Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.120
Filter
2.
Int J Mol Sci ; 25(13)2024 Jul 05.
Article in English | MEDLINE | ID: mdl-39000516

ABSTRACT

The ligands of chemokine receptors 2 and 5 (CCR2 and CCR5, respectively) are associated with the pathomechanism of neuropathic pain development, but their role in painful diabetic neuropathy remains unclear. Therefore, the aim of our study was to examine the function of these factors in the hypersensitivity accompanying diabetes. Additionally, we analyzed the analgesic effect of cenicriviroc (CVC), a dual CCR2/CCR5 antagonist, and its influence on the effectiveness of morphine. An increasing number of experimental studies have shown that targeting more than one molecular target is advantageous compared with the coadministration of individual pharmacophores in terms of their analgesic effect. The advantage of using bifunctional compounds is that they gain simultaneous access to two receptors at the same dose, positively affecting their pharmacokinetics and pharmacodynamics and consequently leading to improved analgesia. Experiments were performed on male and female Swiss albino mice with a streptozotocin (STZ, 200 mg/kg, i.p.) model of diabetic neuropathy. We found that the blood glucose level increased, and the mechanical and thermal hypersensitivity developed on the 7th day after STZ administration. In male mice, we observed increased mRNA levels of Ccl2, Ccl5, and Ccl7, while in female mice, we observed additional increases in Ccl8 and Ccl12 levels. We have demonstrated for the first time that a single administration of cenicriviroc relieves pain to a similar extent in male and female mice. Moreover, repeated coadministration of cenicriviroc with morphine delays the development of opioid tolerance, while the best and longest-lasting analgesic effect is achieved by repeated administration of cenicriviroc alone, which reduces pain hypersensitivity in STZ-exposed mice, and unlike morphine, no tolerance to the analgesic effects of CVC is observed until Day 15 of treatment. Based on these results, we suggest that targeting CCR2 and CCR5 with CVC is a potent therapeutic option for novel pain treatments in diabetic neuropathy patients.


Subject(s)
CCR5 Receptor Antagonists , Diabetic Neuropathies , Disease Models, Animal , Receptors, CCR2 , Receptors, CCR5 , Animals , Mice , Diabetic Neuropathies/drug therapy , Male , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Female , Receptors, CCR5/metabolism , Receptors, CCR5/genetics , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Morphine/pharmacology , Morphine/therapeutic use , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/complications , Analgesics/pharmacology , Analgesics/therapeutic use , Hyperalgesia/drug therapy , Imidazoles , Sulfoxides
3.
Sci Rep ; 14(1): 15736, 2024 07 08.
Article in English | MEDLINE | ID: mdl-38977751

ABSTRACT

Benign Prostatic Hyperplasia (BPH) is a complex condition leading to Lower Urinary Tract Symptoms in aging men, characterized by cellular proliferation, smooth muscle dysfunction, inflammation, and fibrosis. While BPH is known to involve heightened macrophage infiltration, the specific contribution of infiltrating monocytes/macrophages to the disease mechanism remains uncertain. This research explores the impact of reducing circulating monocytes and subsequently limiting their tissue infiltration by using Ccr2 knockout (Ccr2-KO) mice. Ccr2-KO and wild type mice were implanted with testosterone and estradiol (T + E2, 25 mg + 2.5 mg) pellets. Urinary function was assessed via weekly void spot assays over 12 weeks, and prostatic macrophage levels were visualized and quantified in tissue sections using an F4/80 antibody. Additionally, Ki-67 staining was used to evaluate cell proliferation, and picrosirius red staining to assess collagen accumulation. Increased voiding frequency which developed in T + E2 mice, was significantly ameliorated in Ccr2-KO mice, however, both Ccr2-KO and wild type (WT) mice showed increased bladder weights after three month, representing a hypertrophic response to bladder outlet obstruction. T + E2 substantially increased the density of macrophages in WT but not Ccr2-KO mouse prostate. Proliferation rate, as indicated by Ki-67 positivity, was elevated in the vental and anterior prostate lobes but was only marginally reduced in Ccr2-KO mice. Most importantly, a significant prostatic collagen accumulation was observed in WT mice that was markedly reduced by Ccr2 deficiency post T + E2 treatment. The absence of Ccr2 mitigates urinary dysfunction and alters prostatic macrophage levels and collagen accumulation in steroid hormone imbalance. These findings suggest a crucial role for monocyte infiltration, giving rise to macrophages or other cell derivatives, to drive fibrosis.


