Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters










Publication year range
1.
Int J Radiat Oncol Biol Phys ; 113(2): 390-400, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35143888

ABSTRACT

PURPOSE: Cranial radiation therapy for the treatment of pediatric brain tumors results in changes to brain development that are detectable with magnetic resonance imaging. We have previously demonstrated similar structural changes in both humans and mice. The goal of the current study was to examine the role of inflammation in this response. Because neuroanatomic volume deficits in pediatric survivors are more pronounced in female patients, we also evaluated possible dependence on sex. METHODS AND MATERIALS: Other studies have shown that male mice deficient in the C-C chemokine ligand 2 gene (Ccl2; previously Mcp-1) have a muted neuroinflammatory response after irradiation. We irradiated Ccl2-/- (HOM; female = 12, male = 13), Ccl2-/+ (HET; female = 13, male = 16), and Ccl2+/+ (WT; female = 11, male = 13) mice with a whole brain dose of 7 Gy during infancy. Control mice (with approximately equal group sizes) were anesthetized but not irradiated. In vivo magnetic resonance images were acquired at 4 time points up to 3 months after irradiation, and deformation-based morphometry was used to identify volume differences. RESULTS: Irradiation of WT mice resulted in a deficit in neuroanatomic growth with limited sex dependence. HOM and HET male mice were significantly protected from this radiation-induced damage, whereas HOM and HET female mice were not. CONCLUSIONS: Interventions aimed at mitigating the effects of cranial radiation therapy in pediatric cancer survivors by modulating inflammatory response will need to consider patient sex.


Subject(s)
Brain , Chemokine CCL2 , Cranial Irradiation , Radiation Injuries, Experimental , Animals , Brain/diagnostic imaging , Brain/radiation effects , Chemokine CCL2/deficiency , Cranial Irradiation/adverse effects , Female , Magnetic Resonance Imaging , Male , Mice , Radiation Injuries, Experimental/diagnostic imaging , Radiation Injuries, Experimental/metabolism
2.
PLoS One ; 16(3): e0249142, 2021.
Article in English | MEDLINE | ID: mdl-33760886

ABSTRACT

CCR2 is the major chemokine receptor that regulates appropriate trafficking of inflammatory monocytes, but the role of this chemokine receptor and its ligands during primary and secondary infection with intracellular infections remains incompletely understood. Here we used murine infection with the Live Vaccine Strain (LVS) of Francisella tularensis to evaluate the role of CCR2 during primary and secondary parenteral responses to this prototype intracellular bacterium. We find that mice deficient in CCR2 are highly compromised in their ability to survive intradermal infection with LVS, indicating the importance of this receptor during primary parenteral responses. Interestingly, this defect could not be readily attributed to the activities of the known murine CCR2 ligands MCP-1/CCL2, MCP-3/CCL7, or MCP-5/CCL12. Nonetheless, CCR2 knockout mice vaccinated by infection with low doses of LVS generated optimal T cell responses that controlled the intramacrophage replication of Francisella, and LVS-immune CCR2 knockout mice survived maximal lethal Francisella challenge. Thus, fully protective adaptive immune memory responses to this intracellular bacterium can be readily generated in the absence of CCR2.


Subject(s)
Francisella tularensis/physiology , Receptors, CCR2/genetics , Tularemia/immunology , Animals , Bacterial Vaccines/administration & dosage , Bacterial Vaccines/immunology , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Chemokine CCL7/deficiency , Chemokine CCL7/genetics , Chemokine CCL7/immunology , Disease Models, Animal , Disease Susceptibility , Francisella tularensis/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , Monocytes/metabolism , Receptors, CCR2/deficiency , Survival Rate , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Tularemia/mortality , Tularemia/pathology , Tularemia/prevention & control , Vaccines, Attenuated/administration & dosage , Vaccines, Attenuated/immunology
3.
Int J Mol Sci ; 22(3)2021 Feb 02.
Article in English | MEDLINE | ID: mdl-33540898

ABSTRACT

The mechanisms of how obesity contributes to the development of cardio-metabolic diseases are not entirely understood. Obesity is frequently associated with adipose tissue dysfunction, characterized by, e.g., adipocyte hypertrophy, ectopic fat accumulation, immune cell infiltration, and the altered secretion of adipokines. Factors secreted from adipose tissue may induce and/or maintain a local and systemic low-grade activation of the innate immune system. Attraction of macrophages into adipose tissue and altered crosstalk between macrophages, adipocytes, and other cells of adipose tissue are symptoms of metabolic inflammation. Among several secreted factors attracting immune cells to adipose tissue, chemotactic C-C motif chemokine ligand 2 (CCL2) (also described as monocyte chemoattractant protein-1 (MCP-1)) has been shown to play a crucial role in adipose tissue macrophage infiltration. In this review, we aimed to summarize and discuss the current knowledge on CCL2 with a focus on its role in linking obesity to cardio-metabolic diseases.


