Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.140
Filter
1.
Biomaterials ; 312: 122731, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39153324

ABSTRACT

Tumor-associated inflammation drives cancer progression and therapy resistance, often linked to the infiltration of monocyte-derived tumor-associated macrophages (TAMs), which are associated with poor prognosis in various cancers. To advance immunotherapies, testing on immunocompetent pre-clinical models of human tissue is crucial. We have developed an in vitro model of microvascular networks with tumor spheroids or patient tissues to assess monocyte trafficking into tumors and evaluate immunotherapies targeting the human tumor microenvironment. Our findings demonstrate that macrophages in vascularized breast and lung tumor models can enhance monocyte recruitment via CCL7 and CCL2, mediated by CSF-1R. Additionally, a multispecific antibody targeting CSF-1R, CCR2, and neutralizing TGF-ß (CSF1R/CCR2/TGF-ß Ab) repolarizes TAMs towards an anti-tumoral M1-like phenotype, reduces monocyte chemoattractant protein secretion, and blocks monocyte migration. This antibody also inhibits monocyte recruitment in patient-specific vascularized tumor models. In summary, this vascularized tumor model recapitulates the monocyte recruitment cascade, enabling functional testing of innovative therapeutic antibodies targeting TAMs in the tumor microenvironment.


Subject(s)
Monocytes , Receptor, Macrophage Colony-Stimulating Factor , Receptors, CCR2 , Tumor Microenvironment , Humans , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Monocytes/metabolism , Monocytes/immunology , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Tumor Microenvironment/immunology , Animals , Cell Line, Tumor , Female , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Mice , Cell Movement/drug effects , Neoplasms/immunology , Neoplasms/pathology
2.
Sci Adv ; 10(38): eadq1476, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39292787

ABSTRACT

Cross-talk between hematopoietic stem and progenitor cells (HSPCs) and bone marrow (BM) cells is critical for homing and sustained engraftment after transplantation. In particular, molecular and physical adaptation of sinusoidal endothelial cells (ECs) promote HSPC BM occupancy; however, signals that govern these events are not well understood. Extracellular vesicles (EVs) are mediators of cell-cell communication crucial in shaping tissue microenvironments. Here, we demonstrate that integrin α4ß7 on murine HSPC EVs targets uptake into ECs. In BM ECs, HSPC EVs induce up-regulation of C-C motif chemokine receptor 2 (CCR2) ligands that synergize with CXCL12-CXCR4 signaling to promote BM homing. In nonirradiated murine models, marrow preconditioning with HSPC EVs or recombinant CCR2 ligands improves homing and early graft occupancy after transplantation. These findings identify a role for HSPC EVs in remodeling ECs, newly define CCR2-dependent graft homing, and inform novel translational conditioning strategies to improve HSPC transplantation.


Subject(s)
Bone Marrow , Extracellular Vesicles , Hematopoietic Stem Cells , Receptors, CCR2 , Animals , Mice , Bone Marrow/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Cell Movement , Chemokine CXCL12/metabolism , Endothelial Cells/metabolism , Endothelial Cells/cytology , Extracellular Vesicles/metabolism , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Mice, Inbred C57BL , Receptors, CCR2/metabolism , Receptors, CXCR4/metabolism , Signal Transduction , Humans
3.
Immunohorizons ; 8(9): 688-694, 2024 Sep 01.
Article in English | MEDLINE | ID: mdl-39264736

ABSTRACT

Chronic suppurative otitis media (CSOM) is a neglected disease that afflicts 330 million people worldwide and is the most common cause of permanent hearing loss among children in the developing world. Previously, we discovered that outer hair cell (OHC) loss occurred in the basal turn of the cochlea and that macrophages are the major immune cells associated with OHC loss in CSOM. Macrophage-associated cytokines are upregulated. Specifically, CCL-2, an important member of the MCP family, is elevated over time following middle ear infection. CCR2 is a common receptor of the MCP family and the unique receptor of CCL2. CCR2 knockout mice (CCR2-/-) have been used extensively in studies of monocyte activation in neurodegenerative diseases. In the present study, we investigated the effect of CCR2 deletion on the cochlear immune response and OHC survival in CSOM. The OHC survival rate was 84 ± 12.5% in the basal turn of CCR2+/+ CSOM cochleae, compared with was 63 ± 19.9% in the basal turn of CCR2-/- CSOM cochleae (p ≤ 0.05). Macrophage numbers were significantly reduced in CCR2-/- CSOM cochleae compared with CCR2+/+ CSOM cochleae (p ≤ 0.001). In addition, CCL7 was upregulated, whereas IL-33 was downregulated, in CCR2-/- CSOM cochleae. Finally, the permeability of the blood-labyrinth barrier in the stria vascularis remained unchanged in CCR2-/- CSOM compared with CCR2+/+ CSOM. Taken together, the data suggest that CCR2 plays a protective role through cochlear macrophages in the CSOM cochlea.


