Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.870
Filter
1.
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
2.
Cells ; 13(10)2024 May 10.
Article in English | MEDLINE | ID: mdl-38786041

ABSTRACT

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


Subject(s)
CX3C Chemokine Receptor 1 , Flow Cytometry , Monocytes , Receptors, CCR2 , Animals , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Monocytes/metabolism , CX3C Chemokine Receptor 1/metabolism , CX3C Chemokine Receptor 1/genetics , Mice , Antibodies/immunology , Genes, Reporter , Phenotype , Mice, Inbred C57BL , Mice, Transgenic , Green Fluorescent Proteins/metabolism , Antigens, Ly/metabolism , Antigens, Ly/genetics
3.
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
4.
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
5.
Int J Mol Sci ; 25(9)2024 May 04.
Article in English | MEDLINE | ID: mdl-38732244

ABSTRACT

Cardiovascular outcome in Marfan syndrome (MFS) patients most prominently depends on aortic aneurysm progression with subsequent aortic dissection. Angiotensin II receptor blockers (ARBs) prevent aneurysm formation in MFS mouse models. In patients, ARBs only slow down aortic dilation. Downstream signalling from the angiotensin II type 1 receptor (AT1R) is mediated by G proteins and ß-arrestin recruitment. AT1R also interacts with the monocyte chemoattractant protein-1 (MCP-1) receptor, resulting in inflammation. In this study, we explore the targeting of ß-arrestin signalling in MFS mice by administering TRV027. Furthermore, because high doses of the ARB losartan, which has been proven beneficial in MFS, cannot be achieved in humans, we investigate a potential additive effect by combining lower concentrations of losartan (25 mg/kg/day and 5 mg/kg/day) with barbadin, a ß-arrestin blocker, and DMX20, a C-C chemokine receptor type 2 (CCR2) blocker. A high dose of losartan (50 mg/kg/day) slowed down aneurysm progression compared to untreated MFS mice (1.73 ± 0.12 vs. 1.96 ± 0.08 mm, p = 0.0033). TRV027, the combination of barbadin with losartan (25 mg/kg/day), and DMX-200 (90 mg/kg/day) with a low dose of losartan (5 mg/kg/day) did not show a significant beneficial effect. Our results confirm that while losartan effectively halts aneurysm formation in Fbn1C1041G/+ MFS mice, neither TRV027 alone nor any of the other compounds combined with lower doses of losartan demonstrate a notable impact on aneurysm advancement. It appears that complete blockade of AT1R function, achieved by administrating a high dosage of losartan, may be necessary for inhibiting aneurysm progression in MFS.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Disease Models, Animal , Losartan , Marfan Syndrome , Receptor, Angiotensin, Type 1 , Signal Transduction , Animals , Marfan Syndrome/metabolism , Marfan Syndrome/drug therapy , Marfan Syndrome/complications , Mice , Losartan/pharmacology , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/drug effects , Angiotensin II Type 1 Receptor Blockers/pharmacology , Aortic Aneurysm/metabolism , Aortic Aneurysm/etiology , Aortic Aneurysm/prevention & control , Aortic Aneurysm/drug therapy , Aortic Aneurysm/pathology , Male , beta-Arrestins/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/antagonists & inhibitors , Mice, Inbred C57BL
6.
Cell Mol Life Sci ; 81(1): 220, 2024 May 20.
Article in English | MEDLINE | ID: mdl-38763956

ABSTRACT

Cardiovascular diseases are an array of age-related disorders, and accumulating evidence suggests a link between cardiac resident macrophages (CRMs) and the age-related disorders. However, how does CRMs alter with aging remains elusive. In the present study, aged mice (20 months old) have been employed to check for their cardiac structural and functional alterations, and the changes in the proportion of CRM subsets as well, followed by sorting of CRMs, including C-C Motif Chemokine Receptor 2 (CCR2)+ and CCR2- CRMs, which were subjected to Smart-Seq. Integrated analysis of the Smart-Seq data with three publicly available single-cell RNA-seq datasets revealed that inflammatory genes were drastic upregulated for both CCR2+ and CCR2- CRMs with aging, but genes germane to wound healing were downregulated for CCR2- CRMs, suggesting the differential functions of these two subsets. More importantly, inflammatory genes involved in damage sensing, complement cascades, and phagocytosis were largely upregulated in CCR2- CRMs, implying the imbalance of inflammatory response upon aging. Our work provides a comprehensive framework and transcriptional resource for assessing the impact of aging on CRMs with a potential for further understanding cardiac aging.


