Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 298
Filter
1.
Sci Rep ; 14(1): 23021, 2024 10 03.
Article in English | MEDLINE | ID: mdl-39362935

ABSTRACT

Colorectal cancer (CRC) resulting from chronic inflammation is a crucial issue in patients with inflammatory bowel disease (IBD). Although many reports established that intestinal resident CX3CR1high macrophages play an essential role in suppressing intestinal inflammation, their function in colitis-related CRC remains unclear. In this study, we found that colonic CX3CR1high macrophages, which were positive for MHC-II, F4/80 and CD319, promoted colitis-associated CRC. They highly expressed Col1a1, Tgfb, II10, and II4, and were considered to be fibrocytes with an immunosuppressive M2-like phenotype. CX3CR1 deficiency led to reductions in the absolute numbers of CX3CR1high fibrocytes through increased apoptosis, thereby preventing the development of colitis-associated CRC. We next focused statins as drugs targeting CX3CR1high fibrocytes. Statins have been actively discussed for patients with IBD and reported to suppress the CX3CL1/CX3CR1 axis. Statin treatment after azoxymethane/dextran sulfate sodium-induced inflammation reduced CX3CR1high fibrocyte counts and suppressed colitis-associated CRC. Therefore, CX3CR1high fibrocytes represent a potential target for carcinogenesis-preventing therapy, and statins could be safe therapeutic candidates for IBD.


Subject(s)
CX3C Chemokine Receptor 1 , Colitis , Pravastatin , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Animals , Mice , Colitis/complications , Colitis/metabolism , Colitis/pathology , Colitis/drug therapy , Pravastatin/pharmacology , Pravastatin/therapeutic use , Macrophages/metabolism , Macrophages/drug effects , Colon/pathology , Colon/drug effects , Colon/metabolism , Mice, Inbred C57BL , Colitis-Associated Neoplasms/pathology , Colitis-Associated Neoplasms/prevention & control , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/drug therapy , Carcinogenesis/drug effects , Carcinogenesis/pathology , Disease Models, Animal , Dextran Sulfate , Male , Humans
2.
J Neuroinflammation ; 21(1): 254, 2024 Oct 09.
Article in English | MEDLINE | ID: mdl-39385200

ABSTRACT

In neuroinflammation, distinguishing microglia from macrophages and identifying microglial-specific biomarkers in peripheral blood pose significant challenges. This study comprehensively profiled the extracellular vesicles (EVs) of microglia and macrophages, respectively, revealing co-expressed EVs with UCHL1 and CX3CR1 as EVs derived specifically from microglia in human blood. After extensive validation, using optimized nano flow cytometry, we evaluated plasma CX3CR1+/UCHL1+ EVs across clinical cohorts [multiple sclerosis (MS), HTLV-1 associated myelopathy (HAM), Alzheimer's disease (AD), and Parkinson's disease (PD)], along with established neurodegenerative markers (NMDAR2A and NFL). The findings discovered a notable rise in CX3CR1+/UCHL1+ EVs in MS, particularly heightened in HAM, in contrast to controls. Conversely, AD and PD exhibited unaltered or diminished levels of microglial EVs. An integrated model of CX3CR1+/UCHL1+, NMDAR2A+, and NFL+ EVs demonstrated promising diagnostic potential for distinguishing MS from controls and HAM. As to the disease duration, CX3CR1+/UCHL1+ EVs increased in the initial five years of MS, stabilizing thereafter, whereas NMDAR2A+ and NFL+ EVs remained stable initially but increased significantly in the subsequent five years, suggesting their correlation with disease duration. This study uncovers unique blood microglial EVs with potential as biomarkers for MS diagnosis, differentiation from HAM, and correlation with disease duration.


Subject(s)
Biomarkers , CX3C Chemokine Receptor 1 , Extracellular Vesicles , Microglia , Multiple Sclerosis , Humans , Biomarkers/blood , Biomarkers/metabolism , Extracellular Vesicles/metabolism , Multiple Sclerosis/blood , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Microglia/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Female , Male , Middle Aged , Ubiquitin Thiolesterase/metabolism , Adult , Aged , Cohort Studies
3.
Cell Commun Signal ; 22(1): 457, 2024 Sep 26.
Article in English | MEDLINE | ID: mdl-39327578

