Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 177
Filter
Add more filters

Publication year range
1.
J Pathol ; 2024 Jul 26.
Article in English | MEDLINE | ID: mdl-39056146

ABSTRACT

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

2.
Int J Mol Sci ; 24(4)2023 Feb 05.
Article in English | MEDLINE | ID: mdl-36834561

ABSTRACT

Fibrosis and structural remodeling of the lung tissue can significantly impair lung function, often with fatal consequences. The etiology of pulmonary fibrosis (PF) is diverse and includes different triggers such as allergens, chemicals, radiation, and environmental particles. However, the cause of idiopathic PF (IPF), one of the most common forms of PF, remains unknown. Experimental models have been developed to study the mechanisms of PF, and the murine bleomycin (BLM) model has received the most attention. Epithelial injury, inflammation, epithelial-mesenchymal transition (EMT), myofibroblast activation, and repeated tissue injury are important initiators of fibrosis. In this review, we examined the common mechanisms of lung wound-healing responses after BLM-induced lung injury as well as the pathogenesis of the most common PF. A three-stage model of wound repair involving injury, inflammation, and repair is outlined. Dysregulation of one or more of these three phases has been reported in many cases of PF. We reviewed the literature investigating PF pathogenesis, and the role of cytokines, chemokines, growth factors, and matrix feeding in an animal model of BLM-induced PF.


Subject(s)
Idiopathic Pulmonary Fibrosis , Lung Injury , Mice , Animals , Bleomycin , Lung/pathology , Fibrosis , Inflammation/pathology , Idiopathic Pulmonary Fibrosis/pathology , Lung Injury/pathology , Epithelial-Mesenchymal Transition , Disease Models, Animal
3.
Int J Mol Sci ; 24(9)2023 Apr 25.
Article in English | MEDLINE | ID: mdl-37175553

ABSTRACT

Intense neutrophil infiltration into the liver is a characteristic of acetaminophen-induced acute liver injury. Neutrophil elastase is released by neutrophils during inflammation. To elucidate the involvement of neutrophil elastase in acetaminophen-induced liver injury, we investigated the efficacy of a potent and specific neutrophil elastase inhibitor, sivelestat, in mice with acetaminophen-induced acute liver injury. Intraperitoneal administration of 750 mg/kg of acetaminophen caused severe liver damage, such as elevated serum transaminase levels, centrilobular hepatic necrosis, and neutrophil infiltration, with approximately 50% mortality in BALB/c mice within 48 h of administration. However, in mice treated with sivelestat 30 min after the acetaminophen challenge, all mice survived, with reduced serum transaminase elevation and diminished hepatic necrosis. In addition, mice treated with sivelestat had reduced NOS-II expression and hepatic neutrophil infiltration after the acetaminophen challenge. Furthermore, treatment with sivelestat at 3 h after the acetaminophen challenge significantly improved survival. These findings indicate a new clinical application for sivelestat in the treatment of acetaminophen-induced liver failure through mechanisms involving the regulation of neutrophil migration and NO production.


Subject(s)
Chemical and Drug Induced Liver Injury , Liver Diseases , Mice , Animals , Acetaminophen/toxicity , Leukocyte Elastase/metabolism , Mice, Inbred BALB C , Transaminases , Chemical and Drug Induced Liver Injury/drug therapy , Necrosis
4.
Rheumatology (Oxford) ; 61(7): 3033-3048, 2022 07 06.
Article in English | MEDLINE | ID: mdl-34747459