Subject(s)
Hair Cells, Auditory, Outer , Otitis Media, Suppurative , Receptors, CCR2 , Animals , Female , Male , Mice , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Chronic Disease , Cochlea/metabolism , Cochlea/pathology , Cochlea/immunology , Disease Models, Animal , Hair Cells, Auditory, Outer/metabolism , Hair Cells, Auditory, Outer/pathology , Macrophages/immunology , Macrophages/metabolism , Mice, Inbred C57BL , Mice, Knockout , Otitis Media, Suppurative/immunology , Receptors, CCR2/metabolism , Receptors, CCR2/genetics
4.
Cell Rep Methods ; 4(9): 100846, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39241776

ABSTRACT

Monocytes are critical to innate immunity, participating in chemotaxis during tissue injury, infection, and inflammatory conditions. However, the migration dynamics of human monocytes under different guidance cues are not well characterized. Here, we developed a microfluidic device to profile the migration characteristics of human monocytes under chemotactic and barotactic guidance cues while also assessing the effects of age and cytokine stimulation. Human monocytes preferentially migrated toward the CCL2 gradient through confined microchannels, regardless of donor age and migration pathway. Stimulation with interferon (IFN)-γ, but not granulocyte-macrophage colony-stimulating factor (GM-CSF), disrupted monocyte navigation through complex paths and decreased monocyte CCL2 chemotaxis, velocity, and CCR2 expression. Additionally, monocytes exhibited a bias toward low-hydraulic-resistance pathways in asymmetric environments, which remained consistent across donor ages, cytokine stimulation, and chemoattractants. This microfluidic system provides insights into the unique migratory behaviors of human monocytes and is a valuable tool for studying peripheral immune cell migration in health and disease.


Subject(s)
Cell Movement , Chemotaxis , Monocytes , Humans , Monocytes/immunology , Monocytes/metabolism , Monocytes/drug effects , Cell Movement/drug effects , Chemotaxis/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Chemokine CCL2/metabolism , Interferon-gamma/pharmacology , Interferon-gamma/metabolism , Receptors, CCR2/metabolism , Adult
5.
NPJ Biofilms Microbiomes ; 10(1): 79, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-39227647

ABSTRACT

Alcohol use is an independent risk factor for the development of bacterial pneumonia due, in part, to impaired mucus-facilitated clearance, macrophage phagocytosis, and recruitment of neutrophils. Alcohol consumption is also known to reduce peripheral natural killer (NK) cell numbers and compromise NK cell cytolytic activity, especially NK cells with a mature phenotype. However, the role of innate lymphocytes, such as NK cells during host defense against alcohol-associated bacterial pneumonia is essentially unknown. We have previously shown that indole supplementation mitigates increases in pulmonary bacterial burden and improves pulmonary NK cell recruitment in alcohol-fed mice, which were dependent on aryl hydrocarbon receptor (AhR) signaling. Employing a binge-on-chronic alcohol-feeding model we sought to define the role and interaction of indole and NK cells during pulmonary host defense against alcohol-associated pneumonia. We demonstrate that alcohol dysregulates NK cell effector function and pulmonary recruitment via alterations in two key signaling pathways. We found that alcohol increases transforming growth factor beta (TGF-ß) signaling while suppressing AhR signaling. We further demonstrated that NK cells isolated from alcohol-fed mice have a reduced ability to kill Klebsiella pneumoniae. NK cell migratory capacity to chemokines was also significantly altered by alcohol, as NK cells isolated from alcohol-fed mice exhibited preferential migration in response to CXCR3 chemokines but exhibited reduced migration in response to CCR2, CXCR4, and CX3CR1 chemokines. Together this data suggests that alcohol disrupts NK cell-specific TGF-ß and AhR signaling pathways leading to decreased pulmonary recruitment and cytolytic activity thereby increasing susceptibility to alcohol-associated bacterial pneumonia.