ABSTRACT

Microglial activation-mediated neuroinflammation is a major contributor to neuronal damage after cerebral ischemia. The Fractalkine (FKN)/CX3C chemokine receptor 1 (CX3CR1) axis plays a critical role in regulating microglial activation and neuroinflammation. The aim of this study is to ascertain the role and mechanism of FKN/CX3CR1 axis in hypoxic postconditioning (HPC)-induced anti-inflammatory and neuroprotective effects on transient global cerebral ischemia (tGCI). We found that HPC suppressed microglial activation and alleviated neuroinflammation in hippocampal CA1 after tGCI. Meanwhile, HPC upregulated the expression of FKN and CX3CR1 in neurons, but it downregulated the expression of CX3CR1 in glial cells after tGCI. In addition, the overexpression of FKN induced by the administration of FKN-carried lentivirus reduced microglial activation and inhibited neuroinflammation in CA1 after tGCI. Furthermore, silencing CX3CR1 with CX3CRi-carried lentivirus in CA1 after tGCI suppressed microglial activation and neuroinflammation and exerted neuroprotective effects. Finally, the overexpression of FKN caused a marked increase of neuronal CX3CR1 receptors, upregulated the phosphorylation of Akt, and reduced neuronal loss of rats in CA1 after tGCI. These findings demonstrated that HPC protected against neuronal damage in CA1 of tGCI rats through inhibiting microglial activation and activating Akt signaling pathway via FKN/CX3CR1 axis.


Subject(s)
CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Microglia , Neuroprotection , Animals , Male , Rats , Brain Ischemia/metabolism , Brain Ischemia/pathology , Brain Ischemia/genetics , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/pathology , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/genetics , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Ischemic Postconditioning , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Rats, Sprague-Dawley , Signal Transduction
4.
Nat Commun ; 15(1): 7077, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152118

ABSTRACT

Enoblituzumab, an immunotherapeutic agent targeting CD276, shows both safety and efficacy in activating T cells and oligodendrocyte-like cells against various cancers. Preclinical studies and mouse models suggest that therapies targeting CD276 may outperform PD1/PD-L1 blockade. However, data from mouse models indicate a significant non-responsive population to anti-CD276 treatment, with the mechanisms of resistance still unclear. In this study, we evaluate the activity of anti-CD276 antibodies in a chemically-induced murine model of head and neck squamous cell carcinoma. Using models of induced and orthotopic carcinogenesis, we identify ITGB6 as a key gene mediating differential responses to anti-CD276 treatment. Through single-cell RNA sequencing and gene-knockout mouse models, we find that ITGB6 regulates the expression of the tumor-associated chemokine CX3CL1, which recruits and activates PF4+ macrophages that express high levels of CX3CR1. Inhibition of the CX3CL1-CX3CR1 axis suppresses the infiltration and secretion of CXCL16 by PF4+ macrophages, thereby reinvigorating cytotoxic CXCR6+ CD8+ T cells and enhancing sensitivity to anti-CD276 treatment. Further investigations demonstrate that inhibiting ITGB6 restores sensitivity to PD1 antibodies in mice resistant to anti-PD1 treatment. In summary, our research reveals a resistance mechanism associated with immune checkpoint inhibitor therapy and identifies potential targets to overcome resistance in cancer treatment.


Subject(s)
B7 Antigens , Head and Neck Neoplasms , Mice, Knockout , Animals , Mice , B7 Antigens/metabolism , B7 Antigens/genetics , B7 Antigens/antagonists & inhibitors , Humans , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/drug therapy , Head and Neck Neoplasms/pathology , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/immunology , Macrophages/immunology , Macrophages/metabolism , Cell Line, Tumor , Mice, Inbred C57BL , Squamous Cell Carcinoma of Head and Neck/drug therapy , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Disease Models, Animal , Female , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Gene Expression Regulation, Neoplastic/drug effects
5.
Pharmacol Res ; 208: 107348, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39134186

ABSTRACT

Fibrosis refers to the progressive tissue lesion process characterized by excessive secretion and deposition of extracellular matrix (ECM). Abnormal fibrous tissue deposition distorts tissue architecture and leads to the progressive loss of organ function. Notably, fibrosis is one of the primary pathological appearances of many end stage illnesses, and is considered as a lethal threat to human health, especially in the elderly with ageing-related diseases. CX3C ligand 1 (CX3CL1) is the only member of chemokine CX3C and binds specifically to CX3C receptor 1 (CX3CR1). Different from other chemokines, CX3CL1 possesses both chemotactic and adhesive activity. CX3CL1/CX3CR1 axis involves in various physiological and pathological processes, and exerts a critical role in cells from the immune system, vascular system, and nervous system etc. Notably, increasing evidence has demonstrated that CX3CL1/CX3CR1 signaling pathway is closely related to the pathological process of fibrosis in multiple tissue and organs. We reviewed the crucial role of CX3CL1/CX3CR1 axis in fibrosis and ageing and systematically summarized the underlying mechanism, which offers prospective strategies of targeting CX3C for the therapy of fibrosis and ageing-related diseases.