Subject(s)
Estradiol , Fibrosis , Macrophages , Mice, Knockout , Monocytes , Prostate , Receptors, CCR2 , Testosterone , Animals , Male , Receptors, CCR2/metabolism , Macrophages/metabolism , Mice , Monocytes/metabolism , Prostate/metabolism , Prostate/pathology , Testosterone/metabolism , Estradiol/metabolism , Estradiol/pharmacology , Prostatic Hyperplasia/metabolism , Prostatic Hyperplasia/pathology , Cell Proliferation , Mice, Inbred C57BL
4.
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
5.
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
6.
Pharmacol Res ; 205: 107242, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38823470

ABSTRACT

Targeting the CCL2/CCR2 chemokine axis has been shown to be effective at relieving pain in rodent models of inflammatory and neuropathic pain, therefore representing a promising avenue for the development of non-opioid analgesics. However, clinical trials targeting this receptor for inflammatory conditions and painful neuropathies have failed to meet expectations and have all been discontinued due to lack of efficacy. To overcome the poor selectivity of CCR2 chemokine receptor antagonists, we generated and characterized the function of intracellular cell-penetrating allosteric modulators targeting CCR2, namely pepducins. In vivo, chronic intrathecal administration of the CCR2-selective pepducin PP101 was effective in alleviating neuropathic and bone cancer pain. In the setting of bone metastases, we found that T cells infiltrate dorsal root ganglia (DRG) and induce long-lasting pain hypersensitivity. By acting on CCR2-expressing DRG neurons, PP101 attenuated the altered phenotype of sensory neurons as well as the neuroinflammatory milieu of DRGs, and reduced bone cancer pain by blocking CD4+ and CD8+ T cell infiltration. Notably, PP101 demonstrated its efficacy in targeting the neuropathic component of bone cancer pain, as evidenced by its anti-nociceptive effects in a model of chronic constriction injury of the sciatic nerve. Importantly, PP101-induced reduction of CCR2 signaling in DRGs did not result in deleterious tumor progression or adverse behavioral effects. Thus, targeting neuroimmune crosstalk through allosteric inhibition of CCR2 could represent an effective and safe avenue for the management of chronic pain.


Subject(s)
Chronic Pain , Ganglia, Spinal , Neuralgia , Receptors, CCR2 , Animals , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Chronic Pain/drug therapy , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Neuralgia/drug therapy , Neuralgia/metabolism , Humans , Cancer Pain/drug therapy , Bone Neoplasms/drug therapy , Bone Neoplasms/secondary , Analgesics/pharmacology , Analgesics/therapeutic use , Male , Mice , Female , Mice, Inbred C57BL
7.
J Immunol ; 213(2): 214-225, 2024 07 15.
Article in English | MEDLINE | ID: mdl-38829123

ABSTRACT

The interactions between chemokines and their receptors, particularly in the context of inflammation, are complex, with individual receptors binding multiple ligands and individual ligands interacting with multiple receptors. In addition, there are numerous reports of simultaneous coexpression of multiple inflammatory chemokine receptors on individual inflammatory leukocyte subtypes. Overall, this has previously been interpreted as redundancy and proposed as a protective mechanism to ensure that the inflammatory response is robust. By contrast, we have hypothesized that the system is not redundant but exquisitely subtle. Our interests relate to the receptors CCR1, CCR2, CCR3, and CCR5, which, together, regulate nonneutrophilic myeloid cell recruitment to inflammatory sites. In this study, we demonstrate that although most murine monocytes exclusively express CCR2, there is a small subpopulation that is expanded during inflammation and coexpresses CCR1 and CCR2. Combinations of transcript and functional analysis demonstrate that this is not redundant expression and that coexpression of CCR1 and CCR2 marks a phenotypically distinct population of monocytes characterized by expression of genes otherwise typically associated with neutrophils. Single-cell RNA sequencing confirms this as a monodisperse population of atypical monocytes. This monocytic population has previously been described as having immunosuppressive activity. Overall, our data confirm combinatorial chemokine receptor expression by a subpopulation of monocytes but demonstrate that this is not redundant expression and marks a discrete monocytic population.