Subject(s)
Killer Cells, Natural , Mice, Inbred C57BL , Pneumonia, Bacterial , Receptors, Aryl Hydrocarbon , Signal Transduction , Animals , Killer Cells, Natural/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Mice , Receptors, Aryl Hydrocarbon/metabolism , Lung/immunology , Lung/microbiology , Transforming Growth Factor beta/metabolism , Ethanol , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Disease Models, Animal , Indoles/pharmacology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Basic Helix-Loop-Helix Transcription Factors/genetics , Male , Klebsiella pneumoniae , Receptors, CXCR3/metabolism
6.
ACS Chem Biol ; 19(9): 2070-2080, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39186040

ABSTRACT

Interest in affinity-based probes (AfBPs) as novel tools to interrogate G protein-coupled receptors (GPCRs) has gained traction in recent years. AfBPs represent an interesting and more versatile alternative to antibodies. In the present study, we report the development and validation of AfBPs that target the intracellular allosteric pocket of CCR2, a GPCR of interest for the development of therapies targeting autoimmune and inflammatory diseases and also cancer. Owing to the two-step labeling process of these CCR2 AfBPs through the incorporation of a click handle, we were successful in applying our most efficient probe in a variety of in vitro experiments and making use of multiple different detection techniques, such as SDS-PAGE and LC/MS-based proteomics. Collectively, this novel probe shows high selectivity, versatility, and applicability. Hence, this is a valuable alternative for CCR2-targeting antibodies and other traditional tool compounds and could aid in target validation and engagement in drug discovery.


Subject(s)
Receptors, CCR2 , Receptors, CCR2/metabolism , Receptors, CCR2/chemistry , Humans , Allosteric Regulation , Allosteric Site , HEK293 Cells , Affinity Labels/chemistry , Molecular Probes/chemistry
7.
Neuroscience ; 557: 51-55, 2024 Oct 04.
Article in English | MEDLINE | ID: mdl-39137869

ABSTRACT

Subarachnoid hemorrhage due to rupture of intracranial aneurysms has a poor outcome, making this disease being the social problem. Inflammation evoked by the increase in intracranial pressure and the clot in the subarachnoid space after the onset of SAH exacerbates neuronal death and vasospasm, resulting in the poor outcome and severe aftereffects. Here, FROUNT mediates CCR2 and CCR5 signaling as an intracellular molecule binding to these chemoattractant receptors which facilitate the migration of inflammatory cells, such as macrophages, in situ to trigger inflammation there. Animal model of subarachnoid hemorrhage was established in rats through intrathecal injection of autologous blood. The effect of the FROUNT inhibitor, disulfiram, on survival rate, neuronal death in hippocampus or vasospasm was then examined. The intrathecal administration of disulfiram significantly suppressed the infiltration of CD68-positive macrophages and myeloperoxidase-positive neutrophils toward the clot in the cistern in situ. In this condition, disulfiram ameliorated the death of animals after the onset of subarachnoid hemorrhage in rats. In addition, disulfiram suppressed both the two major events after subarachnoid hemorrhage, the neuronal death in hippocampus and vasospasm. The pharmacological inhibition of CCR2 and CCR5 signaling by disulfiram could thus be the therapeutic strategy to improve the outcome of subarachnoid hemorrhage.


Subject(s)
Disulfiram , Rats, Sprague-Dawley , Subarachnoid Hemorrhage , Animals , Disulfiram/pharmacology , Subarachnoid Hemorrhage/drug therapy , Subarachnoid Hemorrhage/metabolism , Male , Vasospasm, Intracranial/drug therapy , Vasospasm, Intracranial/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Disease Models, Animal , Hippocampus/drug effects , Hippocampus/metabolism , Receptors, CCR5/metabolism , Macrophages/drug effects , Macrophages/metabolism , Rats , Prognosis , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Cell Death/drug effects , Cell Death/physiology , Antigens, Differentiation, Myelomonocytic/metabolism
8.
Nature ; 633(8028): 174-181, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39198639