Subject(s)
Aging , Fibrosis , Humans , Aging/metabolism , Aging/pathology , Animals , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Chemokine CX3CL1/metabolism , Signal Transduction , Chemokines, CX3C/metabolism
6.
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
7.
Development ; 151(16)2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39082371

ABSTRACT

Tissue-resident macrophages contribute to the organogenesis of many tissues. Growth of the prostate is regulated by androgens during puberty, yet androgens are considered immune suppressive. In this study, we characterized the localization, androgen receptor expression and hematopoietic origin of prostate macrophages, and transiently ablated macrophages during postnatal prostate organogenesis in the mouse. We show that myeloid cells were abundant in the prostate during puberty. However, nuclear androgen receptor expression was not detected in most macrophages. We found Cx3cr1, a marker for macrophages, monocytes and dendritic cells, expressed in interstitial macrophages surrounding the prostate and associated with nerve fibers. Furthermore, we provide evidence for the co-existence of embryonic origin, self-renewing, tissue-resident macrophages and recruited macrophages of bone-marrow monocyte origin in the prostate during puberty. Our findings suggest that prostate macrophages promote neural patterning and may shed further light on our understanding of the role of the innate immune system in prostate pathology in response to inflammation and in cancer.


Subject(s)
CX3C Chemokine Receptor 1 , Macrophages , Prostate , Receptors, Androgen , Male , Animals , Prostate/metabolism , Macrophages/metabolism , Mice , Receptors, Androgen/metabolism , Receptors, Androgen/genetics , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Mice, Inbred C57BL , Organogenesis , Monocytes/metabolism
8.
Brain Behav Immun ; 121: 1-12, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39002812

ABSTRACT

induced-pluripotent stem cell (iPSC)-derived neurospheroid (NSPH) models are an emerging in vitro toolkit to study the influence of inflammatory triggers on neurodegeneration and repair in a 3D neural environment. In contrast to their human counterpart, the absence of murine iPSC-derived NSPHs for profound characterisation and validation studies is a major experimental research gap, even though they offer the only possibility to truly compare or validate in vitro NSPH responses with in vivo brain responses. To contribute to these developments, we here describe the generation and characterisation of 5-week-old CX3CR1eGFP+/- CCR2RFP+/- murine (m)iPSC-derived bi-partite (neurons + astrocytes) and tri-partite (neurons + astrocytes + microglia) NSPH models that can be subjected to cellular activation following pro-inflammatory stimulation. First, cytokine analysis demonstrates that both bi-partite and tri-partite NSPHs can be triggered to release IL6 and CXCL10 following three days of stimulation with, respectively, TNFα + IL1ß + IFNγ and LPS + IFNγ. Additionally, immunocytochemical analysis for G3BP1 and PABPC1 revealed the development of stress granules in both bi-partite and tri-partite NSPHs after 3 days of stimulation. To further investigate the observed signs of inflammatory response and cellular stress, we performed an untargeted transcriptomic and proteomic analysis of bi- and tri-partite NSPHs under steady-state and inflammatory conditions. Here, using the combined differential gene and protein expression profiles between unstimulated and stimulated NSPHs, Ingenuity Pathway Analysis (IPA) confirms the activation of canonical pathways associated with inflammation and cellular stress in both bi-partite and tri-partite NSPHs. Moreover, our multi-omics analysis suggests a higher level of downstream inflammatory responses, impairment of homeostatic and developmental processes, as well as activation of cell death processes in stimulated tri-partite NSPHs compared to bi-partite NSPHs. Concluding, these results emphasise the advantages of including microglia in NSPH research to study inflammation-induced neurodegeneration in a 3D neural environment.


Subject(s)
Induced Pluripotent Stem Cells , Inflammation , Microglia , Neurons , Proteomics , Transcriptome , Animals , Mice , Induced Pluripotent Stem Cells/metabolism , Proteomics/methods , Inflammation/metabolism , Microglia/metabolism , Neurons/metabolism , Astrocytes/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Cell Differentiation , Cytokines/metabolism , Proteome/metabolism , Chemokine CXCL10/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/genetics
9.
Neurosci Biobehav Rev ; 165: 105834, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39084583

ABSTRACT

Microglia, the intrinsic neuroimmune cells residing in the central nervous system (CNS), exert a pivotal influence on brain development, homeostasis, and functionality, encompassing critical roles during both aging and pathological states. Recent advancements in comprehending brain plasticity and functions have spotlighted conspicuous variances between male and female brains, notably in neurogenesis, neuronal myelination, axon fasciculation, and synaptogenesis. Nevertheless, the precise impact of microglia on sex-specific brain cell plasticity, sculpting diverse neural network architectures and circuits, remains largely unexplored. This article seeks to unravel the present understanding of microglial involvement in brain development, plasticity, and function, with a specific emphasis on microglial signaling in brain sex polymorphism. Commencing with an overview of microglia in the CNS and their associated signaling cascades, we subsequently probe recent revelations regarding molecular signaling by microglia in sex-dependent brain developmental plasticity, functions, and diseases. Notably, C-X3-C motif chemokine receptor 1 (CX3CR1), triggering receptors expressed on myeloid cells 2 (TREM2), calcium (Ca2+), and apolipoprotein E (APOE) emerge as molecular candidates significantly contributing to sex-dependent brain development and plasticity. In conclusion, we address burgeoning inquiries surrounding microglia's pivotal role in the functional diversity of developing and aging brains, contemplating their potential implications for gender-tailored therapeutic strategies in neurodegenerative diseases.