Subject(s)
Monocytes , Receptors, CCR1 , Receptors, CCR2 , Receptors, CCR1/genetics , Receptors, CCR1/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Monocytes/immunology , Monocytes/metabolism , Animals , Mice , Mice, Inbred C57BL , Inflammation/immunology
8.
Endocrinology ; 165(7)2024 May 27.
Article in English | MEDLINE | ID: mdl-38862137

ABSTRACT

The inhibition of hepatic macrophage and Kupfer cell recruitment and activation is a potential strategy for treating insulin resistance and nonalcoholic steatohepatitis (NASH). Cenicriviroc (CVC), a dual C-C chemokine receptor 2 (CCR2) and CCR5 antagonist, has shown antifibrotic activity in murine models of NASH and has been evaluated in clinical trials on patients with NASH. This study investigated the effects of CVC on macrophage infiltration and polarization in a lipotoxic model of NASH. C57BL/6 mice were fed a high-cholesterol, high-fat (CL) diet or a CL diet containing 0.015% CVC (CL + CVC) for 12 weeks. Macrophage recruitment and activation were assayed by immunohistochemistry and flow cytometry. CVC supplementation attenuated excessive hepatic lipid accumulation and peroxidation and alleviated glucose intolerance and hyperinsulinemia in the mice that were fed the CL diet. Flow cytometry analysis revealed that compared with the CL group, mice fed the CL + CVC diet had fewer M1-like macrophages, more M2-like macrophages, and fewer T cell counts, indicating that CVC caused an M2-dominant shift of macrophages in the liver. Similarly, CVC decreased lipopolysaccharide-stimulated M1-like macrophage activation, whereas it increased interleukin-4-induced M2-type macrophage polarization in vitro. In addition, CVC attenuated hepatic fibrosis by repressing hepatic stellate cell activation. Lastly, CVC reversed insulin resistance as well as steatosis, inflammation, and fibrosis of the liver in mice with pre-existing NASH. In conclusion, CVC prevented and reversed hepatic steatosis, insulin resistance, inflammation, and fibrogenesis in the liver of NASH mice via M2 macrophage polarization.


Subject(s)
Liver , Macrophages , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease , Animals , Macrophages/drug effects , Macrophages/metabolism , Mice , Non-alcoholic Fatty Liver Disease/drug therapy , Non-alcoholic Fatty Liver Disease/pathology , Male , Liver/drug effects , Liver/pathology , Liver/metabolism , Diet, High-Fat/adverse effects , Receptors, CCR2/metabolism , Sulfoxides/pharmacology , Macrophage Activation/drug effects , CCR5 Receptor Antagonists/pharmacology , CCR5 Receptor Antagonists/therapeutic use , Insulin Resistance , Imidazoles
9.
Int Immunopharmacol ; 137: 112528, 2024 Aug 20.
Article in English | MEDLINE | ID: mdl-38908086

ABSTRACT

Low back pain due to epidural fibrosis is a major complication after spine surgery. Macrophages infiltrate the wound area post laminectomy, but the role of macrophages in epidural fibrosis remains largely elusive. In a mouse model of laminectomy, macrophage depletion decreased epidural fibrosis. CD146, an adhesion molecule involved in cell migration, is expressed by macrophages. CD146-defective macrophages exhibited impaired migration, which was mediated by reduced expression of CCR2 and suppression of the MAPK/ERK signaling pathway. CD146-defective macrophages suppress the MAPK/ERK signaling pathway by increasing Erdr1. In vivo, CD146 deficiency decreased macrophage infiltration and reduced extracellular matrix deposition in wound tissues. Moreover, the anti-CD146 antibody AA98 suppressed macrophage infiltration and epidural fibrosis. Taken together, these findings demonstrated that CD146 deficiency alleviates epidural fibrosis by decreasing the migration of macrophages via the Erdr1/ERK/CCR2 pathway. Blocking CD146 and macrophage infiltration may help alleviate epidural fibrosis.