ABSTRACT

Sterile inflammation after myocardial infarction is classically credited to myeloid cells interacting with dead cell debris in the infarct zone1,2. Here we show that cardiomyocytes are the dominant initiators of a previously undescribed type I interferon response in the infarct borderzone. Using spatial transcriptomics analysis in mice and humans, we find that myocardial infarction induces colonies of interferon-induced cells (IFNICs) expressing interferon-stimulated genes decorating the borderzone, where cardiomyocytes experience mechanical stress, nuclear rupture and escape of chromosomal DNA. Cardiomyocyte-selective deletion of Irf3 abrogated IFNIC colonies, whereas mice lacking Irf3 in fibroblasts, macrophages, neutrophils or endothelial cells, Ccr2-deficient mice or plasmacytoid-dendritic-cell-depleted mice did not. Interferons blunted the protective matricellular programs and contractile function of borderzone fibroblasts, and increased vulnerability to pathological remodelling. In mice that died after myocardial infarction, IFNIC colonies were immediately adjacent to sites of ventricular rupture, while mice lacking IFNICs were protected from rupture and exhibited improved survival3. Together, these results reveal a pathological borderzone niche characterized by a cardiomyocyte-initiated innate immune response. We suggest that selective inhibition of IRF3 activation in non-immune cells could limit ischaemic cardiomyopathy while avoiding broad immunosuppression.


Subject(s)
Immunity, Innate , Interferon Regulatory Factor-3 , Interferon Type I , Myocardial Infarction , Myocytes, Cardiac , Animals , Mice , Interferon Type I/metabolism , Interferon Type I/immunology , Interferon Regulatory Factor-3/metabolism , Interferon Regulatory Factor-3/deficiency , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Humans , Male , Female , Fibroblasts/metabolism , Macrophages/metabolism , Macrophages/immunology , Receptors, CCR2/metabolism , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Mice, Inbred C57BL , Endothelial Cells/metabolism
9.
Int J Mol Sci ; 25(16)2024 Aug 13.
Article in English | MEDLINE | ID: mdl-39201480

ABSTRACT

Aging is a prominent risk factor for numerous chronic diseases. Understanding the shared mechanisms of aging can aid in pinpointing therapeutic targets for age-related disorders. Chronic inflammation has emerged as a pivotal mediator of aging and a determinant in various age-related chronic conditions. Recent findings indicate that C-C motif chemokine ligand 2 and receptor 2 (CCL2-CCR2) signaling, an important physiological modulator in innate immune response and inflammatory defense, plays a crucial role in aging-related disorders and is increasingly recognized as a promising therapeutic target, highlighting its significance. This review summarizes recent advances in the investigation of CCL2-CCR2 signaling in cardiovascular and neural aging, as well as in various aging-related disorders. It also explores the underlying mechanisms and therapeutic potentials in these contexts. These insights aim to deepen our understanding of aging pathophysiology and the development of aging-related diseases.


Subject(s)
Aging , Cardiovascular Diseases , Chemokine CCL2 , Receptors, CCR2 , Humans , Aging/metabolism , Receptors, CCR2/metabolism , Animals , Cardiovascular Diseases/metabolism , Chemokine CCL2/metabolism , Signal Transduction , Inflammation/metabolism
10.
Int J Mol Sci ; 25(16)2024 Aug 18.
Article in English | MEDLINE | ID: mdl-39201670

ABSTRACT

CC chemokine receptor 2 (CCR2) has been linked to many inflammatory and immune diseases, making it a relevant drug target. Yet, all CCR2 antagonists developed so far have failed in clinical trials; thus, novel strategies are needed to target this receptor. Targeted protein degradation represents a novel approach to inhibit protein function by hijacking the cellular degradation machinery, such as the proteasome, to degrade the protein of interest. Here, we aimed to determine the amenability of CCR2 to chemically induced degradation by using a CCR2 fusion protein containing a HaloTag7 and HiBiT tag (CCR2-HaloTag-HiBiT). After characterization of the CCR2 construct, we used luminescence-based assays and immunofluorescence to quantify CCR2 levels, as well as a label-free, phenotypic assay to investigate the functional effect of CCR2 degradation. Treatment with HaloPROTAC3, which selectively degrades HaloTag fusion proteins, led to concentration- and time-dependent degradation of CCR2-HaloTag-HiBiT. HaloPROTAC3 induced degradation via the proteasome, as degradation was fully blocked with proteasomal inhibitors. Finally, functional assays showed that degradation of CCR2-HaloTag-HiBiT leads to a reduced functional response after agonist stimulation. Overall, our results indicate that CCR2 is amenable to targeted degradation, paving the way for the future development of CCR2 chemical degraders.