Subject(s)
Chemokine CCL2/physiology , Inflammation/complications , Obesity/etiology , Adipocytes/physiology , Adipose Tissue/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , Cardiovascular Diseases/genetics , Cardiovascular Diseases/metabolism , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokines/metabolism , Humans , Inflammation/genetics , Inflammation/physiopathology , Insulin Resistance , Macrophages/physiology , Mesenchymal Stem Cells/metabolism , Metabolic Syndrome/genetics , Metabolic Syndrome/metabolism , Mice , Mice, Knockout , Models, Animal , Molecular Targeted Therapy , Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Neural Tube Defects/genetics , Neural Tube Defects/metabolism , Obesity/genetics , Obesity/physiopathology , Polymorphism, Single Nucleotide , Signal Transduction
4.
Proc Natl Acad Sci U S A ; 118(6)2021 02 09.
Article in English | MEDLINE | ID: mdl-33536334

ABSTRACT

Macrophages are intimately involved in the pathophysiology of endometriosis, a chronic inflammatory disorder characterized by the growth of endometrial-like tissue (lesions) outside the uterus. By combining genetic and pharmacological monocyte and macrophage depletion strategies we determined the ontogeny and function of macrophages in a mouse model of induced endometriosis. We demonstrate that lesion-resident macrophages are derived from eutopic endometrial tissue, infiltrating large peritoneal macrophages (LpM) and monocytes. Furthermore, we found endometriosis to trigger continuous recruitment of monocytes and expansion of CCR2+ LpM. Depletion of eutopic endometrial macrophages results in smaller endometriosis lesions, whereas constitutive inhibition of monocyte recruitment significantly reduces peritoneal macrophage populations and increases the number of lesions. Reprogramming the ontogeny of peritoneal macrophages such that embryo-derived LpM are replaced by monocyte-derived LpM decreases the number of lesions that develop. We propose a putative model whereby endometrial macrophages are "proendometriosis" while newly recruited monocyte-derived macrophages, possibly in LpM form, are "antiendometriosis." These observations highlight the importance of monocyte-derived macrophages in limiting disease progression.


Subject(s)
Endometriosis/pathology , Macrophages, Peritoneal/pathology , Animals , Antibodies, Monoclonal/metabolism , Chemokine CCL2/deficiency , Chemokine CCL2/metabolism , Endometrium/pathology , Female , Mice, Inbred C57BL , Models, Biological , Monocytes/pathology , Peritoneal Cavity/pathology
5.
Cell ; 178(5): 1102-1114.e17, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442403

ABSTRACT

Caloric restriction is known to improve inflammatory and autoimmune diseases. However, the mechanisms by which reduced caloric intake modulates inflammation are poorly understood. Here we show that short-term fasting reduced monocyte metabolic and inflammatory activity and drastically reduced the number of circulating monocytes. Regulation of peripheral monocyte numbers was dependent on dietary glucose and protein levels. Specifically, we found that activation of the low-energy sensor 5'-AMP-activated protein kinase (AMPK) in hepatocytes and suppression of systemic CCL2 production by peroxisome proliferator-activator receptor alpha (PPARα) reduced monocyte mobilization from the bone marrow. Importantly, we show that fasting improves chronic inflammatory diseases without compromising monocyte emergency mobilization during acute infectious inflammation and tissue repair. These results reveal that caloric intake and liver energy sensors dictate the blood and tissue immune tone and link dietary habits to inflammatory disease outcome.


Subject(s)
Caloric Restriction , Monocytes/metabolism , AMP-Activated Protein Kinases/metabolism , Adult , Animals , Antigens, Ly/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Female , Hepatocytes/cytology , Hepatocytes/metabolism , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/cytology , PPAR alpha/deficiency , PPAR alpha/genetics , PPAR alpha/metabolism
6.
PLoS Pathog ; 15(7): e1007847, 2019 07.
Article in English | MEDLINE | ID: mdl-31306468

ABSTRACT

Salmonella exploit host-derived nitrate for growth in the lumen of the inflamed intestine. The generation of host-derived nitrate is dependent on Nos2, which encodes inducible nitric oxide synthase (iNOS), an enzyme that catalyzes nitric oxide (NO) production. However, the cellular sources of iNOS and, therefore, NO-derived nitrate used by Salmonella for growth in the lumen of the inflamed intestine remain unidentified. Here, we show that iNOS-producing inflammatory monocytes infiltrate ceca of mice infected with Salmonella. In addition, we show that inactivation of type-three secretion system (T3SS)-1 and T3SS-2 renders Salmonella unable to induce CC- chemokine receptor-2- and CC-chemokine ligand-2-dependent inflammatory monocyte recruitment. Furthermore, we show that the severity of the pathology of Salmonella- induced colitis as well as the nitrate-dependent growth of Salmonella in the lumen of the inflamed intestine are reduced in mice that lack Ccr2 and, therefore, inflammatory monocytes in the tissues. Thus, inflammatory monocytes provide a niche for Salmonella expansion in the lumen of the inflamed intestine.


