Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 417
Filter
1.
Immun Inflamm Dis ; 12(10): e70034, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39392260

ABSTRACT

INTRODUCTION: Systemic sclerosis (SSc) is a complex autoimmune disease characterized by fibrosis, vascular damage, and immune dysregulation. Fractalkine or chemokine (C-X3-C motif) ligand 1 (CX3CL1), a chemokine and adhesion molecule, along with its receptor CX3CR1, have been implicated in the inflammatory processes of SSc. CX3CL1 functions as both a chemoattractant and an adhesion molecule, guiding immune cell trafficking. This systematic review examines the role of CX3CL1 and its receptor CX3CR1 in the pathogenesis of SSc, with a focus on pulmonary and vascular complications. METHODS: A systematic literature search was conducted across databases including PubMed, Scopus, and Web of Science from inception to November 2020. The search focused on studies investigating the CX3CL1/CX3CR1 axis in the context of SSc. RESULTS: The review identified elevated CX3CL1 expression in SSc patients, particularly in the skin and lungs, where CX3CR1 is expressed on infiltrating immune cells. Higher levels of CX3CL1 were correlated with the severity of interstitial lung disease in SSc patients, indicating a potential predictive marker for disease progression. CX3CR1-positive monocytes and NK cells were recruited to inflamed tissues, contributing to fibrosis and tissue damage. Animal studies showed that inhibition of the CX3CL1/CX3CR1 axis reduced fibrosis and improved vascular function. CONCLUSION: The CX3CL1/CX3CR1 axis plays a key role in immune cell recruitment and fibrosis in SSc. Elevated CX3CL1 levels are associated with lung and vascular complications, making it a potential biomarker for disease progression and a promising therapeutic target.


Subject(s)
CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Scleroderma, Systemic , Humans , Scleroderma, Systemic/immunology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Chemokine CX3CL1/metabolism , Chemokine CX3CL1/genetics , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Animals , Biomarkers
2.
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
3.
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
4.
EBioMedicine ; 107: 105318, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39217853

ABSTRACT

Severe respiratory syncytial virus (RSV) disease is a significant contributor to the global burden of disease in infants and children. The RSV attachment protein (G) has been shown to be critical in invading airway epithelial cells through its CX3C motif interacting with the host receptor CX3CR1. The ubiquitous expression of this receptor on immune cells may explain their susceptibility to RSV infection. The RSV G protein may enhance disease severity through reprogramming of normal cellular functionality leading to inhibition of antiviral responses. While existing preventives targeting the RSV fusion (F) protein are highly effective, there are no RSV therapeutics based on the G protein to limit RSV pathogenesis. Monoclonal antibodies targeting the RSV G protein administered as post-infection therapeutics in mice have been shown to improve the antiviral response, reduce viral load and limit disease severity. Further research is required to better understand how RSV infection of immune cells contributes to pathogenesis for the development of more targeted and efficacious therapeutics.


Subject(s)
Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Animals , Respiratory Syncytial Virus, Human/immunology , Host-Pathogen Interactions/immunology , CX3C Chemokine Receptor 1/metabolism , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism , Mice , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal/pharmacology
5.
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
6.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337288

ABSTRACT

Respiratory syncytial virus (RSV) is a common respiratory pathogen that causes respiratory illnesses, ranging from mild symptoms to severe lower respiratory tract infections in infants and older adults. This virus is responsible for one-third of pneumonia deaths in the pediatric population; however, there are currently only a few effective vaccines. A better understanding of the RSV-host relationship at the molecular level may lead to a more effective management of RSV-related symptoms. The fractalkine (CX3CL1) receptor (CX3CR1) is a co-receptor for RSV expressed by airway epithelial cells and diverse immune cells. RSV G protein binds to the CX3CR1 receptor via a highly conserved amino acid motif (CX3C motif), which is also present in CX3CL1. The CX3CL1-CX3CR1 axis is involved in the activation and infiltration of immune cells into the infected lung. The presence of the RSV G protein alters the natural functions of the CX3CR1-CX3CL1 axis and modifies the host's immune response, an aspects that need to be considered in the development of an efficient vaccine and specific pharmacological treatment.