Subject(s)
Proteasome Endopeptidase Complex , Proteolysis , Receptors, CCR2 , Receptors, CCR2/metabolism , Humans , Proteolysis/drug effects , Proteasome Endopeptidase Complex/metabolism , HEK293 Cells , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/genetics
11.
Rev Med Virol ; 34(5): e2578, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39192485

ABSTRACT

A wide variety of inflammatory mediators, mainly cytokines and chemokines, are induced during SARS CoV-2 infection. Among these proinflammatory mediators, chemokines tend to play a pivotal role in virus-mediated immunopathology. The C-C chemokine ligand 2 (CCL2), also known as monocyte chemoattractant protein-1 (MCP-1) is a potent proinflammatory cytokine and strong chemoattractant of monocytes, macrophages and CD4+ T cells bearing C-C chemokine receptor type-2 (CCR2). Besides controlling immune cell trafficking, CCL2 is also involved in multiple pathophysiological processes including systemic hyperinflammation associated cytokine release syndrome (CRS), organ fibrosis and blood coagulation. These pathological features are commonly manifested in severe and fatal cases of COVID-19. Given the crucial role of CCL2 in COVID-19 pathogenesis, the CCL2:CCR2 axis may constitute a potential therapeutic target to control virus-induced hyperinflammation and multi-organ dysfunction. Herein we describe recent advances on elucidating the role of CCL2 in COVID-19 pathogenesis, prognosis, and a potential target of anti-inflammatory interventions.


Subject(s)
COVID-19 , Chemokine CCL2 , SARS-CoV-2 , Humans , COVID-19/immunology , COVID-19/virology , COVID-19/pathology , Chemokine CCL2/metabolism , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity , Prognosis , Receptors, CCR2/metabolism , Biomarkers , Anti-Inflammatory Agents/therapeutic use , Cytokine Release Syndrome/immunology , Cytokine Release Syndrome/virology
12.
Front Biosci (Landmark Ed) ; 29(8): 303, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39206918

ABSTRACT

BACKGROUND: Rheumatic heart disease (RHD) is an autoimmune disease caused by recurrent infections of Group A streptococcus (GAS), ultimately leading to inflammation and the fibrosis of heart valves. Recent studies have highlighted the crucial role of C-C chemokine receptor type 2-positive (CCR2+) macrophages in autoimmune diseases and tissue fibrosis. However, the specific involvement of CCR2+ macrophages in RHD remains unclear. METHODS: This study established an RHD rat model using inactivated GAS and complete Freund's adjuvant, demonstrating a correlation between CCR2+ macrophages and fibrosis in the mitral valves of these rats. RESULTS: Intraperitoneal injection of the CCR2 antagonist Rs-504393 significantly reduced macrophage infiltration, inflammation, and fibrosis in valve tissues of RHD rats compared to the solvent-treated group . Existing evidence suggests that C-C motif chemokine ligand 2 (CCL2) acts as the primary recruiting factor for CCR2+ cells. To validate this, human monocytic leukemia cells (THP-1) were cultured in vitro to assess the impact of recombinant CCL2 protein on macrophages. CCL2 exhibited pro-inflammatory effects similar to lipopolysaccharide (LPS), promoting M1 polarization in macrophages. Moreover, the combined effect of LPS and CCL2 was more potent than either alone. Knocking down CCR2 expression in THP-1 cells using small interfering RNA suppressed the pro-inflammatory response and M1 polarization induced by CCL2. CONCLUSIONS: The findings from this study indicate that CCR2+ macrophages are pivotal in the valvular remodeling process of RHD. Targeting the CCL2/CCR2 signaling pathway may therefore represent a promising therapeutic strategy to alleviate valve fibrosis in RHD.


Subject(s)
Inflammation , Macrophages , Receptors, CCR2 , Rheumatic Heart Disease , Animals , Humans , Male , Rats , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Disease Models, Animal , Eicosapentaenoic Acid/analogs & derivatives , Fibrosis , Heart Valves/pathology , Inflammation/metabolism , Macrophages/metabolism , Macrophages/immunology , Rats, Inbred Lew , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Rheumatic Heart Disease/immunology , Rheumatic Heart Disease/microbiology , Rheumatic Heart Disease/metabolism , Rheumatic Heart Disease/pathology , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Streptococcal Infections/metabolism , Streptococcus pyogenes , THP-1 Cells
13.
Dis Model Mech ; 17(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38973385