Subject(s)
Aging , Gene Expression Profiling , Macrophages , Mice, Inbred C57BL , Receptors, CCR2 , Animals , Macrophages/metabolism , Aging/genetics , Aging/metabolism , Mice , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Transcriptome , Myocardium/metabolism , Male , Single-Cell Analysis , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Signal Transduction , Phagocytosis
7.
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
8.
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 , Drugs, Chinese Herbal/pharmacology , Macrophages/drug effects , Receptors, CCR2/metabolism , Mice , p38 Mitogen-Activated Protein Kinases/metabolism , Male , Colitis/drug therapy , Colitis/chemically induced , Colitis, Ulcerative/drug therapy , Signal Transduction/drug effects , Chronic Disease
9.
BMC Pulm Med ; 24(1): 242, 2024 May 16.
Article in English | MEDLINE | ID: mdl-38755605

ABSTRACT

INTRODUCTION: Lung cancer is a common malignant tumor, and different types of immune cells may have different effects on the occurrence and development of lung cancer subtypes, including lung squamous cell carcinoma (LUSC) and lung adenocarcinoma (LUAD). However, the causal relationship between immune phenotype and lung cancer is still unclear. METHODS: This study utilized a comprehensive dataset containing 731 immune phenotypes from the European Bioinformatics Institute (EBI) to evaluate the potential causal relationship between immune phenotypes and LUSC and LUAD using the inverse variance weighted (IVW) method in Mendelian randomization (MR). Sensitivity analyses, including MR-Egger intercept, Cochran Q test, and others, were conducted for the robustness of the results. The study results were further validated through meta-analysis using data from the Transdisciplinary Research Into Cancer of the Lung (TRICL) data. Additionally, confounding factors were excluded to ensure the robustness of the findings. RESULTS: Among the final selection of 729 immune cell phenotypes, three immune phenotypes exhibited statistically significant effects with LUSC. CD28 expression on resting CD4 regulatory T cells (OR 1.0980, 95% CI: 1.0627-1.1344, p < 0.0001) and CD45RA + CD28- CD8 + T cell %T cell (OR 1.0011, 95% CI: 1.0007; 1.0015, p < 0.0001) were associated with increased susceptibility to LUSC. Conversely, CCR2 expression on monocytes (OR 0.9399, 95% CI: 0.9177-0.9625, p < 0.0001) was correlated with a decreased risk of LUSC. However, no significant causal relationships were established between any immune cell phenotypes and LUAD. CONCLUSION: This study demonstrates that specific immune cell types are associated with the risk of LUSC but not with LUAD. While these findings are derived solely from European populations, they still provide clues for a deeper understanding of the immunological mechanisms underlying lung cancer and may offer new directions for future therapeutic strategies and preventive measures.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Mendelian Randomization Analysis , Phenotype , Humans , Lung Neoplasms/genetics , Lung Neoplasms/immunology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Receptors, CCR2/genetics , CD8-Positive T-Lymphocytes/immunology , CD28 Antigens/genetics
10.
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
11.
J Neuroinflammation ; 21(1): 134, 2024 May 27.
Article in English | MEDLINE | ID: mdl-38802868