Subject(s)
Intestinal Mucosa/metabolism , Intestinal Mucosa/microbiology , Monocytes/metabolism , Salmonella typhimurium/metabolism , Salmonella typhimurium/pathogenicity , Animals , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Female , Host Microbial Interactions/genetics , Host Microbial Interactions/physiology , Humans , Inflammation/metabolism , Inflammation/microbiology , Inflammation/pathology , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Monocytes/pathology , Nitric Oxide Synthase Type II/metabolism , Receptors, CCR2/deficiency , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Salmonella Infections, Animal/metabolism , Salmonella Infections, Animal/microbiology , Salmonella Infections, Animal/pathology , Salmonella typhimurium/genetics , Type III Secretion Systems/metabolism
7.
Am J Physiol Renal Physiol ; 317(2): F343-F360, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31091126

ABSTRACT

Polycystic kidney disease (PKD) is characterized by slowly expanding renal cysts that damage the kidney, typically resulting in renal failure by the fifth decade. The most common cause of death in these patients, however, is cardiovascular disease. Expanding cysts in PKD induce chronic kidney injury that is accompanied by immune cell infiltration, including macrophages, which we and others have shown can promote disease progression in PKD mouse models. Here, we show that monocyte chemoattractant protein-1 [MCP-1/chemokine (C-C motif) ligand 2 (CCL2)] is responsible for the majority of monocyte chemoattractant activity produced by renal PKD cells from both mice and humans. To test whether the absence of MCP-1 lowers renal macrophage concentration and slows disease progression, we generated genetic knockout (KO) of MCP-1 in a mouse model of PKD [congenital polycystic kidney (cpk) mice]. Cpk mice are born with rapidly expanding renal cysts, accompanied by a decline in kidney function and death by postnatal day 21. Here, we report that KO of MCP-1 in these mice increased survival, with some mice living past 3 mo. Surprisingly, however, there was no significant difference in renal macrophage concentration, nor was there improvement in cystic disease or kidney function. Examination of mice revealed cardiac hypertrophy in cpk mice, and measurement of cardiac electrical activity via ECG revealed repolarization abnormalities. MCP-1 KO did not affect the number of cardiac macrophages, nor did it alleviate the cardiac aberrancies. However, MCP-1 KO did prevent the development of pulmonary edema, which occurred in cpk mice, and promoted decreased resting heart rate and increased heart rate variability in both cpk and noncystic mice. These data suggest that in this mouse model of PKD, MCP-1 altered cardiac/pulmonary function and promoted death outside of its role as a macrophage chemoattractant.


Subject(s)
Arrhythmias, Cardiac/metabolism , Cardiomegaly/metabolism , Chemokine CCL2/metabolism , Kidney/metabolism , Lung/metabolism , Myocardium/metabolism , Polycystic Kidney Diseases/metabolism , Pulmonary Edema/metabolism , Animals , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Cells, Cultured , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Disease Models, Animal , Disease Progression , Fibrosis , Humans , Inflammation Mediators/metabolism , Kidney/pathology , Kidney/physiopathology , Lung/pathology , Lung/physiopathology , Macrophages/metabolism , Macrophages/pathology , Membrane Proteins/genetics , Mice, Inbred C57BL , Mice, Knockout , Myocardium/pathology , Polycystic Kidney Diseases/pathology , Polycystic Kidney Diseases/physiopathology , Pulmonary Edema/pathology , Pulmonary Edema/physiopathology , Pulmonary Edema/prevention & control , Time Factors
8.
J Endocrinol ; 242(2): 91-101, 2019 08.
Article in English | MEDLINE | ID: mdl-31137011

ABSTRACT

Obesity is strongly associated with chronic inflammation for which adipose tissue macrophages play a critical role. The objective of this study is to identify monocyte chemoattractant protein-1 (MCP-1, CCL2) as a key player governing M1-M2 macrophage polarization and energy balance. We evaluated body weight, fat mass, adipocyte size and energy expenditure as well as core body temperature of Ccl2 knockout mice compared with wild-type mice. Adipose tissues, differentiated adipocyte and bone marrow-derived macrophages were assessed by qPCR, Western blot analysis and histochemistry. MCP-1 deficiency augmented energy expenditure by promoting browning in white adipose tissue and brown adipose tissue activity via increasing the expressions of Ucp1, Prdm16, Tnfrsf9, Ppargc1a, Nrf1 and Th and mitochondrial DNA copy number. MCP-1 abrogation promoted M2 polarization which is characterized by increased expression of Arg1, Chil3, Il10 and Klf4 whereas it decreased M1 polarization by decreased p65 nuclear translocation and attenuated expression of Itgax, Tnf and Nos2, leading to increased browning of adipocytes. Enhanced M2 polarization and attenuated M1 polarization in the absence of MCP-1 are independent. Collectively, our results suggest that the action of MCP-1 in macrophages modulates energy expenditure by impairing browning in adipose tissue.