Subject(s)
Brain , Microglia , Neuronal Plasticity , Sex Characteristics , Signal Transduction , Humans , Microglia/metabolism , Microglia/physiology , Brain/growth & development , Brain/metabolism , Neuronal Plasticity/physiology , Animals , Signal Transduction/physiology , Female , Male , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics
10.
Cell Rep ; 43(7): 114385, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38935500

ABSTRACT

Oligodendrocyte death is common in aging and neurodegenerative disease. In these conditions, dying oligodendrocytes must be efficiently removed to allow remyelination and to prevent a feedforward degenerative cascade. Removal of this cellular debris is thought to primarily be carried out by resident microglia. To investigate the cellular dynamics underlying how microglia do this, we use a single-cell cortical demyelination model combined with longitudinal intravital imaging of dual-labeled transgenic mice. Following phagocytosis, single microglia clear the targeted oligodendrocyte and its myelin sheaths in one day via a precise, rapid, and stereotyped sequence. Deletion of the fractalkine receptor, CX3CR1, delays the microglial phagocytosis of the cell soma but has no effect on clearance of myelin sheaths. Unexpectedly, deletion of the phosphatidylserine receptor, MERTK, has no effect on oligodendrocyte or myelin sheath clearance. Thus, separate molecular signals are used to detect, engage, and clear distinct sub-compartments of dying oligodendrocytes to maintain tissue homeostasis.


Subject(s)
CX3C Chemokine Receptor 1 , Microglia , Oligodendroglia , Phagocytosis , c-Mer Tyrosine Kinase , Animals , Oligodendroglia/metabolism , Microglia/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , c-Mer Tyrosine Kinase/metabolism , c-Mer Tyrosine Kinase/genetics , Mice , Myelin Sheath/metabolism , Mice, Transgenic , Mice, Inbred C57BL , Cell Death
11.
Cancer Res Commun ; 4(7): 1802-1814, 2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38881188

ABSTRACT

Recent progress in single-cell profiling technologies has revealed significant phenotypic and transcriptional heterogeneity in tumor-infiltrating CD8+ T cells. However, the transition between the different states of intratumoral antigen-specific CD8+ T cells remains elusive. Here, we sought to examine the generation, transcriptomic states, and the clinical relevance of melanoma-infiltrating CD8+ T cells expressing a chemokine receptor and T-cell differentiation marker, CX3C chemokine receptor 1 (CX3CR1). Analysis of single-cell datasets revealed distinct human melanoma-infiltrating CD8+ T-cell clusters expressing genes associated with effector T-cell function but with distinguishing expression of CX3CR1 or PDCD1. No obvious impact of CX3CR1 expression in melanoma on the response to immune checkpoint inhibitor therapy was observed while increased pretreatment and on-treatment frequency of a CD8+ T-cell cluster expressing high levels of exhaustion markers was associated with poor response to the treatment. Adoptively transferred antigen-specific CX3CR1- CD8+ T cells differentiated into the CX3CR1+ subset in mice treated with FTY720, which inhibits lymphocyte egress from secondary lymphoid tissues, suggesting the intratumoral generation of CX3CR1+ CD8+ T cells rather than their trafficking from secondary lymphoid organs. Furthermore, analysis of adoptively transferred antigen-specific CD8+ T cells, in which the Cx3cr1 gene was replaced with a marker gene confirmed that CX3CR1+ CD8+ T cells could directly differentiate from the intratumoral CX3CR1- subset. These findings highlight that tumor antigen-specific CX3CR1- CD8+ T cells can fully differentiate outside the secondary lymphoid organs and generate CX3CR1+ CD8+ T cells in the tumor microenvironment, which are distinct from CD8+ T cells that express markers of exhaustion. SIGNIFICANCE: Intratumoral T cells are composed of heterogeneous subpopulations with various phenotypic and transcriptional states. This study illustrates the intratumoral generation of antigen-specific CX3CR1+ CD8+ T cells that exhibit distinct transcriptomic signatures and clinical relevance from CD8+ T cells expressing markers of exhaustion.