Subject(s)
CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus Infections/metabolism , Chemokine CX3CL1/metabolism , CX3C Chemokine Receptor 1/metabolism , Respiratory Syncytial Virus, Human/immunology , Animals
7.
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
8.
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
9.
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
10.
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
11.
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
12.
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
13.
Int J Mol Sci ; 25(14)2024 Jul 09.
Article in English | MEDLINE | ID: mdl-39062768

ABSTRACT

Diabetes mellitus (DM) is the most common metabolic disease in humans, and its prevalence is increasing worldwide in parallel with the obesity pandemic. A lack of insulin or insulin resistance, and consequently hyperglycemia, leads to many systemic disorders, among which diabetic encephalopathy (DE) is a long-term complication of the central nervous system (CNS), characterized by cognitive impairment and motor dysfunctions. The role of oxidative stress and neuroinflammation in the pathomechanism of DE has been proven. Fractalkine (CX3CL1) has unique properties as an adhesion molecule and chemoattractant, and by acting on its only receptor, CX3CR1, it regulates the activity of microglia in physiological states and neuroinflammation. Depending on the clinical context, CX3CL1-CX3CR1 signaling may have neuroprotective effects by inhibiting the inflammatory process in microglia or, conversely, maintaining/intensifying inflammation and neurotoxicity. This review discusses the evidence supporting that the CX3CL1-CX3CR1 pair is neuroprotective and other evidence that it is neurotoxic. Therefore, interrupting the vicious cycle within neuron-microglia interactions by promoting neuroprotective effects or inhibiting the neurotoxic effects of the CX3CL1-CX3CR1 signaling axis may be a therapeutic goal in DE by limiting the inflammatory response. However, the optimal approach to prevent DE is simply tight glycemic control, because the elimination of dysglycemic states in the CNS abolishes the fundamental mechanisms that induce this vicious cycle.


Subject(s)
Chemokine CX3CL1 , Microglia , Signal Transduction , Humans , Chemokine CX3CL1/metabolism , Animals , Microglia/metabolism , Microglia/pathology , CX3C Chemokine Receptor 1/metabolism
14.
Cell Rep ; 43(7): 114490, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38990720

ABSTRACT

Although oral tolerance is a critical system in regulating allergic disorders, the mechanisms by which dietary factors regulate the induction and maintenance of oral tolerance remain unclear. To address this, we explored the differentiation and function of various immune cells in the intestinal immune system under fasting and ad libitum-fed conditions before oral ovalbumin (OVA) administration. Fasting mitigated OVA-specific Treg expansion, which is essential for oral tolerance induction. This abnormality mainly resulted from functional defects in the CX3CR1+ cells responsible for the uptake of luminal OVA and reduction of tolerogenic CD103+ dendritic cells. Eventually, fasting impaired the preventive effect of oral OVA administration on asthma and allergic rhinitis development. Specific food ingredients, namely carbohydrates and arginine, were indispensable for oral tolerance induction by activating glycolysis and mTOR signaling. Overall, prior food intake and nutritional signals are critical for maintaining immune homeostasis by inducing tolerance to ingested food antigens.


Subject(s)
Arginine , Dendritic Cells , Immune Tolerance , Ovalbumin , T-Lymphocytes, Regulatory , TOR Serine-Threonine Kinases , Animals , Arginine/metabolism , T-Lymphocytes, Regulatory/immunology , Ovalbumin/immunology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Mice , TOR Serine-Threonine Kinases/metabolism , Mice, Inbred C57BL , Administration, Oral , CX3C Chemokine Receptor 1/metabolism , Intestines/immunology , Antigens, CD/metabolism , Integrin alpha Chains/metabolism , Sugars/metabolism , Glycolysis , Fasting , Signal Transduction , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Female
15.
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
16.
Cytokine ; 181: 156684, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38936205