Subject(s)
CD146 Antigen , Fibrosis , Macrophages , Mice, Inbred C57BL , Receptors, CCR2 , Animals , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Macrophages/immunology , Macrophages/metabolism , Mice , CD146 Antigen/metabolism , CD146 Antigen/genetics , Cell Movement , Mice, Knockout , Epidural Space/pathology , Male , MAP Kinase Signaling System/immunology , Laminectomy , Disease Models, Animal , Signal Transduction , Humans
10.
Neurosci Lett ; 836: 137879, 2024 Jul 27.
Article in English | MEDLINE | ID: mdl-38880353

ABSTRACT

Peripheral nerves exhibit long-term residual motor dysfunction following injury. The length of the denervation period before nerve and muscle reconnection is an important factor in motor function recovery. We aimed to investigate whether repeated nerve crush injuries to the same site every 7 days would preserve the conditioning lesion (CL) response and to determine the number of nerve crush injuries required to create an experimental animal model that would prolong the denervation period while maintaining peripheral nerve continuity. Rats were grouped according to the number of sciatic nerve crushes. A significant decrease in the soleus muscle fiber cross-sectional area was observed with increased crushes. After a single crush, macrophage accumulation and macrophage chemotaxis factor CCL2 expression in dorsal root ganglia were markedly increased, which aligned with the gene expression of Ccl2 and its receptor Ccr2. Macrophage numbers, histological CCL2 expression, and Ccl2 and Ccr2 gene expression levels decreased, depending on the number of repeated crushes. Histological analysis and gene expression analysis in the group with four repeated crushes did not differ significantly when compared with uninjured animals. Our findings indicated that repeated nerve crushes at the same site every 7 days sustained innervation loss and caused a loss of the CL response. The experimental model did not require nerve stump suturing and is useful for exploring factors causing prolonged denervation-induced motor dysfunction. SIGNIFICANCE STATEMENT: This study elucidates the effects of repeated nerve crush injury to the same site on innervation and conditioning lesion responses and demonstrates the utility of an experimental animal model that recapitulates the persistent residual motor deficits owing to prolonged denervation without requiring nerve transection and transection suturing.


Subject(s)
Chemokine CCL2 , Disease Models, Animal , Nerve Crush , Sciatic Nerve , Animals , Sciatic Nerve/injuries , Male , Nerve Crush/methods , Chemokine CCL2/metabolism , Chemokine CCL2/genetics , Muscle, Skeletal/innervation , Muscle, Skeletal/metabolism , Ganglia, Spinal/metabolism , Rats , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Macrophages/metabolism , Peripheral Nerve Injuries/metabolism , Peripheral Nerve Injuries/physiopathology , Rats, Sprague-Dawley , Denervation/methods , Nerve Regeneration/physiology , Sciatic Neuropathy/pathology , Sciatic Neuropathy/physiopathology
11.
Clin Transl Sci ; 17(6): e13811, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38814167

ABSTRACT

Immune checkpoint inhibitors remained the standard-of-care treatment for advanced non-small cell lung cancer (NSCLC) for the past decade. In unselected patients, anti-PD-(L)1 monotherapy achieved an overall response rate of about 20%. In this analysis, we developed a pharmacokinetic and pharmacodynamic module for our previously calibrated quantitative systems pharmacology model (QSP) to simulate the effectiveness of macrophage-targeted therapies in combination with PD-L1 inhibition in advanced NSCLC. By conducting in silico clinical trials, the model confirmed that anti-CD47 treatment is not an optimal option of second- and later-line treatment for advanced NSCLC resistant to PD-(L)1 blockade. Furthermore, the model predicted that inhibition of macrophage recruitment, such as using CCR2 inhibitors, can potentially improve tumor size reduction when combined with anti-PD-(L)1 therapy, especially in patients who are likely to respond to anti-PD-(L)1 monotherapy and those with a high level of tumor-associated macrophages. Here, we demonstrate the application of the QSP platform on predicting the effectiveness of novel drug combinations involving immune checkpoint inhibitors based on preclinical or early-stage clinical trial data.