Subject(s)
CD8-Positive T-Lymphocytes , CX3C Chemokine Receptor 1 , Lymphocytes, Tumor-Infiltrating , Melanoma , Transcriptome , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Melanoma/immunology , Melanoma/genetics , Melanoma/pathology , Animals , Humans , Mice , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Mice, Inbred C57BL , Single-Cell Analysis , Clinical Relevance
12.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892268

ABSTRACT

The cellular distribution and changes in CX3CL1/fractalkine and its receptor CX3CR1 protein levels in the trigeminal subnucleus caudalis (TSC) of rats with unilateral infraorbital nerve ligation (IONL) were investigated on postoperation days 1, 3, 7, and 14 (POD1, POD3, POD7, and POD14, respectively) and compared with those of sham-operated and naïve controls. Behavioral tests revealed a significant increase in tactile hypersensitivity bilaterally in the vibrissal pads of both sham- and IONL-operated animals from POD1 to POD7, with a trend towards normalization in sham controls at POD14. Image analysis revealed increased CX3CL1 immunofluorescence (IF) intensities bilaterally in the TSC neurons of both sham- and IONL-operated rats at all survival periods. Reactive astrocytes in the ipsilateral TSC also displayed CX3CL1-IF from POD3 to POD14. At POD1 and POD3, microglial cells showed high levels of CX3CR1-IF, which decreased by POD7 and POD14. Conversely, CX3CR1 was increased in TSC neurons and reactive astrocytes at POD7 and POD14, which coincided with high levels of CX3CL1-IF and ADAM17-IF. This indicates that CX3CL1/CX3CR1 may be involved in reciprocal signaling between TSC neurons and reactive astrocytes. The level of CatS-IF in microglial cells suggests that soluble CX3CL1 may be involved in neuron-microglial cell signaling at POD3 and POD7, while ADAM17 allows this release at all studied time points. These results indicate an extended CX3CL1/CX3CR1 signaling axis and its role in the crosstalk between TSC neurons and glial cells during the development of trigeminal neuropathic pain.


Subject(s)
CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Signal Transduction , Animals , Chemokine CX3CL1/metabolism , Rats , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Male , Microglia/metabolism , Trigeminal Neuralgia/metabolism , Trigeminal Neuralgia/pathology , Neurons/metabolism , Astrocytes/metabolism , Neuralgia/metabolism , Neuralgia/pathology , Rats, Sprague-Dawley
13.
Int J Med Sci ; 21(8): 1385-1398, 2024.
Article in English | MEDLINE | ID: mdl-38903915

ABSTRACT

Inflammatory bowel disease (IBD) is a chronic inflammatory intestinal disease, characterized by dysregulated immune response. HDAC3 is reported to be an epigenetic brake in inflammation, playing critical roles in macrophages. However, its role in IBD is unclear. In our study, we found HDAC3 was upregulated in CX3CR1-positive cells in the mucosa from IBD mice. Conditional knockout (cKO) of Hdac3 in CX3CR1 positive cells attenuated the disease severity of Dextran Sulfate Sodium (DSS)-induced colitis. In addition, inhibition of HDAC3 with RGFP966 could also alleviate the DSS-induced tissue injury and inflammation in IBD. The RNA sequencing results revealed that Hdac3 cKO restrained DSS-induced upregulation of genes in the pathways of cytokine-cytokine receptor interaction, complement and coagulation cascades, chemokine signaling, and extracellular matrix receptor interaction. We also identified that Guanylate-Binding Protein 5 (GBP5) was transcriptionally regulated by HDAC3 in monocytes by RNA sequencing. Inhibition of HDAC3 resulted in decreased transcriptional activity of interferon-gamma-induced expression of GBP5 in CX3CR1-positive cells, such as macrophages and microglia. Overexpression of HDAC3 upregulated the transcriptional activity of GBP5 reporter. Lastly, conditional knockout of Hdac3 in macrophages (Hdac3 mKO) attenuated the disease severity of DSS-induced colitis. In conclusion, inhibition of HDAC3 in macrophages could ameliorate the disease severity and inflammatory response in colitis by regulating GBP5-NLRP3 axis, identifying a new therapeutic avenue for the treatment of colitis.


Subject(s)
Colitis , Dextran Sulfate , Histone Deacetylases , Macrophages , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Signal Transduction , Animals , Dextran Sulfate/toxicity , Dextran Sulfate/adverse effects , Histone Deacetylases/metabolism , Histone Deacetylases/genetics , Mice , Macrophages/metabolism , Macrophages/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Colitis/chemically induced , Colitis/genetics , Colitis/pathology , Colitis/metabolism , Humans , Signal Transduction/drug effects , Inflammatory Bowel Diseases/chemically induced , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/pathology , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/drug therapy , GTP-Binding Proteins/genetics , GTP-Binding Proteins/metabolism , GTP-Binding Proteins/antagonists & inhibitors , Disease Models, Animal , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Mice, Inbred C57BL , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Intestinal Mucosa/pathology , Intestinal Mucosa/metabolism , Intestinal Mucosa/drug effects , Acrylamides , Phenylenediamines
14.
Nat Commun ; 15(1): 5402, 2024 Jun 26.
Article in English | MEDLINE | ID: mdl-38926390