ABSTRACT

Despite advancements in treatment, approximately 25% of patients with breast cancer experience long-term skeletal muscle wasting (SMW), which limits mobility, reduces drug tolerance and adversely impacts survival. By understanding the underlying molecular mechanisms of SMW, we may be able to develop new strategies to alleviate this condition and improve the lives of patients with breast cancer. Chemokines are small soluble factors that regulate homing of immune cells to tissues during inflammation. In breast cancers, overexpression of C-C chemokine ligand 2 (CCL2) correlates with unfavorable prognosis. Elevated levels of CCL2 in peripheral blood indicate possible systemic effects of this chemokine in patients with breast cancer. Here, we investigated the role of CCL2 signaling on SMW in tumor and non-tumor contexts. In vitro, increasing concentrations of CCL2 inhibited myoblast and myotube function through C-C chemokine receptor 2 (CCR2)-dependent mechanisms involving JNK, SMAD3 and AMPK signaling. In healthy mice, delivery of recombinant CCL2 protein promoted SMW in a dose-dependent manner. In vivo knockdown of breast tumor-derived CCL2 partially protected against SMW. Overall, chronic, upregulated CCL2-CCR2 signaling positively regulates SMW, with implications for therapeutic targeting.


Subject(s)
Breast Neoplasms , Chemokine CCL2 , Muscle, Skeletal , Signal Transduction , Animals , Chemokine CCL2/metabolism , Female , Muscle, Skeletal/pathology , Muscle, Skeletal/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/metabolism , Humans , Receptors, CCR2/metabolism , Mice , Cell Line, Tumor , Disease Models, Animal , Muscular Atrophy/pathology , Muscular Atrophy/metabolism , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/pathology , Muscle Fibers, Skeletal/drug effects , Myoblasts/metabolism , Mice, Inbred C57BL
14.
Cell ; 187(17): 4637-4655.e26, 2024 Aug 22.
Article in English | MEDLINE | ID: mdl-39043180

ABSTRACT

The medical burden of stroke extends beyond the brain injury itself and is largely determined by chronic comorbidities that develop secondarily. We hypothesized that these comorbidities might share a common immunological cause, yet chronic effects post-stroke on systemic immunity are underexplored. Here, we identify myeloid innate immune memory as a cause of remote organ dysfunction after stroke. Single-cell sequencing revealed persistent pro-inflammatory changes in monocytes/macrophages in multiple organs up to 3 months after brain injury, notably in the heart, leading to cardiac fibrosis and dysfunction in both mice and stroke patients. IL-1ß was identified as a key driver of epigenetic changes in innate immune memory. These changes could be transplanted to naive mice, inducing cardiac dysfunction. By neutralizing post-stroke IL-1ß or blocking pro-inflammatory monocyte trafficking with a CCR2/5 inhibitor, we prevented post-stroke cardiac dysfunction. Such immune-targeted therapies could potentially prevent various IL-1ß-mediated comorbidities, offering a framework for secondary prevention immunotherapy.


Subject(s)
Brain Injuries , Immunity, Innate , Immunologic Memory , Inflammation , Interleukin-1beta , Mice, Inbred C57BL , Monocytes , Animals , Mice , Interleukin-1beta/metabolism , Brain Injuries/immunology , Humans , Male , Monocytes/metabolism , Monocytes/immunology , Inflammation/immunology , Macrophages/immunology , Macrophages/metabolism , Stroke/complications , Stroke/immunology , Heart Diseases/immunology , Female , Receptors, CCR2/metabolism , Fibrosis , Epigenesis, Genetic , Trained Immunity
15.
Circ Res ; 135(5): 596-613, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39056179