ABSTRACT

BACKGROUND: Since the 1990s, evidence has accumulated that macrophages promote peripheral nerve regeneration and are required for enhancing regeneration in the conditioning lesion (CL) response. After a sciatic nerve injury, macrophages accumulate in the injury site, the nerve distal to that site, and the axotomized dorsal root ganglia (DRGs). In the peripheral nervous system, as in other tissues, the macrophage response is derived from both resident macrophages and recruited monocyte-derived macrophages (MDMs). Unresolved questions are: at which sites do macrophages enhance nerve regeneration, and is a particular population needed. METHODS: Ccr2 knock-out (KO) and Ccr2gfp/gfp knock-in/KO mice were used to prevent MDM recruitment. Using these strains in a sciatic CL paradigm, we examined the necessity of MDMs and residents for CL-enhanced regeneration in vivo and characterized injury-induced nerve inflammation. CL paradigm variants, including the addition of pharmacological macrophage depletion methods, tested the role of various macrophage populations in initiating or sustaining the CL response. In vivo regeneration, measured from bilateral proximal test lesions (TLs) after 2 d, and macrophages were quantified by immunofluorescent staining. RESULTS: Peripheral CL-enhanced regeneration was equivalent between crush and transection CLs and was sustained for 28 days in both Ccr2 KO and WT mice despite MDM depletion. Similarly, the central CL response measured in dorsal roots was unchanged in Ccr2 KO mice. Macrophages at both the TL and CL, but not between them, stained for the pro-regenerative marker, arginase 1. TL macrophages were primarily CCR2-dependent MDMs and nearly absent in Ccr2 KO and Ccr2gfp/gfp KO mice. However, there were only slightly fewer Arg1+ macrophages in CCR2 null CLs than controls due to resident macrophage compensation. Zymosan injection into an intact WT sciatic nerve recruited Arg1+ macrophages but did not enhance regeneration. Finally, clodronate injection into Ccr2gfp KO CLs dramatically reduced CL macrophages. Combined with the Ccr2gfp KO background, depleting MDMs and TL macrophages, and a transection CL, physically removing the distal nerve environment, nearly all macrophages in the nerve were removed, yet CL-enhanced regeneration was not impaired. CONCLUSIONS: Macrophages in the sciatic nerve are neither necessary nor sufficient to produce a CL response.


Subject(s)
Macrophages , Nerve Regeneration , Peripheral Nerve Injuries , Receptors, CCR2 , Wallerian Degeneration , Animals , Macrophages/metabolism , Macrophages/pathology , Mice , Nerve Regeneration/physiology , Wallerian Degeneration/pathology , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/deficiency , Peripheral Nerve Injuries/pathology , Peripheral Nerve Injuries/metabolism , Mice, Inbred C57BL , Mice, Knockout , Sciatic Neuropathy/pathology , Axons/pathology , Mice, Transgenic , Disease Models, Animal , Sciatic Nerve/injuries , Sciatic Nerve/pathology , Ganglia, Spinal/metabolism , Ganglia, Spinal/pathology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism
12.
Int Immunopharmacol ; 133: 111877, 2024 May 30.
Article in English | MEDLINE | ID: mdl-38608440

ABSTRACT

The gut microbiome plays an important role in tumor growth by regulating immune cell function. However, the role of the gut microbiome-mediated monocytes in liver metastasis remains unclear. In this study, we found that fecal microbiome transplantation (FMT) from the stool of patients with liver metastasis (LM) significantly promoted liver metastasis compared with healthy donors (HD). Monocytes were upregulated in liver tissues by the CCL2/CCR2 axis in LM patients' stool transplanted mouse model. CCL2/CCR2 inhibition and monocyte depletion significantly suppress liver metastasis. FMT using LM patients' stool enhanced the plasma lipopolysaccharides (LPS) concentration. The LPS/TLR4 signaling pathway is crucial for gut microbiome-mediated liver metastasis. These results indicated that monocytes contribute to liver metastasis via the CCL2/CCR2 axis.