ABSTRACT

Acute brain slices represent a workhorse model for studying the central nervous system (CNS) from nanoscale events to complex circuits. While slice preparation inherently involves tissue damage, it is unclear how microglia, the main immune cells and damage sensors of the CNS react to this injury and shape neuronal activity ex vivo. To this end, we investigated microglial phenotypes and contribution to network organization and functioning in acute brain slices. We reveal time-dependent microglial phenotype changes influenced by complex extracellular ATP dynamics through P2Y12R and CX3CR1 signalling, which is sustained for hours in ex vivo mouse brain slices. Downregulation of P2Y12R and changes of microglia-neuron interactions occur in line with alterations in the number of excitatory and inhibitory synapses over time. Importantly, functional microglia modulate synapse sprouting, while microglial dysfunction results in markedly impaired ripple activity both ex vivo and in vivo. Collectively, our data suggest that microglia are modulators of complex neuronal networks with important roles to maintain neuronal network integrity and activity. We suggest that slice preparation can be used to model time-dependent changes of microglia-neuron interactions to reveal how microglia shape neuronal circuits in physiological and pathological conditions.


Subject(s)
Adenosine Triphosphate , Brain , CX3C Chemokine Receptor 1 , Microglia , Neurons , Receptors, Purinergic P2Y12 , Synapses , Animals , Microglia/metabolism , Adenosine Triphosphate/metabolism , Mice , Neurons/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Receptors, Purinergic P2Y12/metabolism , Receptors, Purinergic P2Y12/genetics , Brain/metabolism , Synapses/metabolism , Mice, Inbred C57BL , Phenotype , Male , Signal Transduction
15.
Biochemistry (Mosc) ; 89(5): 904-911, 2024 May.
Article in English | MEDLINE | ID: mdl-38880650

ABSTRACT

Multiple sclerosis (MS) is a complex autoimmune disease of central nervous system (CNS) characterized by the myelin sheath destruction and compromised nerve signal transmission. Understanding molecular mechanisms driving MS development is critical due to its early onset, chronic course, and therapeutic approaches based only on symptomatic treatment. Cytokines are known to play a pivotal role in the MS pathogenesis with interleukin-6 (IL-6) being one of the key mediators. This study investigates contribution of IL-6 produced by microglia and dendritic cells to the development of experimental autoimmune encephalomyelitis (EAE), a widely used mouse model of MS. Mice with conditional inactivation of IL-6 in the CX3CR1+ cells, including microglia, or CD11c+ dendritic cells, displayed less severe symptoms as compared to their wild-type counterparts. Mice with microglial IL-6 deletion exhibited an elevated proportion of regulatory T cells and reduced percentage of pathogenic IFNγ-producing CD4+ T cells, accompanied by the decrease in pro-inflammatory monocytes in the CNS at the peak of EAE. At the same time, deletion of IL-6 from microglia resulted in the increase of CCR6+ T cells and GM-CSF-producing T cells. Conversely, mice with IL-6 deficiency in the dendritic cells showed not only the previously described increase in the proportion of regulatory T cells and decrease in the proportion of TH17 cells, but also reduction in the production of GM-CSF and IFNγ in the secondary lymphoid organs. In summary, IL-6 functions during EAE depend on both the source and localization of immune response: the microglial IL-6 exerts both pathogenic and protective functions specifically in the CNS, whereas the dendritic cell-derived IL-6, in addition to being critically involved in the balance of regulatory T cells and TH17 cells, may stimulate production of cytokines associated with pathogenic functions of T cells.


Subject(s)
Dendritic Cells , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental , Interleukin-6 , Microglia , Multiple Sclerosis , Animals , Dendritic Cells/metabolism , Dendritic Cells/immunology , Mice , Interleukin-6/metabolism , Multiple Sclerosis/immunology , Multiple Sclerosis/metabolism , Multiple Sclerosis/pathology , Microglia/metabolism , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Mice, Inbred C57BL , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Receptors, CCR6/metabolism , Receptors, CCR6/genetics , Female
16.
Front Immunol ; 15: 1383607, 2024.
Article in English | MEDLINE | ID: mdl-38715600