ABSTRACT

OBJECTIVES: LN comprises various glomerular lesions, including endocapillary hypercellularity with macrophage infiltration. In this study, we aimed to clarify the involvement of macrophage-tropic chemokine receptors in the pathogenesis of these glomerular lesions. METHODS: MRL/lpr mouse-derived monoclonal IgG3 antibody-producing hybridomas, 2B11.3 and B1, were injected intraperitoneally into BALB/c mice [wild type (WT)] to induce endocapillary hypercellularity and wire-loop lesions, respectively. The expression of chemokine and chemokine receptors was analysed by quantitative real-time PCR and IF. The roles of chemokine receptors in these lesions were evaluated using chemokine receptor-deficient mice or a selective CCR5 antagonist, maraviroc. RESULTS: 2B11.3 caused glomerular endocapillary hypercellularity with a significant number of glomerular CD68-positive macrophages. Further, enhanced expression of CCL2, CCL3, CCR2, CCR5 and CX3CR1 was observed in the renal cortex, compared with B1 injection, which induced wire-loop lesions. In 2B11.3-induced glomerular lesions, CD68 -positive glomerular macrophages expressed CCL2, CCL3, CCR2, CCR5 and CX3CR1, while glomerular endothelial cells expressed CCL2, CCL3, CX3CL1 and CCR2. When 2B11.3 was injected, CCR2-/- and CCR5-/-, but not CX3CR1-/-, mice exhibited reduced endocapillary hypercellularity, attenuated glomerular macrophage infiltration and improved serum blood urea nitrogen levels. Only CCR2-/- mice developed wire-loop lesions. B1 injection caused wire-loop lesions in these chemokine receptor-deficient mice to a similar extent as WT. Maraviroc treatment reduced 2B11.3-induced endocapillary hypercellularity and improved serum blood urea nitrogen levels. CONCLUSION: CCR2 and CCR5 regulate glomerular macrophage infiltration and contribute to the development of glomerular endocapillary hypercellularity in LN. CCR5 inhibition can be a specific therapy for endocapillary hypercellularity without inducing wire-loop lesions.


Subject(s)
Kidney Diseases , Lupus Nephritis , Receptors, CCR2 , Receptors, CCR5 , Animals , Antibodies, Monoclonal , Endothelial Cells/metabolism , Immunoglobulin G/metabolism , Kidney Diseases/metabolism , Lupus Nephritis/pathology , Macrophages/metabolism , Maraviroc/metabolism , Mice , Mice, Inbred MRL lpr , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Receptors, CCR5/genetics , Receptors, CCR5/metabolism
5.
Immunity ; 38(3): 570-80, 2013 Mar 21.
Article in English | MEDLINE | ID: mdl-23434060

ABSTRACT

Monocytes and macrophages are important effectors and regulators of inflammation, and both can be divided into distinct subsets based on their phenotypes. The developmental and functional relationship between individual subsets of monocytes and those of macrophages has not been fully elucidated, although Ly6C(+)CCR2(+) inflammatory and Ly6C(-)CCR2(-) resident monocytes are generally thought to differentiate into M1 (classically activated) and M2 (alternatively activated) macrophages, respectively. Here we show that inflammatory monocytes recruited to allergic skin acquired an M2-like phenotype in response to basophil-derived interleukin-4 (IL-4) and exerted an anti-inflammatory function. CCR2-deficient mice unexpectedly displayed an exacerbation rather than alleviation of allergic inflammation, in spite of impaired recruitment of inflammatory monocytes to skin lesions. Adoptive transfer of inflammatory monocytes from wild-type but not IL-4 receptor-deficient mice dampened the exacerbated inflammation in CCR2-deficient mice. Thus, inflammatory monocytes can be converted from being proinflammatory to anti-inflammatory under the influence of basophils in allergic reactions.


Subject(s)
Basophils/immunology , Interleukin-4/immunology , Macrophages/immunology , Monocytes/immunology , Skin/immunology , Adoptive Transfer , Animals , Antigens, Ly/genetics , Antigens, Ly/immunology , Antigens, Ly/metabolism , Basophils/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Cells, Cultured , Flow Cytometry , Hypersensitivity/genetics , Hypersensitivity/immunology , Hypersensitivity/metabolism , Inflammation/genetics , Inflammation/immunology , Inflammation/metabolism , Interleukin-4/metabolism , Interleukin-4 Receptor alpha Subunit/genetics , Interleukin-4 Receptor alpha Subunit/immunology , Interleukin-4 Receptor alpha Subunit/metabolism , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Monocytes/metabolism , Receptors, CCR2/genetics , Receptors, CCR2/immunology , Receptors, CCR2/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Skin/metabolism , Skin/pathology
6.
Curr Issues Mol Biol ; 43(3): 1726-1740, 2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34698088