Subject(s)
Adipose Tissue, Brown/metabolism , Chemokine CCL2/genetics , Energy Metabolism/genetics , Macrophages/metabolism , Adipocytes/metabolism , Animals , Chemokine CCL2/deficiency , Gene Expression , Kruppel-Like Factor 4 , Macrophage Activation/genetics , Macrophages/classification , Mice, Inbred C57BL , Mice, Knockout , Obesity/genetics , Obesity/metabolism , Uncoupling Protein 1/genetics , Uncoupling Protein 1/metabolism
9.
Anticancer Res ; 39(4): 1729-1738, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30952712

ABSTRACT

AIM: Monocyte chemotactic protein-1 (MCP1) is a potent adipokine. This study tested the hypothesis that adipose-produced MCP1 contributes to metastasis. MATERIALS AND METHODS: In a spontaneous metastasis model of Lewis lung carcinoma (LLC), male adipose MCP1-deficient (Mcp1-/-) and wild-type (WT) mice were fed the AIN93G diet or a high-fat diet (HFD) for 11 weeks. Lung metastasis from a subcutaneous tumor was the primary endpoint. RESULTS: The adipose expression of MCP1 was lower in Mcp1-/- mice than in WT controls. The HFD increased the number of lung metastases in WT mice. The number of metastasis was significantly lower in the HFD-fed Mcp1-/- mice than in the HFD-fed WT mice. Compared to the WT mice, adipose MCP1 deficiency lowered plasma concentrations of insulin, proinflammatory adipokines (leptin, plasminogen activator inhibitor-1, and resistin), and angiogenic markers (vascular endothelial growth factor, hepatocyte growth factor, and angiopoietin-2). CONCLUSION: Adipose MCP1 deficiency attenuates HFD-enhanced pulmonary metastasis of LLC.


Subject(s)
Adipose Tissue/metabolism , Carcinoma, Lewis Lung/metabolism , Carcinoma, Lewis Lung/secondary , Chemokine CCL2/deficiency , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Adipokines/metabolism , Adiposity , Angiogenic Proteins/metabolism , Animals , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/prevention & control , Chemokine CCL2/genetics , Diet, High-Fat , Inflammation Mediators/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Neoplasm Invasiveness , Signal Transduction
10.
Biochim Biophys Acta Mol Basis Dis ; 1865(6): 1555-1566, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30905786

ABSTRACT

The risk of non-alcoholic fatty liver disease increases with obesity. Vulnerability to oxidative stress and/or inflammation represents a crucial step in non-alcoholic fatty liver disease progression through abnormal metabolic responses. In this study, we investigated the role of CCL2 gene ablation in mice that were double deficient in low density lipoprotein receptor and in paraoxonase-1. Mass spectrometry methods were used to assess the liver metabolic response in mice fed either regular chow or a high-fat diet. Dietary fat caused liver steatosis, oxidative stress and the accumulation of pro-inflammatory macrophages in the livers of double deficient mice. We observed alterations in energy metabolism-related pathways and in metabolites associated with the methionine cycle and the glutathione reduction pathway. This metabolic response was associated with impaired autophagy. Conversely, when we established CCL2 deficiency, histologic features of fatty liver disease were abrogated, hepatic liver oxidative stress decreased, and anti-inflammatory macrophage marker expression levels increased. These changes were associated with the normalization of metabolic disturbances and increased lysosome-associated membrane protein 2, expression, which suggests enhanced chaperone-mediated autophagy. This study demonstrates that CCL2 is a key molecule for the development of metabolic and histological alterations in the liver of mice sensitive to the development of hyperlipidemia and hepatic steatosis, a finding with potential to identify new therapeutic targets in liver diseases.


Subject(s)
Aryldialkylphosphatase/genetics , Chemokine CCL2/genetics , Hyperlipidemias/genetics , Lipoproteins, LDL/metabolism , Non-alcoholic Fatty Liver Disease/genetics , Receptors, LDL/genetics , Animals , Aryldialkylphosphatase/deficiency , Autophagy/genetics , Chemokine CCL2/deficiency , Diet, High-Fat/adverse effects , Energy Metabolism/genetics , Gene Expression Regulation , Glutathione/metabolism , Hyperlipidemias/etiology , Hyperlipidemias/metabolism , Hyperlipidemias/pathology , Liver/metabolism , Liver/pathology , Lysosomal-Associated Membrane Protein 2/genetics , Lysosomal-Associated Membrane Protein 2/metabolism , Macrophages/metabolism , Macrophages/pathology , Male , Metabolome/genetics , Methionine/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Oxidative Stress , Receptors, LDL/deficiency , Signal Transduction
11.
J Ocul Pharmacol Ther ; 35(2): 98-105, 2019 03.
Article in English | MEDLINE | ID: mdl-30547709