ABSTRACT

As a versatile element for maintaining homeostasis, the chemokine system has been reported to be implicated in the pathogenesis of immune thrombocytopenia (ITP). However, research pertaining to chemokine receptors and related ligands in adult ITP is still limited. The states of several typical chemokine receptors and cognate ligands in the circulation were comparatively assessed through various methodologies. Multiple variable analyses of correlation matrixes were conducted to characterize the correlation signatures of various chemokine receptors or candidate ligands with platelet counts. Our data illustrated a significant decrease in relative CXCR3 expression and elevated plasma levels of CXCL4, 9-11, 13, and CCL3 chemokines in ITP patients with varied platelet counts. Flow cytometry assays revealed eminently diminished CXCR3 levels on T and B lymphocytes and increased CXCR5 on cytotoxic T cell (Tc) subsets in ITP patients with certain platelet counts. Meanwhile, circulating CX3CR1 levels were markedly higher on T cells with a concomitant increase in plasma CX3CL1 level in ITP patients, highlighting the importance of aberrant alterations of the CX3CR1-CX3CL1 axis in ITP pathogenesis. Spearman's correlation analyses revealed a strong positive association of peripheral CXCL4 mRNA level, and negative correlations of plasma CXCL4 concentration and certain chemokine receptors with platelet counts, which might serve as a potential biomarker of platelet destruction in ITP development. Overall, these results indicate that the differential expression patterns and distinct activation states of peripheral chemokine network, and the subsequent expansion of circulating CXCR5+ Tc cells and CX3CR1+ T cells, may be a hallmark during ITP progression, which ultimately contributes to thrombocytopenia in ITP patients.


Subject(s)
CX3C Chemokine Receptor 1 , Purpura, Thrombocytopenic, Idiopathic , Receptors, CXCR3 , Receptors, CXCR5 , Humans , Receptors, CXCR3/metabolism , Purpura, Thrombocytopenic, Idiopathic/blood , Purpura, Thrombocytopenic, Idiopathic/immunology , CX3C Chemokine Receptor 1/metabolism , Male , Receptors, CXCR5/metabolism , Female , Adult , Middle Aged , Platelet Count , Platelet Factor 4/blood , Platelet Factor 4/metabolism , Aged , B-Lymphocytes/immunology , B-Lymphocytes/metabolism
17.
Cell Mol Life Sci ; 81(1): 267, 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38884678

ABSTRACT

Neutralizing antibodies are considered a correlate of protection against severe human respiratory syncytial virus (HRSV) disease. Currently, HRSV neutralization assays are performed on immortalized cell lines like Vero or A549 cells. It is known that assays on these cell lines exclusively detect neutralizing antibodies (nAbs) directed to the fusion (F) protein. For the detection of nAbs directed to the glycoprotein (G), ciliated epithelial cells expressing the cellular receptor CX3CR1 are required, but generation of primary cell cultures is expensive and labor-intensive. Here, we developed a high-throughput neutralization assay based on the interaction between clinically relevant HRSV grown on primary cells with ciliated epithelial cells, and validated this assay using a panel of infant sera. To develop the high-throughput neutralization assay, we established a culture of differentiated apical-out airway organoids (Ap-O AO). CX3CR1 expression was confirmed, and both F- and G-specific monoclonal antibodies neutralized HRSV in the Ap-O AO. In a side-by-side neutralization assay on Vero cells and Ap-O AO, neutralizing antibody levels in sera from 125 infants correlated well, although titers on Ap-O AO were consistently lower. We speculate that these lower titers might be an actual reflection of the neutralizing antibody capacity in vivo. The organoid-based neutralization assay described here holds promise for further characterization of correlates of protection against HRSV disease.