ABSTRACT

The chemokine monocyte chemoattractant protein-1 (MCP-1/CCL2) is shown to promote the progression of breast cancer. We previously identified cancer cell-derived granulocyte-macrophage colony-stimulating factor (GM-CSF) as a potential regulator of MCP-1 production in the murine 4T1 breast cancer, but it played a minimum role in overall MCP-1 production. Here, we evaluated the crosstalk between 4T1 cells and fibroblasts. When fibroblasts were co-cultured with 4T1 cells or stimulated with the culture supernatants of 4T1 cells (4T1-sup), MCP-1 production by fibroblasts markedly increased. 4T1 cells expressed mRNA for platelet-derived growth factor (PDGF)-a, b and c, and the PDGF receptor inhibitor crenolanib almost completely inhibited 4T1-sup-induced MCP-1 production by fibroblasts. However, PDGF receptor antagonists failed to reduce MCP-1 production in tumor-bearing mice. Histologically, 4T1 tumors contained a small number of αSMA-positive fibroblasts, and Mcp-1 mRNA was mainly associated with macrophages, especially those surrounding necrotic lesions on day 14, by in situ hybridization. Thus, although cancer cells have the capacity to crosstalk with fibroblasts via PDGFs, this crosstalk does not play a major role in MCP-1 production or cancer progression in this model. Unraveling complex crosstalk between cancer cells and stromal cells will help us identify new targets to help treat breast cancer patients.


Subject(s)
Breast Neoplasms/metabolism , Cancer-Associated Fibroblasts/metabolism , Cell Communication , Chemokine CCL2/biosynthesis , Animals , Biomarkers , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cancer-Associated Fibroblasts/drug effects , Cancer-Associated Fibroblasts/pathology , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Immunohistochemistry , In Situ Hybridization , Macrophage Activation/genetics , Macrophage Activation/immunology , Mice , Organ Specificity , Platelet-Derived Growth Factor/metabolism , Receptors, Platelet-Derived Growth Factor/antagonists & inhibitors , Tumor Microenvironment , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
7.
Cancer Sci ; 112(12): 4883-4893, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34632664

ABSTRACT

From a mouse triple-negative breast cancer cell line, 4T1, we previously established 4T1.3 clone with a high capacity to metastasize to bone after its orthotopic injection into mammary fat pad of immunocompetent mice. Subsequent analysis demonstrated that the interaction between cancer cells and fibroblasts in a bone cavity was crucial for bone metastasis focus formation arising from orthotopic injection of 4T1.3 cells. Here, we demonstrated that a member of the adhesion G-protein-coupled receptor (ADGR) family, G-protein-coupled receptor 56 (GPR56)/adhesion G-protein-coupled receptor G1 (ADGRG1), was expressed selectively in 4T1.3 grown in a bone cavity but not under in vitro conditions. Moreover, fibroblasts present in bone metastasis sites expressed type III collagen, a ligand for GPR56/ADGRG1. Consistently, GPR56/ADGRG1 proteins were detected in tumor cells in bone metastasis foci of human breast cancer patients. Deletion of GPR56/ADGRG1 from 4T1.3 cells reduced markedly intraosseous tumor formation upon their intraosseous injection. Conversely, intraosseous injection of GPR56/ADGRG1-transduced 4T1, TS/A (mouse breast cancer cell line), or MDA-MB-231 (human breast cancer cell line) exhibited enhanced intraosseous tumor formation. Furthermore, we proved that the cleavage at the extracellular region was indispensable for GPR56/ADGRG1-induced increase in breast cancer cell growth upon its intraosseous injection. Finally, inducible suppression of Gpr56/Adgrg1 gene expression in 4T1.3 cells attenuated bone metastasis formation with few effects on primary tumor formation in the spontaneous breast cancer bone metastasis model. Altogether, GPR56/ADGRG1 can be a novel target molecule to develop a strategy to prevent and/or treat breast cancer metastasis to bone.


Subject(s)
Bone Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Experimental/genetics , Receptors, G-Protein-Coupled/genetics , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/secondary , Cell Line, Tumor , Cell Proliferation/genetics , Collagen Type III/metabolism , Female , Gene Deletion , Humans , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice, Inbred BALB C , Mice, SCID , Protein Binding , Receptors, G-Protein-Coupled/metabolism , Tumor Burden/genetics
8.
Cytokine ; 126: 154862, 2020 02.
Article in English | MEDLINE | ID: mdl-31634687