Subject(s)
B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/immunology , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Lung Neoplasms/immunology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/administration & dosage , Immune Checkpoint Inhibitors/pharmacokinetics , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , CD47 Antigen/antagonists & inhibitors , CD47 Antigen/metabolism , Macrophages/metabolism , Macrophages/drug effects , Macrophages/immunology , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Network Pharmacology/methods , Computer Simulation , Models, Biological , Tumor-Associated Macrophages/drug effects , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism
12.
Int Immunopharmacol ; 135: 112328, 2024 Jun 30.
Article in English | MEDLINE | ID: mdl-38796962

ABSTRACT

Alzheimer's disease (AD), as a neurodegenerative disorder, distresses the elderly in large numbers and is characterized by ß-amyloid (Aß) accumulation, elevated tau protein levels, and chronic inflammation. The brain's immune system is aided by microglia and astrocytes, which produce chemokines and cytokines. Nevertheless, dysregulated expression can cause hyperinflammation and lead to neurodegeneration. CCL2/CCR2 chemokines are implicated in neurodegenerative diseases exacerbating. Inflicting damage on nerves and central nervous system (CNS) cells is the function of this axis, which recruits and migrates immune cells, including monocytes and macrophages. It has been shown that targeting the CCL2/CCR2 axis may be a therapeutic option for inflammatory diseases. Using the current knowledge about the involvement of the CCL2/CCR2 axis in the immunopathogenesis of AD, this comprehensive review synthesizes existing information. It also explores potential therapeutic options, including modulation of the CCL2/CCR2 axis as a possible strategy in AD.


Subject(s)
Alzheimer Disease , Chemokine CCL2 , Receptors, CCR2 , Alzheimer Disease/metabolism , Alzheimer Disease/drug therapy , Alzheimer Disease/immunology , Humans , Receptors, CCR2/metabolism , Chemokine CCL2/metabolism , Animals , Amyloid beta-Peptides/metabolism , Amyloid beta-Peptides/immunology , Brain/metabolism , Brain/immunology
13.
Front Immunol ; 15: 1360700, 2024.
Article in English | MEDLINE | ID: mdl-38736886

ABSTRACT

Introduction: Myocardial infarction (MI) is a significant contributor to morbidity and mortality worldwide. Many individuals who survive the acute event continue to experience heart failure (HF), with inflammatory and healing processes post-MI playing a pivotal role. Polymorphonuclear neutrophils (PMN) and monocytes infiltrate the infarcted area, where PMN release high amounts of the heme enzyme myeloperoxidase (MPO). MPO has numerous inflammatory properties and MPO plasma levels are correlated with prognosis and severity of MI. While studies have focused on MPO inhibition and controlling PMN infiltration into the infarcted tissue, less is known on MPO's role in monocyte function. Methods and results: Here, we combined human data with mouse and cell studies to examine the role of MPO on monocyte activation and migration. We revealed a correlation between plasma MPO levels and monocyte activation in a patient study. Using a mouse model of MI, we demonstrated that MPO deficiency led to an increase in splenic monocytes and a decrease in cardiac monocytes compared to wildtype mice (WT). In vitro studies further showed that MPO induces monocyte migration, with upregulation of the chemokine receptor CCR2 and upregulation of inflammatory pathways identified as underlying mechanisms. Conclusion: Taken together, we identify MPO as a pro-inflammatory mediator of splenic monocyte recruitment and activation post-MI and provide mechanistic insight for novel therapeutic strategies after ischemic injury.


Subject(s)
Monocytes , Myocardial Infarction , Peroxidase , Animals , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Myocardial Infarction/metabolism , Peroxidase/metabolism , Monocytes/immunology , Monocytes/metabolism , Humans , Mice , Male , Cell Movement , Disease Models, Animal , Mice, Inbred C57BL , Female , Neutrophils/immunology , Neutrophils/metabolism , Mice, Knockout , Receptors, CCR2/metabolism , Middle Aged
14.
Phytomedicine ; 129: 155694, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38733904