Subject(s)
Chemokine CCL2 , Fecal Microbiota Transplantation , Gastrointestinal Microbiome , Liver Neoplasms , Monocytes , Receptors, CCR2 , Toll-Like Receptor 4 , Gastrointestinal Microbiome/immunology , Animals , Humans , Liver Neoplasms/secondary , Liver Neoplasms/immunology , Monocytes/immunology , Chemokine CCL2/metabolism , Mice , Receptors, CCR2/metabolism , Toll-Like Receptor 4/metabolism , Male , Lipopolysaccharides/immunology , Mice, Inbred C57BL , Female , Signal Transduction , Cell Line, Tumor , Liver/pathology , Liver/immunology , Liver/metabolism
13.
Cancer Cell ; 42(5): 885-903.e4, 2024 May 13.
Article in English | MEDLINE | ID: mdl-38608702

ABSTRACT

With limited treatment options, cachexia remains a major challenge for patients with cancer. Characterizing the interplay between tumor cells and the immune microenvironment may help identify potential therapeutic targets for cancer cachexia. Herein, we investigate the critical role of macrophages in potentiating pancreatic cancer induced muscle wasting via promoting TWEAK (TNF-like weak inducer of apoptosis) secretion from the tumor. Specifically, depletion of macrophages reverses muscle degradation induced by tumor cells. Macrophages induce non-autonomous secretion of TWEAK through CCL5/TRAF6/NF-κB pathway. TWEAK promotes muscle atrophy by activating MuRF1 initiated muscle remodeling. Notably, tumor cells recruit and reprogram macrophages via the CCL2/CCR2 axis and disrupting the interplay between macrophages and tumor cells attenuates muscle wasting. Collectively, this study identifies a feedforward loop between pancreatic cancer cells and macrophages, underlying the non-autonomous activation of TWEAK secretion from tumor cells thereby providing promising therapeutic targets for pancreatic cancer cachexia.


Subject(s)
Cachexia , Cytokine TWEAK , Macrophages , Pancreatic Neoplasms , Cachexia/metabolism , Cachexia/etiology , Cachexia/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/complications , Cytokine TWEAK/metabolism , Animals , Humans , Macrophages/metabolism , Mice , NF-kappa B/metabolism , Cell Line, Tumor , Tumor Microenvironment , Muscular Atrophy/metabolism , Muscular Atrophy/etiology , Muscular Atrophy/pathology , Chemokine CCL5/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism , Tumor Necrosis Factors/metabolism , Receptors, CCR2/metabolism , Chemokine CCL2/metabolism , Mice, Inbred C57BL
14.
Nat Immunol ; 25(5): 802-819, 2024 May.
Article in English | MEDLINE | ID: mdl-38684922

ABSTRACT

Sepsis induces immune alterations, which last for months after the resolution of illness. The effect of this immunological reprogramming on the risk of developing cancer is unclear. Here we use a national claims database to show that sepsis survivors had a lower cumulative incidence of cancers than matched nonsevere infection survivors. We identify a chemokine network released from sepsis-trained resident macrophages that triggers tissue residency of T cells via CCR2 and CXCR6 stimulations as the immune mechanism responsible for this decreased risk of de novo tumor development after sepsis cure. While nonseptic inflammation does not provoke this network, laminarin injection could therapeutically reproduce the protective sepsis effect. This chemokine network and CXCR6 tissue-resident T cell accumulation were detected in humans with sepsis and were associated with prolonged survival in humans with cancer. These findings identify a therapeutically relevant antitumor consequence of sepsis-induced trained immunity.


Subject(s)
Macrophages , Neoplasms , Sepsis , Humans , Sepsis/immunology , Macrophages/immunology , Female , Neoplasms/immunology , Neoplasms/therapy , Male , Receptors, CXCR6/metabolism , Animals , T-Lymphocytes/immunology , Receptors, CCR2/metabolism , Middle Aged , Mice , Aged , Chemokines/metabolism , Adult
15.
J Clin Invest ; 134(10)2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38564300