ABSTRACT

Stimulation of H295R adrenocortical carcinoma cells with angiotensin II or cytokines induces the secretion of the chemokine interleukin-8 (IL-8). Here, we have analyzed the molecular mechanism of stimulus-induced IL-8 expression. IL-8 expression and IL-8 promoter activity increased in H295R cells expressing an activated Gαq-coupled designer receptor. H295R cells stimulated with either interleukin-1ß (IL-1ß) or phorbol ester also showed elevated IL-8 mRNA levels and higher IL-8 promoter activities. Deletion and point mutations of the IL-8 promoter revealed that the AP-1 binding site within the IL-8 promoter is essential to connect designer receptor stimulation with the transcriptional activation of the IL-8 gene. Expression of a constitutively active mutant of c-Jun, or expression of constitutively active mutants of the protein kinases MEKK1 and MKK6 confirmed that the IL-8 gene is a bona fide target of AP-1 in adrenocortical carcinoma cells. Upregulation of IL-8 expression in IL-1ß-treated H295R cells required NF-κB while the phorbol ester TPA used both the AP-1 and NF-κB sites of the IL-8 gene to stimulate IL-8 expression. These data were corroborated in experiments with chromatin-embedded AP-1 or NF-κB-responsive reporter genes. While stimulation of Gαq-coupled designer receptors increased the AP-1 activity in the cells, IL-1ß specifically stimulated NF-κB-regulated transcription. Stimulation of the cells with TPA increased both AP-1 and NF-κB activities. We conclude that stimulation of Gαq-coupled designer receptors or IL-1 receptors triggers distinct signaling pathways in H295R cells leading to the activation of either AP-1 or NF-κB. Nevertheless, both signaling cascades converge to the IL-8 gene, inducing IL-8 gene transcription.


Subject(s)
Adrenal Cortex Neoplasms/metabolism , Adrenocortical Carcinoma/metabolism , Interleukin-8/metabolism , NF-kappa B/metabolism , Transcription Factor AP-1/metabolism , Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/genetics , Genes, jun/genetics , Humans , Interleukin-1beta/pharmacology , Interleukin-8/genetics , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , MAP Kinase Kinase Kinase 1/genetics , MAP Kinase Kinase Kinase 1/metabolism , Point Mutation , Promoter Regions, Genetic , Sequence Deletion , Tetradecanoylphorbol Acetate/pharmacology , Transcriptional Activation/drug effects , Transcriptional Activation/genetics , Up-Regulation
9.
J Immunol ; 200(5): 1839-1852, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29378914

ABSTRACT

Macrophages in lungs can be classified into two subpopulations, alveolar macrophages (AMs) and interstitial macrophages (IMs), which reside in the alveolar and interstitial spaces, respectively. Accumulating evidence indicates the involvement of IMs in lung metastasis, but the roles of AMs in lung metastasis still remain elusive. An i.v. injection of a mouse hepatocellular carcinoma (HCC) cell line, BNL, caused lung metastasis foci with infiltration of AMs and IMs. Comprehensive determination of arachidonic acid metabolite levels revealed increases in leukotrienes and PGs in lungs in this metastasis model. A 5-lipoxygenase (LOX) inhibitor but not a cyclooxygenase inhibitor reduced the numbers of metastatic foci, particularly those of a larger size. A major 5-LOX metabolite, LTB4, augmented in vitro cell proliferation of human HCC cell lines as well as BNL cells. Moreover, in this lung metastasis course, AMs exhibited higher expression levels of the 5-LOX and LTB4 than IMs. Consistently, 5-LOX-expressing AMs increased in the lungs of human HCC patients with lung metastasis, compared with those without lung metastasis. Furthermore, intratracheal clodronate liposome injection selectively depleted AMs but not IMs, together with reduced LTB4 content and metastatic foci numbers in this lung metastasis process. Finally, IMs in mouse metastatic foci produced CCL2, thereby recruiting blood-borne, CCR2-expressing AMs into lungs. Thus, AMs can be recruited under the guidance of IM-derived CCL2 into metastatic lungs and can eventually contribute to the progression of lung metastasis by providing a potent arachidonic acid-derived tumor growth promoting mediator, LTB4.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Leukotriene B4/metabolism , Liver Neoplasms/metabolism , Lung Neoplasms/metabolism , Lung/metabolism , Macrophages, Alveolar/metabolism , Macrophages, Alveolar/pathology , Animals , Arachidonate 5-Lipoxygenase/metabolism , Arachidonic Acid/pharmacology , Bronchoalveolar Lavage Fluid , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Cyclooxygenase Inhibitors/pharmacology , Humans , Leukotrienes/metabolism , Liver Neoplasms/pathology , Lung/drug effects , Lung/pathology , Lung Neoplasms/pathology , Macrophages/drug effects , Macrophages/metabolism , Macrophages/pathology , Macrophages, Alveolar/drug effects , Male , Mice , Mice, Inbred BALB C
10.
Adv Exp Med Biol ; 1231: 23-32, 2020.
Article in English | MEDLINE | ID: mdl-32060843