ABSTRACT

PURPOSE: The purpose of these studies was (1) to investigate the ability of human M1 phenotype macrophages to secrete vascular endothelial growth factor (VEGF) and the influence of prostacyclin receptor (IP) stimulation (2) to evaluate the contribution of the proangiogenic prostanoid prostacyclin to experimental choroidal neovascularization Methods: Human macrophages derived from primary blood mononuclear cells were functionally biased toward the M1 phenotype by using tumor necrosis factor α (TNFα). Experimental choroidal neovascularization was produced by laser photocoagulation. Antagonist drugs RO-3244794 (IP antagonist) and GW 627368 (EP4 antagonist) were administered according to an optimal dosing regimen that was predetermined by bioavailability studies. RESULTS: IP receptor stimulation had diametrically opposed effects on VEGF release compared with reported data on cytokine/chemokine secretion from human macrophages. For example, the IP agonist cicaprost stimulated VEGF secretion although it inhibits monocyte chemoattractant protein-1 (MCP-1) secretion: both would favor a proangiogenic effect. The IP receptor antagonist RO-3244794 produced an ∼20% statistically significant reduction in the neovascularized lesion area in the choroidal neovascularization model, which was a similar level to that produced by the EP4 antagonist GW 627368. Combining the 2 drugs produced a statistically significant reduction in neovascularization but only of slightly greater magnitude than that obtained with each antagonist administered alone. CONCLUSIONS: IP receptor stimulation potently and highly efficaciously promoted VEGF release from human M1 macrophages, indicating a possible contribution of the M1 macrophage subtype to VEGF-induced choroidal neovascularization. Studies in living animals suggest that prostacyclin and its target IP receptor contribute to choroidal neovascularization, although to a more modest extent than might have been expected.


Subject(s)
Antihypertensive Agents/pharmacology , Choroidal Neovascularization/drug therapy , Epoprostenol/pharmacology , Macrophages/drug effects , Ophthalmic Solutions/pharmacology , Animals , Cells, Cultured , Chemokine CCL2/analysis , Chemokine CCL2/deficiency , Chemokine CCL2/metabolism , Choroidal Neovascularization/metabolism , Choroidal Neovascularization/pathology , Disease Models, Animal , Humans , Lipopolysaccharides/pharmacology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Knockout , Rats , Rats, Sprague-Dawley , Vascular Endothelial Growth Factors/analysis , Vascular Endothelial Growth Factors/metabolism
12.
J Am Soc Nephrol ; 29(10): 2471-2481, 2018 10.
Article in English | MEDLINE | ID: mdl-30209078

ABSTRACT

BACKGROUND: In patients with autosomal dominant polycystic kidney disease (ADPKD), most of whom have a mutation in PKD1 or PKD2, abnormally large numbers of macrophages accumulate around kidney cysts and promote their growth. Research by us and others has suggested that monocyte chemoattractant protein-1 (Mcp1) may be a signal for macrophage-mediated cyst growth. METHODS: To define the role of Mcp1 and macrophages in promoting cyst growth, we used mice with inducible knockout of Pkd1 alone (single knockout) or knockout of both Pkd1 and Mcp1 (double knockout) in the murine renal tubule. Levels of Mcp1 RNA expression were measured in single-knockout mice and controls. RESULTS: In single-knockout mice, upregulation of Mcp1 precedes macrophage infiltration. Macrophages accumulating around nascent cysts (0-2 weeks after induction) are initially proinflammatory and induce tubular cell injury with morphologic flattening, oxidative DNA damage, and proliferation-independent cystic dilation. At 2-6 weeks after induction, macrophages switch to an alternative activation phenotype and promote further cyst growth because of an additional three-fold increase in tubular cell proliferative rates. In double-knockout mice, there is a marked reduction in Mcp1 expression and macrophage numbers, resulting in less initial tubular cell injury, slower cyst growth, and improved renal function. Treatment of single-knockout mice with an inhibitor to the Mcp1 receptor Ccr2 partially reproduced the morphologic and functional improvement seen with Mcp1 knockout. CONCLUSIONS: Mcp1 is upregulated after knockout of Pkd1 and promotes macrophage accumulation and cyst growth via both proliferation-independent and proliferation-dependent mechanisms in this orthologous mouse model of ADPKD.


Subject(s)
Chemokine CCL2/genetics , Chemokine CCL2/physiology , Macrophages/physiology , Polycystic Kidney, Autosomal Dominant/genetics , Polycystic Kidney, Autosomal Dominant/pathology , Animals , Chemokine CCL2/deficiency , DNA Damage , Disease Models, Animal , Humans , Kidney Tubules/pathology , Kidney Tubules/physiopathology , Macrophage Activation/drug effects , Macrophage Activation/genetics , Macrophage Activation/physiology , Macrophages/drug effects , Macrophages/pathology , Male , Mice , Mice, Knockout , Polycystic Kidney, Autosomal Dominant/physiopathology , Pyrrolidines/pharmacology , Reactive Oxygen Species/metabolism , Receptors, CCR2/antagonists & inhibitors , TRPP Cation Channels/deficiency , TRPP Cation Channels/genetics , Up-Regulation
13.
Sci Rep ; 8(1): 8598, 2018 06 05.
Article in English | MEDLINE | ID: mdl-29872089