ABSTRACT

Nuclear factor κ-B (NFκB) is activated in iPSC-cardiac myocytes from patients with arrhythmogenic cardiomyopathy (ACM) under basal conditions, and inhibition of NFκB signaling prevents disease in Dsg2mut/mut mice, a robust mouse model of ACM. Here, we used genetic approaches and single-cell RNA-Seq to define the contributions of immune signaling in cardiac myocytes and macrophages in the natural progression of ACM using Dsg2mut/mut mice. We found that NFκB signaling in cardiac myocytes drives myocardial injury, contractile dysfunction, and arrhythmias in Dsg2mut/mut mice. NFκB signaling in cardiac myocytes mobilizes macrophages expressing C-C motif chemokine receptor-2 (CCR2+ cells) to affected areas within the heart, where they mediate myocardial injury and arrhythmias. Contractile dysfunction in Dsg2mut/mut mice is caused both by loss of heart muscle and negative inotropic effects of inflammation in viable muscle. Single nucleus RNA-Seq and cellular indexing of transcriptomes and epitomes (CITE-Seq) studies revealed marked proinflammatory changes in gene expression and the cellular landscape in hearts of Dsg2mut/mut mice involving cardiac myocytes, fibroblasts, and CCR2+ macrophages. Changes in gene expression in cardiac myocytes and fibroblasts in Dsg2mut/mut mice were dependent on CCR2+ macrophage recruitment to the heart. These results highlight complex mechanisms of immune injury and regulatory crosstalk between cardiac myocytes, inflammatory cells, and fibroblasts in the pathogenesis of ACM.


Subject(s)
Desmoglein 2 , Disease Models, Animal , Macrophages , NF-kappa B , Receptors, CCR2 , Signal Transduction , Animals , Mice , Macrophages/metabolism , Macrophages/pathology , Macrophages/immunology , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Desmoglein 2/genetics , Desmoglein 2/metabolism , NF-kappa B/metabolism , NF-kappa B/genetics , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Myocytes, Cardiac/immunology , Humans , Arrhythmogenic Right Ventricular Dysplasia/genetics , Arrhythmogenic Right Ventricular Dysplasia/metabolism , Arrhythmogenic Right Ventricular Dysplasia/pathology , Myocardium/pathology , Myocardium/metabolism , Myocardium/immunology
16.
J Ethnopharmacol ; 329: 118169, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38621463

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: The Ba-Qi-Rougan formula (BQRGF) is a traditional and effective compound prescription from Traditional Chinese Medicine (TCM) utilized in treating hepatic fibrosis (HF). AIM OF THE STUDY: We aimed to evaluate the therapeutic efficacy of BQRGF on HF and explore the underlying mechanisms of action. MATERIALS AND METHODS: UPLC-Q-TOF-MS technology was employed to identify the material basis of BQRGF. Mice with carbon tetrachloride (CCl4)-induced HF received BQRGF at three doses (3.87, 7.74, and 15.48 g/kg per day). We examined serum and liver biochemical indicators and liver histology to assess the therapeutic impact. Primary mouse cells were isolated and utilized for experimental analysis. MSMP expression levels were examined in vitro and in vivo experimental models, including human and mouse tissue. Furthermore, lentivirus and small interfering RNA (siRNA) transfections were employed to manipulate microseminoprotein (MSMP) expression in LO2 cells (human normal liver cells). These manipulated LO2 cells were then co-cultured with LX2 human hepatic stellate cells (HSCs). Through the modulation of MSMP expression in co-cultured cells, administering recombinant MSMP (rMSMP) with or without BQRGF-medicated serum, and using specific pathway inhibitors or agonists in LX2 cells, we elucidated the underlying mechanisms. RESULTS: A total of 48 compounds were identified from BQRGF, with 12 compounds being absorbed into the bloodstream and 9 compounds being absorbed into the liver. Four weeks of BQRGF treatment in the HF mouse model led to significant improvements in biochemical and molecular assays and histopathology, particularly in the medium and high-dose groups. These improvements included a reduction in the level of liver injury and fibrosis-related factors. MSMP levels were elevated in human and mouse fibrotic liver tissues, and this increase was mitigated in HF mice treated with BQRGF. Moreover, primary cells and co-culture studies revealed that BQRGF reduced MSMP expression, decreased the expression of the hepatic stellate cell (HSC) activation markers, and suppressed critical phosphorylated protein levels in the CCR2/PI3K/AKT pathway. These findings were further validated using CCR2/PI3K/AKT signaling inhibitors and agonists in MSMP-activated LX2 cells. CONCLUSIONS: Collectively, our results suggest that BQRGF combats HF by diminishing MSMP levels and inhibiting MSMP-induced HSC activation through the CCR2/PI3K/AKT pathway.


