dramatic changes at 1 dprd, with 2297 DEG identified. The KEGG pathways and biological process GO groups related to innate immune responses were significantly enriched. Lipocalin 2 (Lcn2) and several chemokines were upregulated, while numerous genes related to RPE functions, such as pigment synthesis, visual cycle, phagocytosis, and tight junctions were downregulated at 1 dprd. The response was largely transient, with only 18 significant DEG identified at 7 dprd, including upregulation of complement gene C4b. Validation studies confirmed RNA-Seq results. Thus, the RPE quickly downregulates cell-specific functions and mounts an innate immune defense response following RD. Our data demonstrate that the RPE contributes to the inflammatory response to RD and may play a role in attraction of immune cells to the subretinal space.


Subject(s)
Macular Degeneration , Retinal Detachment , Mice , Animals , Humans , Retinal Pigment Epithelium/metabolism , Retinal Detachment/metabolism , Retina/metabolism , Macular Degeneration/metabolism , Phagocytosis/genetics , Bone Morphogenetic Protein Receptors/metabolism
18.
Alzheimers Dement ; 20(7): 4914-4934, 2024 07.
Article in English | MEDLINE | ID: mdl-38506634

ABSTRACT

BACKGROUND: Variants in ABCA7, a member of the ABC transporter superfamily, have been associated with increased risk for developing late onset Alzheimer's disease (LOAD). METHODS: CRISPR-Cas9 was used to generate an Abca7V1613M variant in mice, modeling the homologous human ABCA7V1599M variant, and extensive characterization was performed. RESULTS: Abca7V1613M microglia show differential gene expression profiles upon lipopolysaccharide challenge and increased phagocytic capacity. Homozygous Abca7V1613M mice display elevated circulating cholesterol and altered brain lipid composition. When crossed with 5xFAD mice, homozygous Abca7V1613M mice display fewer Thioflavin S-positive plaques, decreased amyloid beta (Aß) peptides, and altered amyloid precursor protein processing and trafficking. They also exhibit reduced Aß-associated inflammation, gliosis, and neuronal damage. DISCUSSION: Overall, homozygosity for the Abca7V1613M variant influences phagocytosis, response to inflammation, lipid metabolism, Aß pathology, and neuronal damage in mice. This variant may confer a gain of function and offer a protective effect against Alzheimer's disease-related pathology. HIGHLIGHTS: ABCA7 recognized as a top 10 risk gene for developing Alzheimer's disease. Loss of function mutations result in increased risk for LOAD. V1613M variant reduces amyloid beta plaque burden in 5xFAD mice. V1613M variant modulates APP processing and trafficking in 5xFAD mice. V1613M variant reduces amyloid beta-associated damage in 5xFAD mice.


Subject(s)
ATP-Binding Cassette Transporters , Alzheimer Disease , Amyloid beta-Peptides , Mice, Transgenic , Plaque, Amyloid , Animals , Mice , ATP-Binding Cassette Transporters/genetics , ATP-Binding Cassette Transporters/metabolism , Plaque, Amyloid/pathology , Plaque, Amyloid/genetics , Plaque, Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Alzheimer Disease/metabolism , Neurons/metabolism , Neurons/pathology , Disease Models, Animal , Humans , Brain/pathology , Brain/metabolism , Microglia/metabolism , Microglia/pathology , Phagocytosis/genetics , Amyloid beta-Protein Precursor/genetics
19.
Int Immunopharmacol ; 130: 111708, 2024 Mar 30.
Article in English | MEDLINE | ID: mdl-38394889

ABSTRACT

Immunoevasion has been a severe obstacle for the clinical treatment of breast cancer (BC). CD47, known as an anti-phagocytic molecule, plays a key role in governing the evasion of tumor cells from immune surveillance by interacting with signal-regulated protein α (SIRPα) on macrophages. Here, we report for the first time that miR-299-3p is a direct regulator of CD47 with tumor suppressive effects both in vitro and in vivo. miRNA expression profiles and overall survival of BC cohorts from the Cancer Genome Atlas, METABRIC, or GSE19783 datasets showed that miR-299-3p is downregulated in BC tissues and that BC patients with low levels of miR-299-3p have poorer prognoses. Using dual-luciferase reporter, qRT-PCR, Western blot, and phagocytosis assays, we proved that restoration of miR-299-3p can suppress CD47 expression by directly targeting the predicted seed sequence "CCCACAU" in its 3'-UTR, leading to phagocytosis of BC cells by macrophages, whereas miR-299-3p inhibition or deletion reversed this effect. Additionally, Gene Ontology (GO) analysis and a variety of confirmatory experiments revealed that miR-299-3p was inversely correlated with cell proliferation, migration, and the cell cycle process. Mechanistically, miR-299-3p can also directly target ABCE1, an essential ribosome recycling factor, alleviating these malignant phenotypes of BC cells. In vivo BC xenografts based on nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice further proved that restoration of miR-299-3p resulted in a significant suppression of tumorigenesis and a promotion of macrophage activation and infiltration. Overall, our study suggested that miR-299-3p is a potent inhibitor of CD47 and ABCE1 to exhibit bifunctional BC-suppressing effects through immune activation conjugated with malignant behavior inhibition in breast carcinogenesis and thus can potentially serve as a novel therapeutic target for BC.


Subject(s)
ATP-Binding Cassette Transporters , Breast Neoplasms , CD47 Antigen , MicroRNAs , Tumor Escape , Animals , Female , Humans , Mice , ATP-Binding Cassette Transporters/metabolism , Breast Neoplasms/immunology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinogenesis , CD47 Antigen/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Macrophages/metabolism , Mice, Inbred NOD , Mice, SCID , MicroRNAs/metabolism , Phagocytosis/genetics , Phenotype
20.
Mol Neurobiol ; 61(9): 6920-6933, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38363533

ABSTRACT

Multiple sclerosis (MS) is a leading cause of chronic neurological dysfunction in young to middle-aged adults, affecting approximately 2.5 million people worldwide. It is characterized by inflammation, multifocal demyelination, axonal loss, and white and gray matter gliosis. Autophagy is a highly conserved protein degradation pathway. Polymorphisms in autophagy-related genes have been implicated in a variety of autoimmune diseases, including systemic lupus erythematosus, rheumatoid arthritis, psoriasis and MS. However, the significance of autophagy in MS remains to be elucidated. This paper aims to explore the potential role of autophagy-related genes in MS diseases by using bioinformatics combined with machine learning methods. Finally, we obtained 9 autophagy genes with the highest correlation with MS, and further changes in these autophagy genes were verified in the experimental autoimmune encephalomyelitis (EAE) model and oligodendrocyte precursor cells (OPCs) engulfed myelin debris (MD). Combined with bioinformatic analysis and experimental data, Becn1 showed obvious expression abnormalities suggesting that this gene has vital functions in autophagy and MD engulfed by OPCs. This work will be of great significance for the further exploration of autophagy-related genes in demyelinating diseases.


Subject(s)
Autophagy , Multiple Sclerosis , Oligodendrocyte Precursor Cells , Phagocytosis , Autophagy/genetics , Multiple Sclerosis/genetics , Multiple Sclerosis/immunology , Multiple Sclerosis/pathology , Phagocytosis/genetics , Oligodendrocyte Precursor Cells/immunology , Computational Biology , Machine Learning , Animals , Mice , Female , Mice, Inbred C57BL , Beclin-1/genetics , Beclin-1/immunology , Gene Expression Profiling , Myelin Sheath/metabolism , Myelin Sheath/pathology
SELECTION OF CITATIONS
SEARCH DETAIL