ABSTRACT

Inflammation plays an important role in the pathogenesis of renal and cardiovascular disease in renovascular hypertension (RVH). Ccl2 is an important mediator of inflammation, and is induced within 24 hours following surgery to establish RVH in the murine 2 kidney 1 clip model, a time prior to onset of interstitial inflammation, fibrosis, or tubular atrophy. We tested the hypothesis that Ccl2 deficiency protects the stenotic kidney (STK) from development of chronic renal damage in mice with renovascular hypertension due to renal artery stenosis (RAS). RAS surgery was performed on wild type (WT) and Ccl2 knock out (KO) mice; animals were studied for four weeks. Renal blood flow was reduced to similar extent in both WT and Ccl2 KO mice with RVH. Perfusion of the stenotic kidney was significantly reduced in Ccl2 KO mice as assessed by magnetic resonance imaging (MRI). Stenotic kidney volume in WT, but not in Ccl2 KO mice, was significantly reduced following surgery. Cortical hypoxia was observed in the stenotic kidney of Ccl2 KO mice, as assessed by blood oxygen level-dependent MRI (BOLD-MRI). Ccl2 KO mice showed less cortical atrophy than WT RAS mice. Ccl2 deficiency reduced the number of infiltrating mononuclear cells and expression of Ccl5, Ccl7, Ccl8, Ccr2 and Cd206. We conclude that Ccl2 is a critical mediator of chronic renal injury in RVH.


Subject(s)
Chemokine CCL2/metabolism , Hypertension, Renovascular/complications , Renal Insufficiency, Chronic/pathology , Renal Insufficiency, Chronic/physiopathology , Animals , Atrophy/pathology , Chemokine CCL2/deficiency , Disease Models, Animal , Hypoxia/pathology , Magnetic Resonance Imaging , Mice , Mice, Knockout , Renal Circulation
14.
APMIS ; 125(11): 945-956, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28836736

ABSTRACT

CCL2 is a chemokine that can be induced during neuroinflammation to recruit immune cells, but its role in the central nervous system (CNS) is unclear. Our aim was to better understand its role. We induced CCL2 in CNS of naive CCL2-deficient mice using intrathecally administered replication-defective adenovirus and examined cell infiltration by flow cytometry. CCL2 expression induced pronounced and unexpected recruitment of regulatory and IFNγ-producing T cells to CNS from blood, possibly related to defective egress of monocytes from CCL2-deficient bone marrow. Infiltration also occurred in mice lacking CCR2, a receptor for CCL2. Expression of another receptor for CCL2, CCR4, and CXCR3, a receptor for CXCL10, which was also induced, were both increased in CCL2-treated CNS. CCR4 was expressed by neurons and astrocytes as well as CD4 T cells, and CXCR3 was expressed by CD4 and CD8 T cells. Chemokine-recruited T cells did not lead to CNS pathology. Our findings show a role for CCL2 in recruitment of CD4 T cells to the CNS and show that redundancy among chemokine receptors ensures optimal response.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Central Nervous System/immunology , Chemokine CCL2/immunology , Receptors, CCR2/immunology , Adenoviridae/genetics , Adenoviridae/metabolism , Animals , Astrocytes/cytology , Astrocytes/immunology , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Movement , Central Nervous System/cytology , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Female , Gene Expression Regulation , Genes, Reporter , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Injections, Spinal , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Mice, Knockout , Neurons/cytology , Neurons/immunology , Receptors, CCR2/genetics , Receptors, CCR4/genetics , Receptors, CCR4/immunology , Receptors, CXCR3/genetics , Receptors, CXCR3/immunology , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Signal Transduction , Red Fluorescent Protein
15.
Cytokine ; 97: 108-116, 2017 09.
Article in English | MEDLINE | ID: mdl-28628889

ABSTRACT

Neonates have greater morbidity/mortality from lower respiratory tract infections (LRTI) compared to older children. Lack of conditioning of the pulmonary immune system due to limited environmental exposures and/or infectious challenges likely contributes to the increase susceptibility in the neonate. In this study, we sought to gain insights into the nature and dynamics of the neonatal pulmonary immune response to LRTI using a murine model. METHODS: Wildtype (WT) and Ccr2-/- C57BL/6 neonatal and juvenile mice received E. coli or PBS by direct pharyngeal aspiration. Flow cytometry was used to measure immune cell dynamics and identify cytokine-producing cells. Real-time PCR and ELISA were used to measure cytokine/chemokine expression. RESULTS: Innate immune cell recruitment in response to E. coli-induced LRTI was delayed in the neonatal lung compared to juvenile lung. Lung clearance of bacteria was also significantly delayed in the neonate. Ccr2-/- neonates, which lack an intact CCL2-CCR2 axis, had higher mortality after E. coli challenged than Ccr2+/+ neonates. A greater percentage of CD8+ T cells and monocytes from WT neonates challenged with E. coli produced TNF compared to controls. CONCLUSION: The pulmonary immune response to E. coli-induced LRTI differed significantly between neonatal and juvenile mice. Neonates were more susceptible to increasing doses of E. coli and exhibited greater mortality than juveniles. In the absence of an intact CCL2-CCR2 axis, susceptibility to LRTI-induced mortality was further increased in neonatal mice. Taken together these findings underscore the importance of age-related differences in the innate immune response to LRTI during early stages of postnatal life.


Subject(s)
Chemokine CCL2/immunology , Escherichia coli Infections/immunology , Immunity, Innate , Lung/microbiology , Receptors, CCR2/metabolism , Respiratory Tract Infections/immunology , Age Factors , Animals , Animals, Newborn , Bronchi/microbiology , Chemokine CCL2/deficiency , Chemokines/immunology , Cytokines/immunology , Disease Models, Animal , Escherichia coli , Escherichia coli Infections/microbiology , Escherichia coli Infections/mortality , Inflammation , Lung/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Pneumonia, Bacterial/immunology , Pneumonia, Bacterial/microbiology , Real-Time Polymerase Chain Reaction , Receptors, CCR2/deficiency
16.
Oncotarget ; 8(14): 23303-23311, 2017 Apr 04.
Article in English | MEDLINE | ID: mdl-28177896

ABSTRACT

Bone loss occurs in obesity and cancer-associated complications including wasting. This study determined whether a high-fat diet and a deficiency in monocyte chemotactic protein-1 (MCP-1) altered bone structural defects in male C57BL/6 mice with Lewis lung carcinoma (LLC) metastases in lungs. Compared to non-tumor-bearing mice, LLC reduced bone volume fraction, connectivity density, trabecular number, trabecular thickness and bone mineral density and increased trabecular separation in femurs. Similar changes occurred in vertebrae. The high-fat diet compared to the AIN93G diet exacerbated LLC-induced detrimental structural changes; the exacerbation was greater in femurs than in vertebrae. Mice deficient in MCP-1 compared to wild-type mice exhibited increases in bone volume fraction, connectivity density, trabecular number and decreases in trabecular separation in both femurs and vertebrae, and increases in trabecular thickness and bone mineral density and a decrease in structure model index in vertebrae. Lewis lung carcinoma significantly decreased osteocalcin but increased tartrate-resistant acid phosphatase 5b (TRAP 5b) in plasma. In LLC-bearing mice, the high-fat diet increased and MCP-1 deficiency decreased plasma TRAP 5b; neither the high-fat diet nor MCP-1 deficiency resulted in significant changes in plasma concentration of osteocalcin. In conclusion, pulmonary metastasis of LLC is accompanied by detrimental bone structural changes; MCP-1 deficiency attenuates and high-fat diet exacerbates the metastasis-associated bone wasting.


Subject(s)
Bone and Bones/pathology , Carcinoma, Lewis Lung/pathology , Chemokine CCL2/deficiency , Animals , Bone and Bones/metabolism , Carcinoma, Lewis Lung/blood , Carcinoma, Lewis Lung/genetics , Carcinoma, Lewis Lung/metabolism , Chemokine CCL2/metabolism , Diet, High-Fat/adverse effects , Male , Mice , Mice, Inbred C57BL , Neoplasm Metastasis , Osteocalcin/blood , Tartrate-Resistant Acid Phosphatase/blood
17.
Sci Rep ; 7: 41258, 2017 01 24.
Article in English | MEDLINE | ID: mdl-28117437

ABSTRACT

Systemic lupus erythematosus (SLE) is a multi-organ autoimmune disease characterized by autoantibody production. Mesenchymal stem cells (MSCs) ameliorate SLE symptoms by targeting T cells, whereas the mechanisms of their efficacy remain incompletely understood. In this study, we show that transfer of human MSCs increased MRL.Faslpr mouse survival, decreased T cell infiltration in the kidneys, and reduced T cell cytokine expression. In vitro, allogeneic mouse MSCs inhibited MRL.Faslpr T cell proliferation and cytokine production. Time-lapse imaging revealed that MSCs recruited MRL.Faslpr T cells establishing long-lasting cellular contacts by enhancing T cell VCAM-1 expression in a CCL2-dependent manner. In contrast, CCL2 deficient MSCs did not induce T cell migration and VCAM-1 expression, resulting in insufficient cell-cell contact. Consequently, CCL2 deficient MSCs did not inhibit IFN-γ production by T cells and upon transfer no longer prolonged survival of MRL.Faslpr mice. Taken together, our imaging study demonstrates that CCL2 enables the prolonged MSC-T cell interactions needed for sufficient suppression of autoreactive T cells and helps to understand how MSCs ameliorate symptoms in lupus-prone MRL.Faslpr mice.


Subject(s)
Cell Communication , Chemokine CCL2/deficiency , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Mesenchymal Stem Cells/metabolism , T-Lymphocytes/metabolism , Animals , Cell Movement , Humans , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Solubility , Vascular Cell Adhesion Molecule-1/metabolism
18.
PLoS One ; 11(11): e0165595, 2016.
Article in English | MEDLINE | ID: mdl-27820834