Subject(s)
Drugs, Chinese Herbal , Hepatic Stellate Cells , Liver Cirrhosis , Phosphatidylinositol 3-Kinases , Signal Transduction , Animals , Hepatic Stellate Cells/drug effects , Hepatic Stellate Cells/metabolism , Drugs, Chinese Herbal/pharmacology , Humans , Mice , Male , Liver Cirrhosis/drug therapy , Liver Cirrhosis/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/chemically induced , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/antagonists & inhibitors , Mice, Inbred C57BL , Carbon Tetrachloride , Cell Line
17.
PLoS Negl Trop Dis ; 18(4): e0012112, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38669292

ABSTRACT

Visceral leishmaniasis (VL) is a potentially fatal parasitic infection caused by Leishmania donovani in India. L. donovani is an obligate intracellular protozoan residing mostly in macrophages of the reticuloendothelial system throughout chronic infection. Monocytic phagocytes are critical in the pathogenesis of different forms of leishmaniasis. Subsets of monocytes are distinguished by their surface markers into CD14+CD16- classical monocytes, CD14+CD16+ intermediate monocytes, and CD16++CD14low non-classical monocyte subsets. During cutaneous leishmaniasis (CL), intermediate monocyte are reported to be a source of inflammatory cytokines IL-1ß and TNF, and they express CCR2 attracting them to sites of inflammatory pathology. We examined monocyte subsets in the blood and bone marrow of patients with VL from an endemic site in Bihar, India, and found these contrasted with the roles of monocytes in CL. During VL, intermediate and non-classical CD16+ monocyte subsets expressed instead a non-inflammatory phenotype with low CCR2, high CX3CR1 and low microbicidal oxidant generation, making them more similar to patrolling monocytes than inflammatory cells. Bone marrow CD16+ monocyte subsets expressed a phenotype that might be more similar to the inflammatory subsets of CL, although our inability to obtain bone marrow from healthy donors in the endemic region hampered this interpretation Overall the data suggest that CD16+ intermediate monocyte subsets in VL patients express a phenotypes that contributes to an immunosuppressed pathologic immune state, but in contrast to CL, these do not mediate localized inflammatory responses.


Subject(s)
Bone Marrow , Leishmaniasis, Visceral , Monocytes , Leishmaniasis, Visceral/immunology , Leishmaniasis, Visceral/parasitology , Humans , Monocytes/immunology , India , Adult , Male , Bone Marrow/parasitology , Female , Receptors, IgG/analysis , Receptors, IgG/metabolism , Leishmania donovani/immunology , Leishmania donovani/physiology , Young Adult , Adolescent , Receptors, CCR2/metabolism , Middle Aged , Child , Receptors, Chemokine/metabolism , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/metabolism , Cytokines/metabolism
18.
Cell Rep ; 43(4): 114120, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38625796

ABSTRACT

Border-associated macrophages (BAMs) are tissue-resident macrophages that reside at the border of the central nervous system (CNS). Since BAMs originate from yolk sac progenitors that do not persist after birth, the means by which this population of cells is maintained is not well understood. Using two-photon microscopy and multiple lineage-tracing strategies, we determine that CCR2+ monocytes are significant contributors to BAM populations following disruptions of CNS homeostasis in adult mice. After BAM depletion, while the residual BAMs possess partial self-repopulation capability, the CCR2+ monocytes are a critical source of the repopulated BAMs. In addition, we demonstrate the existence of CCR2+ monocyte-derived long-lived BAMs in a brain compression model and in a sepsis model after the initial disruption of homeostasis. Our study reveals that the short-lived CCR2+ monocytes transform into long-lived BAM-like cells at the CNS border and subsequently contribute to BAM populations.


Subject(s)
Brain , Macrophages , Monocytes , Receptors, CCR2 , Animals , Receptors, CCR2/metabolism , Monocytes/metabolism , Macrophages/metabolism , Mice , Brain/pathology , Brain/metabolism , Mice, Inbred C57BL , Homeostasis
19.
Adv Drug Deliv Rev ; 209: 115318, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643840

ABSTRACT

The communication between cells and their microenvironment represents an intrinsic and essential attribute that takes place in several biological processes, including tissue homeostasis and tissue repair. Among these interactions, inflammation is certainly a central biological response that occurs through cytokines and the crosstalk with their respective receptors. In particular, the interaction between CCL2 and its main receptor, CCR2, plays a pivotal role in both harmful and protective inflammatory states, including cancer-mediated inflammation. The activation of the CCL2/CCR2 axis was shown to dictate the migration of macrophages with immune-suppressive phenotype and to aggravate the progression of different cancer types. In addition, this interaction mediates metastasis formation, further limiting the potential therapeutic outcome of anti-cancer drugs. Attempts to inhibit pharmacologically the CCL2/CCR2 axis have yet to show its anti-cancer efficacy as a single agent, but it sheds light on its role as a powerful tool to selectively alleviate pro-tumorigenic and anti-repair inflammation. In this review, we will elucidate the role of CCL2/CCR2 axis in promoting cancer inflammation by activating the host pro-tumorigenic phenotype. Moreover, we will provide some insight into the potential therapeutic benefit of targeting the CCL2/CCR2 axis for cancer and inflammation using novel delivery systems, aiming to sensitize non-responders to currently approved immunotherapies and offer new combinatory approaches.


Subject(s)
Chemokine CCL2 , Inflammation , Nanomedicine , Neoplasms , Receptors, CCR2 , Humans , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Neoplasms/pathology , Inflammation/drug therapy , Inflammation/metabolism , Chemokine CCL2/metabolism , Chemokine CCL2/antagonists & inhibitors , Animals , Tumor Microenvironment/drug effects , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/administration & dosage , Drug Delivery Systems
20.
J Cell Mol Med ; 28(9): e18315, 2024 May.
Article in English | MEDLINE | ID: mdl-38680032

ABSTRACT

Oestrogen is known to be strongly associated with ovarian cancer. There was much work to show the importance of lncRNA SNHG17 in ovarian cancer. However, no study has revealed the molecular regulatory mechanism and functional effects between oestrogen and SNHG17 in the development and metastasis of ovarian cancer. In this study, we found that SNHG17 expression was significantly increased in ovarian cancer and positively correlated with oestrogen treatment. Oestrogen could promote M2 macrophage polarization as well as ovarian cancer cells SKOV3 and ES2 cell exosomal SNHG17 expression. When exposure to oestrogen, exosomal SNHG17 promoted ovarian cancer cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT) in vitro, and tumour growth and lung metastasis in vivo by accelerating M2-like phenotype of macrophages. Mechanically, exosomal SNHG17 could facilitate the release of CCL13 from M2 macrophage via the PI3K-Akt signalling pathway. Moreover, CCL13-CCR2 axis was identified to be involved in ovarian cancer tumour behaviours driven by oestrogen. There results demonstrate a novel mechanism that exosomal SNHG17 exerts an oncogenic effect on ovarian cancer via the CCL13-CCR2-M2 macrophage axis upon oestrogen treatment, of which SNHG17 may be a potential biomarker and therapeutic target for ovarian cancer responded to oestrogen.


Subject(s)
Cell Proliferation , Epithelial-Mesenchymal Transition , Estrogens , Exosomes , Gene Expression Regulation, Neoplastic , Macrophages , Ovarian Neoplasms , RNA, Long Noncoding , Receptors, CCR2 , Female , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Humans , Macrophages/metabolism , Macrophages/drug effects , Exosomes/metabolism , Estrogens/metabolism , Estrogens/pharmacology , Cell Line, Tumor , Animals , Receptors, CCR2/metabolism , Receptors, CCR2/genetics , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Mice , Epithelial-Mesenchymal Transition/drug effects , Cell Movement/drug effects , Disease Progression , Signal Transduction , Mice, Nude
SELECTION OF CITATIONS
SEARCH DETAIL
...