ABSTRACT

BACKGROUND: Ulcerative colitis (UC) is associated with intestinal macrophage infiltration due to disruption of the mucosal barrier and bacterial invasion. Therefore, it is crucial to identify therapeutic agents capable of attenuating the macrophage-induced inflammatory response to preserve mucosal homeostasis and immune tolerance. The modified Zhenwu decoction (CDD-2103) is a novel herbal formulation developed based on the principles of Traditional Chinese medicine. To date, there are no clinically approved herbal formulations for UC with a well-known mechanism of action on macrophages. PURPOSE: The objective of this study was to systematically investigate the inhibitory effect of the active fraction of CDD-2103 in a mouse model of chronic colitis and delineate the mechanisms underlying its inhibitory action. METHODS: CDD-2103 was extracted into four fractions using organic solvents with increasing polarity. A chronic 49-day dextran sulfate sodium (DSS)-induced colitis mice model, closely resembling human clinical conditions, was used to examine the effect of CDD-2103 on chronic colitis. To confirm the effect of CDD-2103 on macrophages in this chronic colitis model, adoptive macrophage transfer and CCL2 supplementation were conducted. The mechanisms of action of CDD-2103 were further elucidated utilizing bone marrow-derived macrophages (BMDMs). Transcriptome analysis was conducted to gain insights into the underlying mechanism of action of CDD-2103 in BMDMs. RESULTS: Our in vitro and in vivo findings demonstrated that the ethanol-enriched fraction of CDD-2103 exhibited significant anti-inflammatory effects, leading to the suppression of colitis severity. This effect was associated with diminished accumulation of colonic macrophages in the lamina propria of CDD-2103-intervened colitis mice. Specifically, CDD-2103 inhibited CCR2/L2-mediated proinflammatory macrophage infiltration into the colon without affecting macrophage proliferation. Mechanistically, CDD-2103 inhibited Fyn expression-mediated p38 MAPK activation and subsequently suppressed CCR2 expression in BMDMs. CONCLUSIONS: Collectively, our study supports the potential use of CDD-2103 to limit macrophage infiltration, thereby reducing inflammation during UC treatment. CDD-2103 and the components in the ethanolic fraction are promising candidates for the development of novel drugs for UC management. Additionally, our study underscores Fyn-mediated CCR2 expression as a potential therapeutic target for the management of UC.


Subject(s)
Dextran Sulfate , Disease Models, Animal , Drugs, Chinese Herbal , Macrophages , Mice, Inbred C57BL , Receptors, CCR2 , p38 Mitogen-Activated Protein Kinases , Animals , Male , Mice , Chronic Disease , Colitis/drug therapy , Colitis/chemically induced , Colitis, Ulcerative/drug therapy , Drugs, Chinese Herbal/pharmacology , Macrophages/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism , Receptors, CCR2/metabolism , Signal Transduction/drug effects
15.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747296

ABSTRACT

Arrhythmogenic cardiomyopathy (ACM) is an inherited cardiac condition characterized by cardiac remodeling and life-threatening ventricular arrhythmias. In this issue of the JCI, Chelko, Penna, and colleagues mechanistically addressed the intricate contribution of immune-mediated injury in ACM pathogenesis. Inhibition of nuclear factor κ-B (NF-κB) and infiltration of monocyte-derived macrophages expressing C-C motif chemokine receptor-2 (CCR2) alleviated the phenotypic ACM features (i.e., fibrofatty replacement, contractile dysfunction, and ventricular arrhythmias) in desmoglein 2-mutant (Dsg2mut/mut) mice. These findings pave the way for efficacious and targetable immune therapy for patients with ACM.


Subject(s)
Desmoglein 2 , Macrophages , Receptors, CCR2 , Animals , Macrophages/metabolism , Macrophages/immunology , Macrophages/pathology , Mice , Humans , Desmoglein 2/genetics , Desmoglein 2/metabolism , Desmoglein 2/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , NF-kappa B/metabolism , NF-kappa B/genetics , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/immunology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/pathology , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Cardiomyopathies/genetics , Cardiomyopathies/pathology , Cardiomyopathies/immunology , Cardiomyopathies/metabolism
16.
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
17.
Immunohorizons ; 8(5): 363-370, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38775688