ABSTRACT

Background: The crucial role of inflammation in aortic aneurysm (AA) is gaining prominence, while there is still a lack of key cytokines or targets for effective clinical translation. Methods: Mendelian randomization (MR) analysis was performed to identify the causal relationship between 91 circulating inflammatory proteins and AA and between 731 immune traits and AA. Bulk RNA sequencing data was utilized to demonstrate the expression profile of the paired ligand-receptor. Gene enrichment analysis, Immune infiltration, and correlation analysis were employed to deduce the potential role of CX3CR1. We used single-cell RNA sequencing data to pinpoint the localization of CX3CL1 and CX3CR1, which was further validated by multiplex immunofluorescence staining. Cellchat analysis was utilized to infer the CX3C signaling pathway. Trajectory analysis and the Cytosig database were exploited to determine the downstream effect of CX3CL1-CX3CR1. Results: We identified 4 candidates (FGF5, CX3CL1, IL20RA, and SCF) in multiple two-sample MR analyses. Subsequent analysis of the expression profile of the paired receptor revealed the significant upregulation of CX3CR1 in AA and its positive correlation with pro-inflammatory macrophages. Two sample MR between immune cell traits and AA demonstrated the potential causality between intermediate monocytes and AA. We finally deciphered in single-cell sequencing data that CX3CL1 sent by endothelial cells (ECs) acted on CX3CR1 of intermediated monocytes, leading to its recruitment and pro-inflammatory responses. Conclusion: Our study presented a genetic insight into the pathogenetic role of CX3CL1-CX3CR1 in AA, and further deciphered the CX3C signaling pathway between ECs and intermediate monocytes.


Subject(s)
Aortic Aneurysm , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Mendelian Randomization Analysis , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Humans , Chemokine CX3CL1/genetics , Chemokine CX3CL1/metabolism , Aortic Aneurysm/genetics , Aortic Aneurysm/metabolism , Gene Expression Profiling , Transcriptome , Signal Transduction , Genetic Predisposition to Disease
17.
J Am Heart Assoc ; 13(9): e034731, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38700011

ABSTRACT

BACKGROUND: Cardiac damage induced by ischemic stroke, such as arrhythmia, cardiac dysfunction, and even cardiac arrest, is referred to as cerebral-cardiac syndrome (CCS). Cardiac macrophages are reported to be closely associated with stroke-induced cardiac damage. However, the role of macrophage subsets in CCS is still unclear due to their heterogeneity. Sympathetic nerves play a significant role in regulating macrophages in cardiovascular disease. However, the role of macrophage subsets and sympathetic nerves in CCS is still unclear. METHODS AND RESULTS: In this study, a middle cerebral artery occlusion mouse model was used to simulate ischemic stroke. ECG and echocardiography were used to assess cardiac function. We used Cx3cr1GFPCcr2RFP mice and NLRP3-deficient mice in combination with Smart-seq2 RNA sequencing to confirm the role of macrophage subsets in CCS. We demonstrated that ischemic stroke-induced cardiac damage is characterized by severe cardiac dysfunction and robust infiltration of monocyte-derived macrophages into the heart. Subsequently, we identified that cardiac monocyte-derived macrophages displayed a proinflammatory profile. We also observed that cardiac dysfunction was rescued in ischemic stroke mice by blocking macrophage infiltration using a CCR2 antagonist and NLRP3-deficient mice. In addition, a cardiac sympathetic nerve retrograde tracer and a sympathectomy method were used to explore the relationship between sympathetic nerves and cardiac macrophages. We found that cardiac sympathetic nerves are significantly activated after ischemic stroke, which contributes to the infiltration of monocyte-derived macrophages and subsequent cardiac dysfunction. CONCLUSIONS: Our findings suggest a potential pathogenesis of CCS involving the cardiac sympathetic nerve-monocyte-derived macrophage axis.


Subject(s)
Disease Models, Animal , Ischemic Stroke , Macrophages , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , Macrophages/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/deficiency , Ischemic Stroke/physiopathology , Ischemic Stroke/metabolism , Ischemic Stroke/pathology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Male , Mice, Knockout , Mice , Infarction, Middle Cerebral Artery/physiopathology , Infarction, Middle Cerebral Artery/pathology , Sympathetic Nervous System/physiopathology , Myocardium/pathology , Myocardium/metabolism , Heart Diseases/etiology , Heart Diseases/physiopathology , Heart Diseases/pathology , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/deficiency
18.
Int J Mol Sci ; 25(9)2024 Apr 25.
Article in English | MEDLINE | ID: mdl-38731899

ABSTRACT

The chemotactic cytokine fractalkine (FKN, chemokine CX3CL1) has unique properties resulting from the combination of chemoattractants and adhesion molecules. The soluble form (sFKN) has chemotactic properties and strongly attracts T cells and monocytes. The membrane-bound form (mFKN) facilitates diapedesis and is responsible for cell-to-cell adhesion, especially by promoting the strong adhesion of leukocytes (monocytes) to activated endothelial cells with the subsequent formation of an extracellular matrix and angiogenesis. FKN signaling occurs via CX3CR1, which is the only known member of the CX3C chemokine receptor subfamily. Signaling within the FKN-CX3CR1 axis plays an important role in many processes related to inflammation and the immune response, which often occur simultaneously and overlap. FKN is strongly upregulated by hypoxia and/or inflammation-induced inflammatory cytokine release, and it may act locally as a key angiogenic factor in the highly hypoxic tumor microenvironment. The importance of the FKN/CX3CR1 signaling pathway in tumorigenesis and cancer metastasis results from its influence on cell adhesion, apoptosis, and cell migration. This review presents the role of the FKN signaling pathway in the context of angiogenesis in inflammation and cancer. The mechanisms determining the pro- or anti-tumor effects are presented, which are the cause of the seemingly contradictory results that create confusion regarding the therapeutic goals.