ABSTRACT

BACKGROUND: Macrophages are key players in obesity-associated cardiovascular diseases, which are marked by inflammatory and immune alterations. However, the pathophysiological mechanisms underlying macrophage's role in obesity-induced cardiac inflammation are incompletely understood. Our study aimed to identify the key macrophage population involved in obesity-induced cardiac dysfunction and investigate the molecular mechanism that contributes to the inflammatory response. METHODS: In this study, we used single-cell RNA-sequencing analysis of Cd45+CD11b+F4/80+ cardiac macrophages to explore the heterogeneity of cardiac macrophages. The CCR2+ (C-C chemokine receptor 2) macrophages were specifically removed by a dual recombinase approach, and the macrophage CCR2 was deleted to investigate their functions. We also performed cleavage under target and tagmentation analysis, chromatin immunoprecipitation-polymerase chain reaction, luciferase assay, and macrophage-specific lentivirus transfection to define the impact of lysozyme C in macrophages on obesity-induced inflammation. RESULTS: We find that the Ccr2 cluster undergoes a functional transition from homeostatic maintenance to proinflammation. Our data highlight specific changes in macrophage behavior during cardiac dysfunction under metabolic challenge. Consistently, inducible ablation of CCR2+CX3CR1+ macrophages or selective deletion of macrophage CCR2 prevents obesity-induced cardiac dysfunction. At the mechanistic level, we demonstrate that the obesity-induced functional shift of CCR2-expressing macrophages is mediated by the CCR2/activating transcription factor 3/lysozyme 1/NF-κB (nuclear factor kappa B) signaling. Finally, we uncover a noncanonical role for lysozyme 1 as a transcription activator, binding to the RelA promoter, driving NF-κB signaling, and strongly promoting inflammation and cardiac dysfunction in obesity. CONCLUSIONS: Our findings suggest that lysozyme 1 may represent a potential target for the diagnosis of obesity-induced inflammation and the treatment of obesity-induced heart disease.


Subject(s)
Macrophages , Muramidase , Obesity , Receptors, CCR2 , Animals , Obesity/complications , Obesity/metabolism , Macrophages/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Mice , Muramidase/metabolism , Muramidase/genetics , Mice, Inbred C57BL , Male , Mice, Knockout , Signal Transduction , Inflammation/metabolism , Inflammation/genetics , Heart Diseases/etiology , Heart Diseases/metabolism , Heart Diseases/genetics
16.
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
17.
J Pathol ; 264(2): 174-185, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39056146

ABSTRACT

The CCL2-CCR2 axis is involved in lupus nephritis, however the precise roles in the mechanisms by which different pathological lesions develop after glomerular immune complex deposition remain elusive. Previously, we demonstrated that genetic CCR2 inhibition induced a histological switch from glomerular endocapillary hypercellularity to wire-loop lesions in murine lupus nephritis. This study aimed to clarify the CCL2-CCR2 axis-mediated cellular mechanism in the formation of these different pathological lesions. We injected MRL/lpr mouse-derived monoclonal IgG3 antibody-producing hybridomas, 2B11.3 or B1, into wild-type (WT) mice to selectively induce glomerular endocapillary hypercellularity or wire-loop lesions. The expression of chemokine and chemokine receptors was analyzed using RT-quantitative PCR and/or immunofluorescence. We found 2B11.3 caused glomerular endocapillary hypercellularity in WT mice with glomerular infiltration of larger numbers of CCR2-expressing macrophages and neutrophils phagocyting immune complex, whereas B1 induced wire-loop lesions. In glomerular endocapillary hypercellularity, CCL2 was identified as the ligand involved in the CCR2-positive cell infiltration; it was expressed by glomerular endothelial cells and macrophages. Notably, 2B11.3-induced glomerular endocapillary hypercellularity converted to wire-loop lesions with reduced glomerular macrophage and neutrophil infiltration in CCL2-deficient (Ccl2-/-) mice similarly observed in Ccr2-/- mice. Moreover, this histological conversion was also observed when both glomerular macrophage and neutrophil infiltration were inhibited in anti-Ly6G antibody-treated Ccr5-/- mice but not when only glomerular macrophage infiltration was inhibited in Ccr5-/- mice or when only glomerular neutrophil infiltration was inhibited in anti-Ly6G antibody-treated WT mice. In contrast, B1 injection caused wire-loop lesions in Ccl2-/- and Ccr2-/- mice, as observed in WT mice. Moreover, 2B11.3 induced CCL2 from glomerular endothelial cells to a larger extent than B1 when injected into Ccr2-/- mice. In conclusion, the CCL2-CCR2 axis determines whether glomerular endocapillary hypercellularity or wire-loop lesions develop by regulating glomerular infiltration of phagocytic cells: macrophages and neutrophils. © 2024 The Pathological Society of Great Britain and Ireland.