Subject(s)
Antibodies, Neutralizing , CX3C Chemokine Receptor 1 , Neutralization Tests , Organoids , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Human , Humans , Respiratory Syncytial Virus, Human/immunology , Antibodies, Neutralizing/immunology , Organoids/metabolism , Organoids/immunology , Organoids/virology , Organoids/cytology , Animals , Neutralization Tests/methods , Chlorocebus aethiops , Vero Cells , Respiratory Syncytial Virus Infections/immunology , Respiratory Syncytial Virus Infections/virology , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/immunology , Antibodies, Viral/immunology , Viral Fusion Proteins/immunology , Viral Fusion Proteins/metabolism , Infant , Epithelial Cells/metabolism , Epithelial Cells/immunology , Epithelial Cells/virology , Antibodies, Monoclonal/immunology
18.
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
19.
Biomed Pharmacother ; 177: 116929, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38889644

ABSTRACT

Acute kidney injury (AKI) is a devastating consequence of sepsis, accompanied by high mortality rates. It was suggested that inflammatory pathways are closely linked to the pathogenesis of lipopolysaccharide (LPS)-induced AKI. Inflammatory signaling, including PCSK9, HMGB1/RAGE/TLR4/MYD88/NF-κB, NLRP3/caspase-1 and Fractalkine/CX3CR1 are considered major forerunners in this link. Alirocumab, PCSK9 inhibitor, with remarkable anti-inflammatory features. Accordingly, this study aimed to elucidate the antibacterial effect of alirocumab against E. coli in vitro. Additionally, evaluation of the potential nephroprotective effects of alirocumab against LPS-induced AKI in rats, highlighting the potential underlying mechanisms involved in these beneficial actions. Thirty-six adult male Wistar rats were assorted into three groups (n=12). Group I; was a normal control group, whereas sepsis-mediated AKI was induced in groups II and III through single-dose intraperitoneal injection of LPS on day 16. In group III, animals were given alirocumab. The results revealed that LPS-induced AKI was mitigated by alirocumab, evidenced by amelioration in renal function tests (creatinine, cystatin C, KIM-1, and NGAL); oxidative stress biomarkers (Nrf2, HO-1, TAC, and MDA); apoptotic markers and renal histopathological findings. Besides, alirocumab pronouncedly hindered LPS-mediated inflammatory response, confirmed by diminishing HMGB1, TNF-α, IL-1ß, and caspase-1 contents; the gene expression of PCSK9, RAGE, NF-ᴋB and Fractalkine/CX3CR1, along with mRNA expression of TLR4, MYD88, and NLRP3. Regarding the antibacterial actions, results showed that alirocumab displayed potential anti-bacterial activity against pathogenic gram-negative E. coli. In conclusion, alirocumab elicited nephroprotective activities against LPS-induced AKI via modulation of Nrf2/HO-1, PCSK9, HMGB1/RAGE/TLR4/MYD88/NF-ᴋB/NLRP3/Caspase-1, Fractalkine/CX3R1 and apoptotic axes.


Subject(s)
Acute Kidney Injury , Antibodies, Monoclonal, Humanized , Antioxidants , CX3C Chemokine Receptor 1 , Chemokine CX3CL1 , HMGB1 Protein , Heme Oxygenase (Decyclizing) , NF-E2-Related Factor 2 , NF-kappa B , NLR Family, Pyrin Domain-Containing 3 Protein , Rats, Wistar , Sepsis , Signal Transduction , Animals , Male , Sepsis/complications , Sepsis/drug therapy , Sepsis/metabolism , HMGB1 Protein/metabolism , Chemokine CX3CL1/metabolism , Acute Kidney Injury/metabolism , Acute Kidney Injury/drug therapy , Acute Kidney Injury/chemically induced , Acute Kidney Injury/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Rats , Heme Oxygenase (Decyclizing)/metabolism , Antibodies, Monoclonal, Humanized/pharmacology , NF-kappa B/metabolism , NF-E2-Related Factor 2/metabolism , Antioxidants/pharmacology , CX3C Chemokine Receptor 1/metabolism , Signal Transduction/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Disease Models, Animal , Lipopolysaccharides , PCSK9 Inhibitors , Kidney/drug effects , Kidney/pathology , Kidney/metabolism , Proprotein Convertase 9/metabolism , Proprotein Convertase 9/genetics , Oxidative Stress/drug effects , Anti-Inflammatory Agents/pharmacology
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL