Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 155
Filter
1.
Bull Exp Biol Med ; 172(2): 263-269, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34855085

ABSTRACT

Activation and migration of donor T cells to the host target organs are critical mechanisms in the pathogenesis of graft-versus-host disease (GVHD). The role of monocyte chemoattractant protein-1 (MCP-1/CCL2) and its receptor CCR2 in the recruitment of T cells during immune or inflammatory response is also well known. For elucidation of the mechanism of the therapeutic effect of human bone marrow derived-mesenchymal stem cells (MSC) in GVHD, we studied the effect of these cells on migration of activated donor T cells through the CCL2-CCR2 axis in vitro. MSC were expanded from donors' bone marrow mononuclear cells. After co-culturing of IL-2-activated T cells with allogeneic MSC at different ratios, the levels of CCL2 in supernatants were measured by ELISA, and CCR2 expression in CD4+/CD8+ T cells subsets were detected by flow cytometry. The effect of MSC on the migration of activated T cells in the Transwell system was studied in the absence or presence of CCL2. Our results show that CCL2 levels in supernatants of co-cultures were significantly higher than in MSC monoculture and this increase depended on the number of MSC. MSC inhibited proliferation of T cells, but did not change the percentages of CD4+ and CD8+ T cells subsets. MSC can up-regulate the CCR2 expression in CD8+ subsets rather than in CD4+ subsets; MSC enhanced migration of IL-2-activated T cells to CCL2 by increasing the expression of CCR2. The data demonstrate that MSC can enhance chemotaxis of cytokine-activated T cells through the CCL2-CCR2 axis in vitro.


Subject(s)
Chemotaxis, Leukocyte/physiology , Mesenchymal Stem Cells/physiology , T-Lymphocytes/physiology , Adult , Cell Differentiation/immunology , Cells, Cultured , Chemokine CCL2/physiology , Coculture Techniques , Humans , Immunophenotyping , Lymphocyte Activation , Mesenchymal Stem Cells/cytology , Receptors, CCR2/physiology , Signal Transduction , T-Lymphocytes/immunology
2.
Sci Rep ; 11(1): 24345, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34934133

ABSTRACT

Mutations in the NF1 tumor suppressor gene are linked to arteriopathy. Nf1 heterozygosity (Nf1+/-) results in robust neointima formation, similar to humans, and myeloid-restricted Nf1+/- recapitulates this phenotype via MEK-ERK activation. Here we define the contribution of myeloid subpopulations to NF1 arteriopathy. Neutrophils from WT and Nf1+/- mice were functionally assessed in the presence of MEK and farnesylation inhibitors in vitro and neutrophil recruitment to lipopolysaccharide was assessed in WT and Nf1+/- mice. Littermate 12-15 week-old male wildtype and Nf1+/- mice were subjected to carotid artery ligation and provided either a neutrophil depleting antibody (1A8), liposomal clodronate to deplete monocytes/macrophages, or PD0325901 and neointima size was assessed 28 days after injury. Bone marrow transplant experiments assessed monocyte/macrophage mobilization during neointima formation. Nf1+/- neutrophils exhibit enhanced proliferation, migration, and adhesion via p21Ras activation of MEK in vitro and in vivo. Neutrophil depletion suppresses circulating Ly6Clow monocytes and enhances neointima size, while monocyte/macrophage depletion and deletion of CCR2 in bone marrow cells abolish neointima formation in Nf1+/- mice. Taken together, these findings suggest that neurofibromin-MEK-ERK activation in circulating neutrophils and monocytes during arterial remodeling is nuanced and points to important cross-talk between these populations in the pathogenesis of NF1 arteriopathy.


Subject(s)
Carotid Artery Injuries/pathology , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Myeloid Progenitor Cells/pathology , Neointima/pathology , Neurofibromatosis 1/pathology , Neurofibromin 1/physiology , Receptors, CCR2/physiology , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Monocytes/pathology , Myeloid Progenitor Cells/metabolism , Neointima/etiology , Neointima/metabolism , Neurofibromatosis 1/etiology , Neurofibromatosis 1/metabolism
3.
Front Immunol ; 12: 763379, 2021.
Article in English | MEDLINE | ID: mdl-34691085

ABSTRACT

Every immune response has accelerators and brakes. Depending on the pathogen or injury, monocytes can play either role, promoting or resolving immunity. Poly I:C, a potent TLR3 ligand, licenses cross-presenting dendritic cells (DC1) to accelerate a robust cytotoxic T cells response against a foreign antigen. Poly I:C thus has promise as an adjuvant in cancer immunotherapy and viral subunit vaccines. Like DC1s, monocytes are also abundant in the LNs. They may act as either immune accelerators or brakes, depending on the inflammatory mediator they encounter. However, little is known about their contribution to adaptive immunity in the context of antigen and Poly I:C. Using monocyte-deficient and chimeric mice, we demonstrate that LN monocytes indirectly dampen a Poly I:C induced antigen-specific cytotoxic T cell response, exerting a "braking" function. This effect is mediated by IL-10 production and induction of suppressor CD4+ T cells. In a metastatic melanoma model, we show that a triple-combination prophylactic treatment consisting of anti-IL-10, tumor peptides and Poly I:C works because removing IL-10 counteracts the monocytic brake, resulting in significantly fewer tumors compared to mice treated with tumor peptides and Poly I:C alone. Finally, in human LN tissue, we observed that monocytes (unlike DCs) express high levels of IL-10, suggesting that anti-IL-10 may be an important addition to treatments. Overall, our data demonstrates that LN monocytes regulate the induction of a robust DC1-mediated immune response. Neutralization of either IL-10 or monocytes can augment Poly I:C-based treatments and enhance T cell cytotoxicity.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Interleukin-10/physiology , Lymph Nodes/immunology , Monocytes/physiology , Poly I-C/pharmacology , T-Lymphocytes, Cytotoxic/drug effects , Animals , Humans , Interleukin-10/antagonists & inhibitors , Melanoma, Experimental/drug therapy , Mice , Mice, Inbred C57BL , Receptors, CCR2/physiology , T-Lymphocytes, Cytotoxic/immunology
4.
Life Sci ; 272: 118808, 2021 May 01.
Article in English | MEDLINE | ID: mdl-33245967

ABSTRACT

BACKGROUND/AIMS: The theory of inflammation is one of the important theories in the pathogenesis of diabetic nephropathy (DN). We herein aimed to explore whether loganin affected macrophage infiltration and activation upon diabetic nephropathy (DN) by a spontaneous DN mice and a co-culture system of glomerular mesangial cells (GMCs) and macrophage cells (RAW264.7) which was induced by advanced glycation end products (AGEs). METHODS AND KEY FINDINGS: Loganin showed remarkable capacity on protecting renal from damage by mitigating diabetic symptoms, improving the histomorphology of the kidney, decreasing the expression of extracellular matrix such as FN, COL-IV and TGF-ß, reversing the production of IL-12 and IL-10 and decreasing the number of infiltrating macrophages in the kidney. Moreover, loganin showed markedly effects by suppressing iNOS and CD16/32 expressions (M1 markers), increasing Arg-1 and CD206 expressions (M2 markers), which were the phenotypic transformation of macrophage. These effects may be attributed to the inhibition of the receptor for AGEs (RAGE) /monocyte chemotactic protein-1 (MCP-1)/CC chemokine receptor 2 (CCR2) signaling pathway, with significantly down-regulated expressions of RAGE, MCP-1 and CCR2 by loganin. Loganin further decreased MCP-1 secretion when RAGE was silenced, which means other target was involved in regulating the MCP-1 expression. While loganin combinated with the inhibitor of CCR2 exerted stronger anti-inhibition effects of iNOS expression, suggesting that CCR2 was the target of loganin in regulating the activation of macrophages. SIGNIFICANCE: Loganin could ameliorate DN kidney damage by inhibiting macrophage infiltration and activation via the MCP-1/CCR2 signaling pathway in DN.


Subject(s)
Diabetic Nephropathies/drug therapy , Iridoids/pharmacology , Macrophages/metabolism , Animals , Chemokine CCL2/metabolism , Chemokine CCL2/physiology , Chemokine CCL8/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Fibronectins/metabolism , Glycation End Products, Advanced/metabolism , Iridoids/metabolism , Kidney/metabolism , Macrophage Activation/drug effects , Macrophages/drug effects , Male , Mesangial Cells/metabolism , Mice , Mice, Inbred C57BL , RAW 264.7 Cells , Receptors, CCR1/metabolism , Receptors, CCR2/metabolism , Receptors, CCR2/physiology , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism
5.
JCI Insight ; 5(18)2020 09 17.
Article in English | MEDLINE | ID: mdl-32780724

ABSTRACT

Tumor-associated macrophages (TAMs) affect cancer progression and therapy. Ovarian carcinoma often metastasizes to the peritoneal cavity. Here, we found 2 peritoneal macrophage subsets in mice bearing ID8 ovarian cancer based on T cell immunoglobulin and mucin domain containing 4 (Tim-4) expression. Tim-4+ TAMs were embryonically originated and locally sustained while Tim-4- TAMs were replenished from circulating monocytes. Tim-4+ TAMs, but not Tim-4- TAMs, promoted tumor growth in vivo. Relative to Tim-4- TAMs, Tim-4+ TAMs manifested high oxidative phosphorylation and adapted mitophagy to alleviate oxidative stress. High levels of arginase-1 in Tim-4+ TAMs contributed to potent mitophagy activities via weakened mTORC1 activation due to low arginine resultant from arginase-1-mediated metabolism. Furthermore, genetic deficiency of autophagy element FAK family-interacting protein of 200 kDa resulted in Tim-4+ TAM loss via ROS-mediated apoptosis and elevated T cell immunity and ID8 tumor inhibition in vivo. Moreover, human ovarian cancer-associated macrophages positive for complement receptor of the immunoglobulin superfamily (CRIg) were transcriptionally, metabolically, and functionally similar to murine Tim-4+ TAMs. Thus, targeting CRIg+ (Tim-4+) TAMs may potentially treat patients with ovarian cancer with peritoneal metastasis.


Subject(s)
Autophagy , Macrophages, Peritoneal/pathology , Membrane Proteins/metabolism , Membrane Proteins/physiology , Ovarian Neoplasms/pathology , Oxidative Stress , Peritoneal Neoplasms/secondary , Adaptation, Physiological , Animals , Autophagy-Related Proteins/physiology , Female , Humans , Leukocyte Common Antigens/physiology , Macrophages, Peritoneal/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/metabolism , Receptors, CCR2/physiology
6.
J Exp Med ; 217(10)2020 10 05.
Article in English | MEDLINE | ID: mdl-32667673

ABSTRACT

C-C chemokine receptor type 2 (CCR2) is expressed on monocytes and facilitates their recruitment to tumors. Though breast cancer cells also express CCR2, its functions in these cells are unclear. We found that Ccr2 deletion in cancer cells led to reduced tumor growth and approximately twofold longer survival in an orthotopic, isograft breast cancer mouse model. Deletion of Ccr2 in cancer cells resulted in multiple alterations associated with better immune control: increased infiltration and activation of cytotoxic T lymphocytes (CTLs) and CD103+ cross-presenting dendritic cells (DCs), as well as up-regulation of MHC class I and down-regulation of checkpoint regulator PD-L1 on the cancer cells. Pharmacological or genetic targeting of CCR2 increased cancer cell sensitivity to CTLs and enabled the cancer cells to induce DC maturation toward the CD103+ subtype. Consistently, Ccr2-/- cancer cells did not induce immune suppression in Batf3-/- mice lacking CD103+ DCs. Our results establish that CCR2 signaling in cancer cells can orchestrate suppression of the immune response.


Subject(s)
Adaptive Immunity/immunology , Immune Tolerance , Mammary Neoplasms, Experimental/immunology , Receptors, CCR2/physiology , Adaptive Immunity/physiology , Animals , Apoptosis , B7-H1 Antigen/metabolism , Dendritic Cells/immunology , Dendritic Cells/physiology , Female , Histocompatibility Antigens Class I/metabolism , Immune Tolerance/immunology , Immune Tolerance/physiology , Interferons/metabolism , Mice , Mice, Inbred C57BL , Receptors, CCR2/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/physiology
7.
Cancer Cell ; 37(6): 786-799.e5, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32516589

ABSTRACT

Generation of tumor-infiltrating lymphocytes begins when tumor antigens reach the lymph node (LN) to stimulate T cells, yet we know little of how tumor material is disseminated among the large variety of antigen-presenting dendritic cell (DC) subsets in the LN. Here, we demonstrate that tumor proteins are carried to the LN within discrete vesicles inside DCs and are then transferred among DC subsets. A synapse is formed between interacting DCs and vesicle transfer takes place in the absence of free exosomes. DCs -containing vesicles can uniquely activate T cells, whereas DCs lacking them do not. Understanding this restricted sharing of tumor identity provides substantial room for engineering better anti-tumor immunity.


Subject(s)
Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/immunology , Melanoma, Experimental/immunology , Myeloid Cells/immunology , Synapses/immunology , T-Lymphocytes/immunology , Animals , Dendritic Cells/cytology , Dendritic Cells/metabolism , Male , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/cytology , Myeloid Cells/metabolism , Receptors, CCR2/physiology , Receptors, CCR7/physiology , Synapses/metabolism , Synapses/pathology , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
8.
Front Immunol ; 11: 675, 2020.
Article in English | MEDLINE | ID: mdl-32425929

ABSTRACT

Sepsis is characterized by a systemic inflammation that can cause an immune dysfunction, for which the underlying mechanisms are unclear. We investigated the impact of cecal ligature and puncture (CLP)-mediated polymicrobial sepsis on monocyte (Mo) mobilization and functions. Our results show that CLP led to two consecutive phases of Mo deployment. The first one occurred within the first 3 days after the induction of the peritonitis, while the second phase was of a larger amplitude and extended up to a month after apparent clinical recovery. The latter was associated with the expansion of Mo in the tissue reservoirs (bone marrow and spleen), their release in the blood and their accumulation in the vasculature of peripheral non-lymphoid tissues. It occurred even after antibiotic treatment but relied on inflammatory-dependent pathways and inversely correlated with increased susceptibility and severity to a secondary infection. The intravascular lung Mo displayed limited activation capacity, impaired phagocytic functions and failed to transfer efficient protection against a secondary infection into monocytopenic CCR2-deficient mice. In conclusion, our work unveiled key dysfunctions of intravascular inflammatory Mo during the recovery phase of sepsis and provided new insights to improve patient protection against secondary infections.


Subject(s)
Inflammation/immunology , Monocytes/immunology , Sepsis/immunology , Animals , Antigens, Ly/analysis , CX3C Chemokine Receptor 1/physiology , Lung/immunology , Mice , Mice, Inbred C57BL , Monocytes/physiology , Neutrophils/immunology , Phagocytosis , Receptors, CCR2/physiology
9.
Cell Commun Signal ; 18(1): 82, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32471499

ABSTRACT

Chemokines are a family of small cytokines, which guide a variety of immune/inflammatory cells to the site of tumor in tumorigenesis. A dysregulated expression of chemokines is implicated in different types of cancer including prostate cancer. The progression and metastasis of prostate cancer involve a complex network of chemokines that regulate the recruitment and trafficking of immune cells. The chemokine CCL2 and its main receptor CCR2 have been receiving particular interest on their roles in cancer pathogenesis. The up-regulation of CCL2/CCR2 and varied immune conditions in prostate cancer, are associated with cancer advancement, metastasis, and relapse. Here we reviewed recent findings, which link CCL2/CCR2 to the inflammation and cancer pathogenesis, and discussed the therapeutic potential of CCL2/CCR2 axis in cancer treatment based on results from our group and other investigators, with a major focus on prostate cancer. Video Abstract.


Subject(s)
Chemokine CCL2/physiology , Inflammation/metabolism , Prostatic Neoplasms/metabolism , Receptors, CCR2/physiology , Animals , Humans , Male
10.
Mol Hum Reprod ; 26(5): 289-300, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32159806

ABSTRACT

Expression of immune function genes within follicle cells has been reported in ovaries from many species. Recent work from our laboratory showed a direct effect of the monocyte chemoattractant protein 1/C-C motif chemokine receptor 2 system within the feline cumulus oocyte complex, by increasing the mRNA levels of key genes involved in the ovulatory cascade in vitro. Studies were designed to evaluate if C-C motif chemokine receptor 2 acts as a novel mediator of the ovulatory cascade in vitro. Therefore, feline cumulus oocyte complexes were cultured in the presence or absence of a highly selective C-C motif chemokine receptor 2 antagonist together with known inducers of cumulus-oocyte expansion and/or oocyte maturation to assess mRNA expression of key genes related to periovulatory events in other species as well as oocyte maturation. Also, the effects of recombinant monocyte chemoattractant protein 1 on spontaneous or gonadotrophin-induced oocyte maturation were assessed. This is an in vitro system using isolated cumulus oocyte complexes from feline ovaries. The present study reveals the modulation of several key ovulatory genes by a highly selective C-C motif chemokine receptor 2 antagonist. However, this antagonist was not enough to block the oocyte maturation induced by gonadotropins or amphiregulin. Nonetheless, recombinant monocyte chemoattractant protein 1 had a significant effect on spontaneous oocyte maturation, increasing the percentage of metaphase II stage oocytes in comparison to the control. This is the first study in any species to establish C-C motif chemokine receptor 2 as a mediator of some actions of the mid-cycle gonadotrophin surge.


Subject(s)
Ovulation/genetics , Receptors, CCR2/physiology , Animals , Cats , Cells, Cultured , Cumulus Cells/metabolism , Cumulus Cells/physiology , Female , In Vitro Oocyte Maturation Techniques/veterinary , Oocytes/metabolism , Oocytes/physiology , Oogenesis/genetics , Ovarian Follicle/metabolism , Ovarian Follicle/physiology
11.
Nephrol Dial Transplant ; 35(2): 227-239, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30597038

ABSTRACT

BACKGROUND: Glomerulosclerosis and tubulointerstitial fibrosis are hallmarks of chronic kidney injury leading to end-stage renal disease. Inflammatory mechanisms contribute to glomerular and interstitial scarring, including chemokine-mediated recruitment of leucocytes. In particular, accumulation of C-C chemokine receptor type 2 (CCR2)-expressing macrophages promotes renal injury and fibrotic remodelling in diseases like glomerulonephritis and diabetic nephropathy. The functional role of CCR2 in the initiation and progression of primary glomerulosclerosis induced by podocyte injury remains to be characterized. METHODS: We analysed glomerular expression of CCR2 and its chemokine ligand C-C motif chemokine ligand 2 (CCL2) in human focal segmental glomerulosclerosis (FSGS). Additionally, CCL2 expression was determined in stimulated murine glomeruli and glomerular cells in vitro. To explore pro-inflammatory and profibrotic functions of CCR2 we induced adriamycin nephropathy, a murine model of FSGS, in BALB/c wild-type and Ccr2-deficient mice. RESULTS: Glomerular expression of CCR2 and CCL2 significantly increased in human FSGS. In adriamycin-induced FSGS, progressive glomerular scarring and reduced glomerular nephrin expression was paralleled by induced glomerular expression of CCL2. Adriamycin exposure stimulated secretion of CCL2 and tumour necrosis factor-α (TNF) in isolated glomeruli and mesangial cells and CCL2 in parietal epithelial cells. In addition, TNF induced CCL2 expression in all glomerular cell populations, most prominently in podocytes. In vivo, Ccr2-deficient mice with adriamycin nephropathy showed reduced injury, macrophage and fibrocyte infiltration and inflammation in glomeruli and the tubulointerstitium. Importantly, glomerulosclerosis and tubulointerstitial fibrosis were significantly ameliorated. CONCLUSIONS: Our data indicate that CCR2 is an important mediator of glomerular injury and progression of FSGS. CCR2- targeting therapies may represent a novel approach for its treatment.


Subject(s)
Fibrosis/etiology , Glomerulosclerosis, Focal Segmental/complications , Inflammation/etiology , Kidney/pathology , Receptors, CCR2/physiology , Animals , Chemokines/metabolism , Fibrosis/pathology , Inflammation/pathology , Kidney/injuries , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout
12.
Angew Chem Int Ed Engl ; 58(47): 16894-16898, 2019 11 18.
Article in English | MEDLINE | ID: mdl-31535788

ABSTRACT

We report the novel chemical design of fluorescent activatable chemokines as highly specific functional probes for imaging subpopulations of immune cells in live tumours. Activatable chemokines behave as AND-gates since they emit only after receptor binding and intracellular activation, showing enhanced selectivity over existing agents. We have applied this strategy to produce mCCL2-MAF as the first probe for in vivo detection of metastasis-associated macrophages in a preclinical model of lung metastasis. This strategy will accelerate the preparation of new chemokine-based probes for imaging immune cell function in tumours.


Subject(s)
Breast Neoplasms/pathology , Fluorescent Dyes/chemistry , Lung Neoplasms/pathology , Macrophages/pathology , Molecular Imaging/methods , Receptors, CCR2/physiology , Animals , Apoptosis , Breast Neoplasms/metabolism , Cell Proliferation , Female , Humans , Lung Neoplasms/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Spectrometry, Fluorescence , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
J Orthop Res ; 37(12): 2561-2574, 2019 12.
Article in English | MEDLINE | ID: mdl-31424112

ABSTRACT

The role of the inflammatory response in articular cartilage degeneration and/or repair is often debated. Chemokine networks play a critical role in directing the recruitment of immune cells to sites of injury and have been shown to regulate cell behavior. In this study, we investigated the role of the CCL2/CCR2 signaling axis in cartilage regeneration and degeneration. CCL2-/- , CCR2-/- , CCL2-/- CCR2-/- , and control (C57) mice were subjected to full-thickness cartilage defect (FTCD) injuries (n = 9/group) within the femoral groove. Cartilage regeneration at 4 and 12 weeks post-FTCD was assessed using a 14-point histological scoring scale. Mesenchymal stem cells (MSCs) (Sca-1+ , CD140a+ ), macrophages (M1:CD38+ , M2:CD206+ , and M0:F4/80+ ) and proliferating cells (Ki67+ ) were quantified within joints using immunofluorescence. The multi-lineage differentiation capacity of Sca1+ MSCs was determined for all mouse strains. ACL transection (ACL-x) was employed to determine if CCL2-/- CCR2-/- mice were protected against osteoarthritis (OA) (n = 6/group). Absence of CCR2, but not CCL2 nor both (CCL2 and CCR2), enhanced spontaneous articular cartilage regeneration by 4 weeks post-FTCD. Furthermore, increased chondrogenesis was observed in MSCs derived from CCR2-/- mice. CCL2 deficiency promoted MSC homing to the adjacent synovium and FTCD at both 4 and 12 weeks post-injury; with no MSCs present at the surface of the FTCD in the remaining strains. Lower OA scores were observed in CCL2-/- CCR2-/- mice at 12 weeks post-ACL-x compared with C57 mice. Our findings demonstrate an inhibitory role for CCR2 in cartilage regeneration after injury, while CCL2 is required for regeneration, acting through a CCR2 independent mechanism. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2561-2574, 2019.


Subject(s)
Cartilage, Articular/physiology , Chemokine CCL2/physiology , Receptors, CCR2/physiology , Regeneration , Animals , Cell Differentiation , Chondrogenesis , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/physiology , Mice , Mice, Inbred C57BL , Osteoarthritis/pathology
14.
J Am Soc Nephrol ; 30(10): 1825-1840, 2019 10.
Article in English | MEDLINE | ID: mdl-31315923

ABSTRACT

BACKGROUND: After bilateral kidney ischemia/reperfusion injury (IRI), monocytes infiltrate the kidney and differentiate into proinflammatory macrophages in response to the initial kidney damage, and then transition to a form that promotes kidney repair. In the setting of unilateral IRI (U-IRI), however, we have previously shown that macrophages persist beyond the time of repair and may promote fibrosis. METHODS: Macrophage homing/survival signals were determined at 14 days after injury in mice subjected to U-IRI and in vitro using coculture of macrophages and tubular cells. Mice genetically engineered to lack Ccr2 and wild-type mice were treated ±CCR2 antagonist RS102895 and subjected to U-IRI to quantify macrophage accumulation, kidney fibrosis, and inflammation 14 and 30 days after the injury. RESULTS: Failure to resolve tubular injury after U-IRI results in sustained expression of granulocyte-macrophage colony-stimulating factor by renal tubular cells, which directly stimulates expression of monocyte chemoattractant protein-1 (Mcp-1) by macrophages. Analysis of CD45+ immune cells isolated from wild-type kidneys 14 days after U-IRI reveals high-level expression of the MCP-1 receptor Ccr2. In mice lacking Ccr2 and wild-type mice treated with RS102895, the numbers of macrophages, dendritic cells, and T cell decreased following U-IRI, as did the expression of profibrotic growth factors and proimflammatory cytokines. This results in a reduction in extracellular matrix and kidney injury markers. CONCLUSIONS: GM-CSF-induced MCP-1/CCR2 signaling plays an important role in the cross-talk between injured tubular cells and infiltrating immune cells and myofibroblasts, and promotes sustained inflammation and tubular injury with progressive interstitial fibrosis in the late stages of U-IRI.


Subject(s)
Chemokine CCL2/physiology , Granulocyte-Macrophage Colony-Stimulating Factor/physiology , Inflammation/etiology , Kidney/blood supply , Kidney/pathology , Receptors, CCR2/physiology , Reperfusion Injury/complications , Animals , Cells, Cultured , Fibrosis/etiology , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Kidney Tubules/cytology , Kidney Tubules/metabolism , Macrophages , Mice
15.
Inflamm Res ; 68(8): 639-642, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31115587

ABSTRACT

OBJECTIVE: The hypoxic milieu at tumor microenvironment is able to drive the behavior of infiltrating tumor cells. Considering that hypoxia-mediated HMGB1 release is known to promote tumor growth, as well to enhance the pro-tumoral profile of M2 macrophages by a RAGE-dependent mechanism, it is tempting to evaluate the potential contribution of HMGB1 under hypoxia to restrain M2 macrophages mobility. METHODS: CCR-2 expression was evaluated in M2 polarized macrophages by western blotting and immunocytochemistry. The secreted levels of CCL-2 and the migration capability were evaluated using an ELISA and a chemotaxis assay, respectively. RESULTS: HMGB1, under hypoxic conditions, markedly reduce both the production of CCL-2 and the expression of its receptor CCR-2; and reduced the migration capacity of M2 macrophages. CONCLUSIONS: These results provided new insights into the mechanisms that regulate M2 macrophages mobility at the tumor microenvironment.


Subject(s)
HMGB1 Protein/physiology , Macrophages/physiology , Receptors, CCR2/physiology , Tumor Hypoxia/physiology , Cell Movement , Chemokine CCL2/physiology , Humans , Receptor for Advanced Glycation End Products/physiology , THP-1 Cells , Tumor Microenvironment
16.
Brain Behav Immun ; 80: 464-473, 2019 08.
Article in English | MEDLINE | ID: mdl-30981714

ABSTRACT

Spontaneous itch and pain are the most common symptoms in various skin diseases, including allergic contact dermatitis (ACD). The chemokine (C-C motif) ligand 2 (CCL2, also referred to as monocyte chemoattractant protein 1 (MCP-1)) and its receptor CCR2 are involved in the pathophysiology of ACD, but little is known of the role of CCL2/CCR2 for the itch- and pain-behaviors accompanying the murine model of this disorder, termed contact hypersensitivity (CHS). C57BL/6 mice previously sensitized to the hapten, squaric acid dibutyl ester, applied to the abdomen were subsequently challenged twice with the hapten delivered to either the cheek or to the hairy skin of the hind paw resulting in CHS at that site. By 24 h after the 2nd challenge to the hind paw CCL2 and CCR2 mRNA, protein, and signaling activity were upregulated in the dorsal root ganglion (DRG). Calcium imaging and whole-cell current-clamp recordings revealed that CCL2 directly acted on its neuronal receptor, CCR2 to activate a subset of small-diameter, nociceptive-like DRG neurons retrogradely labeled from the CHS site. Intradermal injection of CCL2 into the site of CHS on the cheek evoked site-directed itch- and pain-like behaviors which could be attenuated by prior delivery of an antagonist of CCR2. In contrast, CCL2 failed to elicit either type of behavior in control mice. Results are consistent with the hypothesis that CHS upregulates CCL2/CCR2 signaling in a subpopulation of cutaneous small diameter DRG neurons and that CCL2 can activate these neurons through neuronal CCR2 to elicit itch- and pain-behavior. Targeting the CCL2/CCR2 signaling might be beneficial for the treatment of the itch and pain sensations accompanying ACD in humans.


Subject(s)
Chemokine CCL2/metabolism , Dermatitis, Allergic Contact/metabolism , Receptors, CCR2/metabolism , Animals , Chemokine CCL2/physiology , Cyclobutanes/pharmacology , Dermatitis, Allergic Contact/physiopathology , Disease Models, Animal , Ganglia, Spinal/metabolism , Male , Mice , Mice, Inbred C57BL , Pain/metabolism , Patch-Clamp Techniques , Pruritus/metabolism , Receptors, CCR2/physiology , Sensory Receptor Cells/metabolism , Signal Transduction , Skin/metabolism
17.
J Neurosci ; 39(18): 3412-3433, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30833511

ABSTRACT

Peripheral nerve injury results in persistent motor deficits, even after the nerve regenerates and muscles are reinnervated. This lack of functional recovery is partly explained by brain and spinal cord circuit alterations triggered by the injury, but the mechanisms are generally unknown. One example of this plasticity is the die-back in the spinal cord ventral horn of the projections of proprioceptive axons mediating the stretch reflex (Ia afferents). Consequently, Ia information about muscle length and dynamics is lost from ventral spinal circuits, degrading motor performance after nerve regeneration. Simultaneously, there is activation of microglia around the central projections of peripherally injured Ia afferents, suggesting a possible causal relationship between neuroinflammation and Ia axon removal. Therefore, we used mice (both sexes) that allow visualization of microglia (CX3CR1-GFP) and infiltrating peripheral myeloid cells (CCR2-RFP) and related changes in these cells to Ia synaptic losses (identified by VGLUT1 content) on retrogradely labeled motoneurons. Microgliosis around axotomized motoneurons starts and peaks within 2 weeks after nerve transection. Thereafter, this region becomes infiltrated by CCR2 cells, and VGLUT1 synapses are lost in parallel. Immunohistochemistry, flow cytometry, and genetic lineage tracing showed that infiltrating CCR2 cells include T cells, dendritic cells, and monocytes, the latter differentiating into tissue macrophages. VGLUT1 synapses were rescued after attenuating the ventral microglial reaction by removal of colony stimulating factor 1 from motoneurons or in CCR2 global KOs. Thus, both activation of ventral microglia and a CCR2-dependent mechanism are necessary for removal of VGLUT1 synapses and alterations in Ia-circuit function following nerve injuries.SIGNIFICANCE STATEMENT Synaptic plasticity and reorganization of essential motor circuits after a peripheral nerve injury can result in permanent motor deficits due to the removal of sensory Ia afferent synapses from the spinal cord ventral horn. Our data link this major circuit change with the neuroinflammatory reaction that occurs inside the spinal cord following injury to peripheral nerves. We describe that both activation of microglia and recruitment into the spinal cord of blood-derived myeloid cells are necessary for motor circuit synaptic plasticity. This study sheds new light into mechanisms that trigger major network plasticity in CNS regions removed from injury sites and that might prevent full recovery of function, even after successful regeneration.


Subject(s)
Microglia/physiology , Motor Neurons/physiology , Myelitis/physiopathology , Neuronal Plasticity , Peripheral Nerve Injuries/physiopathology , Receptors, CCR2/physiology , Spinal Cord/physiopathology , Animals , Female , Male , Mice, Inbred C57BL , Mice, Transgenic , Myelitis/etiology , Peripheral Nerve Injuries/complications , Sciatic Nerve/injuries , Sciatic Nerve/physiopathology , Synapses/physiology
18.
Hear Res ; 374: 49-57, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30710792

ABSTRACT

The blood-perilymph barrier serves a critical role by separating the components of blood from inner ear fluids, limiting traffic of cells, proteins and other solutes into the labyrinth, and allowing gas (O2-CO2) exchange. Inflammation produces changes in the blood-perilymph barrier resulting in increased vascular permeability. It is commonly thought that compromise of the blood-inner ear barrier would lead to hearing impairment through loss of the endocochlear potential (EP). In fact, the effect of increasing cochlear vascular permeability on hearing function and EP is poorly understood. We used a novel method to measure the integrity of the blood-perilymph barrier and demonstrated the effects of barrier compromise on ABR threshold and EP. We also investigated the contribution of CX3CR1 cochlear macrophages and CCR2 inflammatory monocytes to barrier function after systemic exposure to lipopolysaccharide (LPS). We found that systemic LPS induced a profound change in vascular permeability, which correlated with minimal change in ABR threshold and EP. Macrophage depletion using CX3CR1-DTR mice did not alter the baseline permeability of cochlear vessels and resulted in preservation of barrier function in LPS-treated animals. We conclude that cochlear macrophages are not required to maintain the barrier in normal mice and activated macrophages are a critical factor in breakdown of the barrier after LPS. CCR2 null mice demonstrated that LPS induction of barrier leakiness occurs in the absence of CCR2 expression. Thus, enhanced aminoglycoside ototoxicity after LPS can be linked to the expression of CCR2 in inflammatory monocytes, and not to preservation of the blood-perilymph barrier in CCR2 knockout mice.


Subject(s)
Cochlea/blood supply , Cochlea/physiology , Macrophages/physiology , Monocytes/physiology , Perilymph/cytology , Perilymph/physiology , Animals , CX3C Chemokine Receptor 1/genetics , CX3C Chemokine Receptor 1/physiology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Cochlea/cytology , Diphtheria Toxin/toxicity , Heparin-binding EGF-like Growth Factor/genetics , Heparin-binding EGF-like Growth Factor/physiology , Lipopolysaccharides/toxicity , Macrophage Activation/physiology , Macrophages/drug effects , Mice , Mice, Knockout , Mice, Transgenic , Ototoxicity/pathology , Ototoxicity/physiopathology , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR2/physiology
19.
Biol Reprod ; 100(4): 1046-1056, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30395163

ABSTRACT

Studies were designed to (a) evaluate the mRNA expression of the C-C motif chemokine receptor 2 (CCR2) and its chemokine ligands, as well as genes related to periovulatory events, within the cumulus oocyte complex (COC) and follicle wall after a luteinizing hormone (LH) stimulus in cultured feline antral follicles; (b) assess the immunolocalization of CCR2 and its main ligand (monocyte chemoattractant protein 1, MCP1) within the feline COC; and (c) examine the direct effects of exogenous recombinant MCP1 on mRNA expression of the CCR2 receptor and MCP1 as well as key periovulatory genes in the COC, using a feline COC culture system. Both culture systems were developed by our laboratory and exhibit physiological response to gonadotropin stimuli. In summary, this study demonstrated mRNA expression of CCR2 receptor and its assessed ligands (MCP1, MCP2, MCP3, and MCP4) within the feline COC and follicle antral wall, and a significant increase in CCR2 mRNA by LH within the COC. Also, CCR2 and MCP1 immunoreactivity was observed in the oocyte and cumulus cells of the feline COC. Remarkably, this is the first report, in any species, describing a direct effect of the recombinant MCP1 in the CCR2/MCP1 system within the COC, by increasing the mRNA levels of key genes involved in the ovulatory cascade, as well as its own receptor CCR2. Together, these data suggest that CCR2 receptor signaling in the COC may regulate events critical for promoting cumulus oocyte expansion and/or oocyte maturation.


Subject(s)
Chemokine CCL2/physiology , Cumulus Cells/metabolism , Oocytes/metabolism , Receptors, CCR2/physiology , Animals , Cats , Cells, Cultured , Chemokine CCL2/genetics , Cumulus Cells/cytology , Female , Oocytes/cytology , Oogenesis/genetics , Ovarian Follicle/cytology , Ovarian Follicle/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Signal Transduction/genetics
20.
Am J Pathol ; 188(12): 2786-2799, 2018 12.
Article in English | MEDLINE | ID: mdl-30470496

ABSTRACT

Although antibiotics are useful, they can also bring negative effects. We found that antibiotic-treated mice exhibit an alteration in the gene expression profile of corneal tissues and a decrease in corneal nerve density. During corneal wound healing, antibiotic treatment was found to impair corneal nerve regeneration, an effect that could be largely reversed by reconstitution of the gut microbiota via fecal transplant. Furthermore, CCR2- corneal macrophages were found to participate in the repair of damaged corneal nerves, and a decrease in CCR2- corneal macrophages in antibiotic-treated mice, which could be reversed by fecal transplant, was observed. Adoptive transfer of CCR2- corneal macrophages promoted corneal nerve regeneration in antibiotic-treated mice. The application of probiotics after administration of antibiotics also restored the proportion of CCR2- corneal macrophages and increased the regeneration of corneal nerve fibers after epithelial abrasion. These results suggest that dysbiosis of the gut microbiota induced by antibiotic treatment impairs corneal nerve regeneration by affecting CCR2- macrophage distribution in the cornea. This study also indicates the potential of probiotics as a therapeutic strategy for promoting the regeneration of damaged corneal nerve fibers when the gut microbiota is in dysbiosis.


Subject(s)
Anti-Bacterial Agents/adverse effects , Corneal Injuries/etiology , Dysbiosis/complications , Gastrointestinal Microbiome/drug effects , Macrophages/immunology , Nerve Regeneration/immunology , Receptors, CCR2/physiology , Animals , Cells, Cultured , Corneal Injuries/metabolism , Corneal Injuries/pathology , Disease Models, Animal , Dysbiosis/chemically induced , Dysbiosis/metabolism , Female , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Mice , Mice, Inbred C57BL , Nerve Regeneration/drug effects , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...