ABSTRACT

Chronic inflammation is a hallmark of cancer. Inflammatory chemokines, such as C-C chemokine ligand 2 (CCL2), are often present in tumors but their roles in cancer initiation and maintenance are not clear. Here we report that CCL2 promotes mammary carcinoma development in a clinically relevant murine model of breast cancer. Targeted disruption of Ccl2 slowed the growth of activated Her2/neu-driven mammary tumors and prolonged host survival. Disruption of Ccl2 was associated with a decrease in the development and mobilization of endothelial precursor cells (EPCs) which can contribute to tumor neovascularization. In contrast, disruption of Ccr2, which encodes CCL2's sole signaling receptor, accelerated tumor development, shortened host survival, and mobilized EPCs. However, pharmacological inhibition of CCR2 phenocopied Ccl2 disruption rather than Ccr2 disruption, suggesting that the Ccr2-/- phenotype is a consequence of unanticipated alterations not linked to intact CCL2/CCR2 signaling. Consistent with this explanation, Ccr2-/- monocytes are more divergent from wild type monocytes than Ccl2-/- monocytes in their expression of genes involved in key developmental and functional pathways. Taken together, our data suggest a tumor-promoting role for CCL2 acting through CCR2 on the tumor microenvironment and support the targeting of this chemokine/receptor pair in breast cancer.


Subject(s)
Chemokine CCL2/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Receptor, ErbB-2/metabolism , Receptors, CCR2/metabolism , Tumor Microenvironment , Animals , Cell Line, Tumor , Chemokine CCL2/deficiency , Chemokine CCL2/genetics , Endothelial Progenitor Cells/pathology , Female , Gene Deletion , Humans , Mammary Neoplasms, Experimental/genetics , Mice , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/deficiency , Receptors, CCR2/genetics
19.
EMBO Rep ; 17(12): 1738-1752, 2016 12.
Article in English | MEDLINE | ID: mdl-27733491

ABSTRACT

Sickness behavior defines the endocrine, autonomic, behavioral, and metabolic responses associated with infection. While inflammatory responses were suggested to be instrumental in the loss of appetite and body weight, the molecular underpinning remains unknown. Here, we show that systemic or central lipopolysaccharide (LPS) injection results in specific hypothalamic changes characterized by a precocious increase in the chemokine ligand 2 (CCL2) followed by an increase in pro-inflammatory cytokines and a decrease in the orexigenic neuropeptide melanin-concentrating hormone (MCH). We therefore hypothesized that CCL2 could be the central relay for the loss in body weight induced by the inflammatory signal LPS. We find that central delivery of CCL2 promotes neuroinflammation and the decrease in MCH and body weight. MCH neurons express CCL2 receptor and respond to CCL2 by decreasing both electrical activity and MCH release. Pharmacological or genetic inhibition of CCL2 signaling opposes the response to LPS at both molecular and physiologic levels. We conclude that CCL2 signaling onto MCH neurons represents a core mechanism that relays peripheral inflammation to sickness behavior.


Subject(s)
Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Inflammation/metabolism , Melanins/metabolism , Neurons/metabolism , Pituitary Hormones/metabolism , Signal Transduction , Animals , Chemokine CCL2/deficiency , Chemokine CCL2/immunology , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/immunology , Hypothalamic Hormones/genetics , Hypothalamic Hormones/immunology , Illness Behavior , Lipopolysaccharides/immunology , Melanins/genetics , Melanins/immunology , Mice , Neurons/immunology , Pituitary Hormones/genetics , Pituitary Hormones/immunology , Receptors, CCR2/metabolism , Weight Loss
20.
Oncotarget ; 7(17): 24792-9, 2016 Apr 26.
Article in English | MEDLINE | ID: mdl-27028862

ABSTRACT

Adipose-produced pro-inflammatory cytokines contribute to obesity and cancer. This 2x2 experiment was designed to investigate effects of monocyte chemotactic protein-1 (MCP-1) deficiency on pulmonary metastasis of Lewis lung carcinoma (LLC) in MCP-1 deficient and wild-type mice fed a modified AIN93G diet containing 16% and 45% of energy from corn oil, respectively. The high-fat diet significantly increased the number and size (cross-sectional area and volume) of lung metastases compared to the AIN93G control diet. Deficiency in MCP-1 reduced lung metastases by 37% in high-fat diet-fed mice; it reduced metastatic cross-sectional area by 46% and volume by 69% compared to wild-type mice. Adipose and plasma concentrations of MCP-1 were significantly higher in high-fat diet-fed wild-type mice than in their AIN93G-fed counterparts; they were not detectable in MCP-1 deficient mice regardless of diet. Plasma concentrations of plasminogen activator inhibitor-1, tumor necrosis factor-α, vascular endothelial growth factor and tissue inhibitor of metalloproteinase-1 were significantly higher in MCP-1 deficient mice compared to wild-type mice. We conclude that adipose-produced MCP-1 contributes to high-fat diet-enhanced metastasis. While MCP-1 deficiency reduces metastasis, the elevation of pro-inflammatory cytokines and angiogenic factors in the absence of MCP-1 may support the metastatic development and growth of LLC in MCP-1 deficient mice.


Subject(s)
Carcinoma, Lewis Lung/genetics , Chemokine CCL2/deficiency , Animals , Carcinoma, Lewis Lung/pathology , Chemokine CCL2/metabolism , Diet, High-Fat , Humans , Mice , Mice, Inbred C57BL , Neoplasm Metastasis
SELECTION OF CITATIONS
SEARCH DETAIL
...