Subject(s)
Chemokine CCL2 , Kidney Glomerulus , Lupus Nephritis , Macrophages , Receptors, CCR2 , Animals , Lupus Nephritis/pathology , Lupus Nephritis/immunology , Lupus Nephritis/metabolism , Chemokine CCL2/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Kidney Glomerulus/pathology , Kidney Glomerulus/metabolism , Kidney Glomerulus/immunology , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Neutrophil Infiltration , Mice, Inbred MRL lpr , Neutrophils/immunology , Neutrophils/metabolism , Neutrophils/pathology , Female , Disease Models, Animal , Mice , Mice, Inbred C57BL , Signal Transduction
18.
Cell Commun Signal ; 22(1): 364, 2024 Jul 17.
Article in English | MEDLINE | ID: mdl-39014433

ABSTRACT

BACKGROUND: Chronic obstructive pulmonary disease (COPD) remains a leading cause of morbidity and mortality worldwide, characterized by persistent respiratory symptoms and airflow limitation. The involvement of C-C motif chemokine ligand 2 (CCL2) in COPD pathogenesis, particularly in macrophage regulation and activation, is poorly understood despite its recognized role in chronic inflammation. Our study aims to elucidate the regulatory role and molecular mechanisms of CCL2 in the pathogenesis of COPD, providing new insights for therapeutic strategies. METHODS: This study focused on the CCL2-CCR2 signaling pathway, exploring its role in COPD pathogenesis using both Ccl2 knockout (KO) mice and pharmacological inhibitors. To dissect the underlying mechanisms, we employed various in vitro and in vivo methods to analyze the secretion patterns and pathogenic effects of CCL2 and its downstream molecular signaling through the CCL2-CCR2 axis. RESULTS: Elevated Ccl2 expression was confirmed in the lungs of COPD mice and was associated with enhanced recruitment and activation of macrophages. Deletion of Ccl2 in knockout mice, as well as treatment with a Ccr2 inhibitor, resulted in protection against CS- and LPS-induced alveolar injury and airway remodeling. Mechanistically, CCL2 was predominantly secreted by bronchial epithelial cells in a process dependent on STAT1 phosphorylation and acted through the CCR2 receptor on macrophages. This interaction activated the PI3K-AKT signaling pathway, which was pivotal for macrophage activation and the secretion of inflammatory cytokines, further influencing the progression of COPD. CONCLUSIONS: The study highlighted the crucial role of CCL2 in mediating inflammatory responses and remodeling in COPD. It enhanced our understanding of COPD's molecular mechanisms, particularly how CCL2's interaction with the CCR2 activates critical signaling pathways. Targeting the CCL2-CCR2 axis emerged as a promising strategy to alleviate COPD pathology.


Subject(s)
Chemokine CCL2 , Macrophages , Mice, Knockout , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Pulmonary Disease, Chronic Obstructive , Receptors, CCR2 , Signal Transduction , Pulmonary Disease, Chronic Obstructive/metabolism , Pulmonary Disease, Chronic Obstructive/pathology , Animals , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Macrophages/metabolism , Macrophages/drug effects , Mice , Humans , Mice, Inbred C57BL , Male
20.
Front Immunol ; 15: 1387651, 2024.
Article in English | MEDLINE | ID: mdl-39076996

ABSTRACT

Osteoarthritis (OA) is characterized by a complex interplay of molecular signals orchestrated by the CCL2/CCR2 axis. The pathogenesis of OA has been revealed to be influenced by a multifaceted effect of CCL2/CCR2 signaling on inflammation, cartilage degradation, and joint homeostasis. The CCL2/CCR2 axis promotes immune cell recruitment and tips the balance toward degeneration by influencing chondrocyte behavior. Insights into these intricate pathways will offer novel therapeutic approaches, paving the way for targeted interventions that may redefine OA management in the future. This review article explores the molecular symphony through the lens of the CCL2/CCR2 axis, providing a harmonious blend of current knowledge and future directions on OA treatment. Furthermore, in this study, through a meticulous review of recent research, the key players and molecular mechanisms that amplify the catabolic cascade within the joint microenvironment are identified, and therapeutic approaches to targeting the CCL2/CCR axis are discussed.


Subject(s)
Chemokine CCL2 , Osteoarthritis , Receptors, CCR2 , Signal Transduction , Humans , Chemokine CCL2/metabolism , Receptors, CCR2/metabolism , Osteoarthritis/metabolism , Osteoarthritis/immunology , Osteoarthritis/therapy , Animals , Chondrocytes/metabolism , Chondrocytes/immunology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cartilage, Articular/immunology , Molecular Targeted Therapy
SELECTION OF CITATIONS
SEARCH DETAIL