Subject(s)
CX3C Chemokine Receptor 1 , Carcinogenesis , Chemokine CX3CL1 , Inflammation , Neovascularization, Pathologic , Signal Transduction , Humans , Chemokine CX3CL1/metabolism , Neovascularization, Pathologic/metabolism , Inflammation/metabolism , Inflammation/pathology , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Neoplasms/metabolism , Neoplasms/pathology , Neoplasms/etiology , Tumor Microenvironment , Angiogenesis
19.
Cells ; 13(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38786041

ABSTRACT

Monocytes, as well as downstream macrophages and dendritic cells, are essential players in the immune system, fulfilling key roles in homeostasis as well as in inflammatory conditions. Conventionally, driven by studies on reporter models, mouse monocytes are categorized into a classical and a non-classical subset based on their inversely correlated surface expression of Ly6C/CCR2 and CX3CR1. Here, we aimed to challenge this concept by antibody staining and reporter mouse models. Therefore, we took advantage of Cx3cr1GFP and Ccr2RFP reporter mice, in which the respective gene was replaced by a fluorescent reporter protein gene. We analyzed the expression of CX3CR1 and CCR2 by flow cytometry using several validated fluorochrome-coupled antibodies and compared them with the reporter gene signal in these reporter mouse strains. Although we were able to validate the specificity of the fluorochrome-coupled flow cytometry antibodies, mouse Ly6Chigh classical and Ly6Clow non-classical monocytes showed no differences in CX3CR1 expression levels in the peripheral blood and spleen when stained with these antibodies. On the contrary, in Cx3cr1GFP reporter mice, we were able to reproduce the inverse correlation of the CX3CR1 reporter gene signal and Ly6C surface expression. Furthermore, differential CCR2 surface expression correlating with the expression of Ly6C was observed by antibody staining, but not in Ccr2RFP reporter mice. In conclusion, our data suggest that phenotyping strategies for mouse monocyte subsets should be carefully selected. In accordance with the literature, the suitability of CX3CR1 antibody staining is limited, whereas for CCR2, caution should be applied when using reporter mice.


Subject(s)
CX3C Chemokine Receptor 1 , Flow Cytometry , Monocytes , Receptors, CCR2 , Animals , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Monocytes/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Mice , Antibodies/immunology , Genes, Reporter , Phenotype , Mice, Inbred C57BL , Mice, Transgenic , Green Fluorescent Proteins/metabolism , Antigens, Ly/metabolism , Antigens, Ly/genetics
20.
Viruses ; 16(5)2024 05 14.
Article in English | MEDLINE | ID: mdl-38793659

ABSTRACT

Respiratory syncytial virus (RSV) is the most prevalent cause of acute lower respiratory infection in young children. Currently, the first RSV vaccines are approved by the FDA. Recently, N6-methyladenosine (m6A) RNA methylation has been implicated in the regulation of the viral life cycle and replication of many viruses, including RSV. m6A methylation of RSV RNA has been demonstrated to promote replication and prevent anti-viral immune responses by the host. Whether m6A is also involved in viral entry and whether m6A can also affect RSV infection via different mechanisms than methylation of viral RNA is poorly understood. Here, we identify m6A reader YTH domain-containing protein 1 (YTHDC1) as a novel negative regulator of RSV infection. We demonstrate that YTHDC1 abrogates RSV infection by reducing the expression of RSV entry receptor CX3C motif chemokine receptor 1 (CX3CR1) on the cell surface of lung epithelial cells. Altogether, these data reveal a novel role for m6A methylation and YTHDC1 in the viral entry of RSV. These findings may contribute to the development of novel treatment options to control RSV infection.


Subject(s)
CX3C Chemokine Receptor 1 , Down-Regulation , RNA Splicing Factors , Respiratory Syncytial Virus Infections , Humans , A549 Cells , Adenosine/analogs & derivatives , Adenosine/metabolism , Cell Line , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Epithelial Cells/virology , Epithelial Cells/metabolism , Host-Pathogen Interactions , Methylation , Nerve Tissue Proteins , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Respiratory Syncytial Virus, Human/physiology , RNA Splicing Factors/metabolism , RNA Splicing Factors/genetics , RNA, Viral/genetics , RNA, Viral/metabolism , Virus Internalization , Virus Replication
SELECTION OF CITATIONS
SEARCH DETAIL