ABSTRACT

Although the pathogenesis of choroidal neovascularization (CNV) is largely unknown in age-related macular degeneration (AMD), inflammasomes may contribute to CNV development and progression. To understand the role NLRP3 inflammasomes in CNV, we used Ccr2RFPCx3cr1GFP dual-reporter mice and immunostaining techniques to confirm localization of NLRP3 inflammasomes in the laser-induced CNV (LCNV) lesions. Confocal microscopy was used to image and quantify LCNV volumes. MCC950 was used as NLRP3 inhibitor. ELISA and quantitative RT-PCR were used to confirm the activation of NLRP3 by monitoring the expression of IL-1ß protein and mRNA in choroidal tissues from LCNV mice. In addition, NLRP3 (-/-) LCNV mice were used to investigate whether NLRP3 inflammasomes contribute to the development of LCNV lesions. We observed that red fluorescent protein (RFP)-positive monocyte-derived macrophages and GFP-positive microglia-derived macrophages, in addition to other cell types, were localized in LCNV lesions at day 7 post-laser injury. In addition, NLRP3 inflammasomes are associated with LCNV lesions. Inhibition of NLRP3 inflammasomes, using MCC950, caused an increased Ccr2RFP-positive macrophages, Cx3cr1GFP-positive microglia, and other cells, resulting in an increase in total lesion size. NLRP3 (-/-) LCNV mice showed significantly increased lesion size compared with age-matched controls. Inhibition of NLRP3 resulted in decreased IL-1ß mRNA and protein expression in the choroidal tissues, suggesting that increased lesion size may not be directly related to IL-1ß.


Subject(s)
Choroidal Neovascularization , Indenes , Inflammasomes , Interleukin-1beta , Microglia , Monocytes , NLR Family, Pyrin Domain-Containing 3 Protein , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Mice , Inflammasomes/metabolism , Interleukin-1beta/metabolism , Microglia/metabolism , Monocytes/metabolism , Mice, Knockout , Sulfones/pharmacology , Mice, Inbred C57BL , Furans/pharmacology , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Macrophages/metabolism , Macrophages/immunology , Sulfonamides/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Carrier Proteins/metabolism , Carrier Proteins/genetics , Choroid/metabolism , Choroid/pathology , Disease Models, Animal , Lasers/adverse effects , Macular Degeneration/pathology , Macular Degeneration/metabolism , Macular Degeneration/genetics
18.
Front Immunol ; 15: 1372957, 2024.
Article in English | MEDLINE | ID: mdl-38779688

ABSTRACT

Background: Schistosomiasis is a common cause of pulmonary hypertension (PH) worldwide. Type 2 inflammation contributes to the development of Schistosoma-induced PH. Specifically, interstitial macrophages (IMs) derived from monocytes play a pivotal role by producing thrombospondin-1 (TSP-1), which in turn activates TGF-ß, thereby driving the pathology of PH. Resident and recruited IM subpopulations have recently been identified. We hypothesized that in Schistosoma-PH, one IM subpopulation expresses monocyte recruitment factors, whereas recruited monocytes become a separate IM subpopulation that expresses TSP-1. Methods: Mice were intraperitoneally sensitized and then intravenously challenged with S. mansoni eggs. Flow cytometry on lungs and blood was performed on wildtype and reporter mice to identify IM subpopulations and protein expression. Single-cell RNA sequencing (scRNAseq) was performed on flow-sorted IMs from unexposed and at day 1, 3 and 7 following Schistosoma exposure to complement flow cytometry based IM characterization and identify gene expression. Results: Flow cytometry and scRNAseq both identified 3 IM subpopulations, characterized by CCR2, MHCII, and FOLR2 expression. Following Schistosoma exposure, the CCR2+ IM subpopulation expanded, suggestive of circulating monocyte recruitment. Schistosoma exposure caused increased monocyte-recruitment ligand CCL2 expression in the resident FOLR2+ IM subpopulation. In contrast, the vascular pathology-driving protein TSP-1 was greatest in the CCR2+ IM subpopulation. Conclusion: Schistosoma-induced PH involves crosstalk between IM subpopulations, with increased expression of monocyte recruitment ligands by resident FOLR2+ IMs, and the recruitment of CCR2+ IMs which express TSP-1 that activates TGF-ß and causes PH.


Subject(s)
Hypertension, Pulmonary , Macrophages , Animals , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/parasitology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/pathology , Mice , Macrophages/immunology , Macrophages/parasitology , Phenotype , Schistosoma mansoni/immunology , Mice, Inbred C57BL , Schistosomiasis/immunology , Schistosomiasis/complications , Schistosomiasis/parasitology , Disease Models, Animal , Schistosomiasis mansoni/immunology , Schistosomiasis mansoni/parasitology , Schistosomiasis mansoni/complications , Schistosomiasis mansoni/pathology , Thrombospondin 1/genetics , Thrombospondin 1/metabolism , Monocytes/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Female , Schistosoma/immunology , Schistosoma/physiology , Lung/immunology , Lung/parasitology , Lung/pathology
19.
Brain Behav Immun ; 119: 818-835, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38735403

ABSTRACT

Survivors of myocardial infarction are at increased risk for vascular dementia. Neuroinflammation has been implicated in the pathogenesis of vascular dementia, yet little is known about the cellular and molecular mediators of neuroinflammation after myocardial infarction. Using a mouse model of myocardial infarction coupled with flow cytometric analyses and immunohistochemistry, we discovered increased monocyte abundance in the brain after myocardial infarction, which was associated with increases in brain-resident perivascular macrophages and microglia. Myeloid cell recruitment and activation was also observed in post-mortem brains of humans that died after myocardial infarction. Spatial and single cell transcriptomic profiling of brain-resident myeloid cells after experimental myocardial infarction revealed increased expression of monocyte chemoattractant proteins. In parallel, myocardial infarction increased crosstalk between brain-resident myeloid cells and oligodendrocytes, leading to neuroinflammation, white matter injury, and cognitive dysfunction. Inhibition of monocyte recruitment preserved white matter integrity and cognitive function, linking monocytes to neurodegeneration after myocardial infarction. Together, these preclinical and clinical results demonstrate that monocyte infiltration into the brain after myocardial infarction initiate neuropathological events that lead to vascular dementia.


Subject(s)
Brain , Cognitive Dysfunction , Monocytes , Myocardial Infarction , White Matter , Animals , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/complications , White Matter/metabolism , White Matter/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/etiology , Monocytes/metabolism , Mice , Male , Humans , Brain/metabolism , Brain/pathology , Receptors, CCR2/metabolism , Mice, Inbred C57BL , Disease Models, Animal , Macrophages/metabolism , Microglia/metabolism , Neuroinflammatory Diseases/metabolism , Dementia, Vascular/metabolism , Dementia, Vascular/pathology , Oligodendroglia/metabolism
20.
Mol Ther ; 32(7): 2248-2263, 2024 Jul 03.
Article in English | MEDLINE | ID: mdl-38796708

ABSTRACT

Acute kidney injury (AKI) is a major worldwide health concern that currently lacks effective medical treatments. PSMP is a damage-induced chemotactic cytokine that acts as a ligand of CCR2 and has an unknown role in AKI. We have observed a significant increase in PSMP levels in the renal tissue, urine, and plasma of patients with AKI. PSMP deficiency improved kidney function and decreased tubular damage and inflammation in AKI mouse models induced by kidney ischemia-reperfusion injury, glycerol, and cisplatin. Single-cell RNA sequencing analysis revealed that Ly6Chi or F4/80lo infiltrated macrophages (IMs) were a major group of proinflammatory macrophages with strong CCR2 expression in AKI. We observed that PSMP deficiency decreased CCR2+Ly6Chi or F4/80lo IMs and inhibited M1 polarization in the AKI mouse model. Moreover, overexpressed human PSMP in the mouse kidney could reverse the attenuation of kidney injury in a CCR2-dependent manner, and this effect could be achieved without CCL2 involvement. Extracellular PSMP played a crucial role, and treatment with a PSMP-neutralizing antibody significantly reduced kidney injury in vivo. Therefore, PSMP might be a therapeutic target for AKI, and its antibody is a promising therapeutic drug for the treatment of AKI.


Subject(s)
Acute Kidney Injury , Disease Models, Animal , Macrophages , Receptors, CCR2 , Animals , Humans , Male , Mice , Acute Kidney Injury/metabolism , Acute Kidney Injury/pathology , Kidney/metabolism , Kidney/pathology , Macrophages/metabolism , Mice, Knockout , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Reperfusion Injury/metabolism , Neoplasm Proteins
SELECTION OF CITATIONS
SEARCH DETAIL