ABSTRACT

CCL4, a CC chemokine, previously known as macrophage inflammatory protein (MIP)-1ß, has diverse effects on various types of immune and nonimmune cells by the virtue of its interaction with its specific receptor, CCR5, in collaboration with related but distinct CC chemokines such as CCL3 and CCL5, which can also bind CCR5. Several lines of evidence indicate that CCL4 can promote tumor development and progression by recruiting regulatory T cells and pro-tumorigenic macrophages, and acting on other resident cells present in the tumor microenvironment, such as fibroblasts and endothelial cells, to facilitate their pro-tumorigenic capacities. These observations suggest the potential efficacy of CCR5 antagonists for cancer treatment. On the contrary, under some situations, CCL4 can enhance tumor immunity by recruiting cytolytic lymphocytes and macrophages with phagocytic ability. Thus, presently, the clinical application of CCR5 antagonists warrants more detailed analysis of the role of CCL4 and other CCR5-binding chemokines in the tumor microenvironment.


Subject(s)
Chemokine CCL4/metabolism , Signal Transduction , Tumor Microenvironment , Animals , Chemokine CCL4/immunology , Humans , Macrophages/immunology , Receptors, CCR5/metabolism , T-Lymphocytes, Regulatory/immunology
11.
Int J Mol Sci ; 21(10)2020 May 14.
Article in English | MEDLINE | ID: mdl-32422991

ABSTRACT

Neutrophils are the most abundant circulating leukocytes in humans. Neutrophil infiltration into tumor tissues has long been observed but its roles have been ignored due to the presumed short life cycle and metabolic incompetence of neutrophils. Recent advances in neutrophil biology research have revealed that neutrophils have a longer life cycle with a potential to express various bioactive molecules. Clinical studies have simultaneously unraveled an increase in the neutrophil-lymphocyte ratio (NLR), a ratio of absolute neutrophil to absolute lymphocyte numbers in cancer patient peripheral blood and an association of higher NLR with more advanced or aggressive disease. As a consequence, tumor-associated neutrophils (TANs) have emerged as important players in tumor microenvironment. The elucidation of the roles of TANs, however, has been hampered by their multitude of plasticity in terms of phenotypes and functionality. Difficulties are further enhanced by the presence of a related cell population-polymorphonuclear leukocyte (PMN)-myeloid-derived suppressor cells (MDSCs)-and various dissimilar aspects of neutrophil biology between humans and mice. Here, we discuss TAN biology in various tumorigenesis processes, and particularly focus on the context-dependent functional heterogeneity of TANs.


Subject(s)
Carcinogenesis/genetics , Neoplasms/blood , Neutrophil Infiltration/genetics , Neutrophils/pathology , Cell Lineage/genetics , Humans , Leukocyte Count , Lymphocyte Count , Myeloid-Derived Suppressor Cells/pathology , Neoplasms/genetics , Neoplasms/pathology , Neoplastic Cells, Circulating/pathology , Tumor Microenvironment/genetics
12.
Arterioscler Thromb Vasc Biol ; 38(11): 2638-2650, 2018 11.
Article in English | MEDLINE | ID: mdl-30354252

ABSTRACT

Objective- Deep vein thrombosis results from a combination of risk factors including genetic conditions, obesity, drugs, pregnancy, aging, and malignancy. We examined pathophysiological roles of the TNF-α (tumor necrosis factor-α)-TNF-Rp55 (tumor necrosis factor receptor p55) axis in thrombus resolution using Tnfrp55-/- (TNF-Rp55-deficient) mice. Approach and Results- On ligating the inferior vena cava of wild-type (WT) mice, venous thrombi formed and grew progressively until 5 days but shrunk to <50% of the thrombus weight at day 14. Concomitantly, inferior vena cava ligation enhanced intrathrombotic gene expression of Tnfa and Tnfrp55, and intrathrombotic macrophages expressed both TNF-α and TNF-Rp55 proteins. In Tnfrp55-/- mice treated with the same manner, thrombus formed at a similar rate for 5 days, but shrinking was delayed compared with WT mice. Moreover, the blood flow recovery in thrombosed inferior vena cava was suspended in Tnfrp55-/- mice compared with WT mice. Intrathrombotic Plau (urokinase-type plasminogen activator), Mmp2 (matrix metalloproteinase 2), and Mmp9 (matrix metalloproteinase 9) mRNA expression was significantly reduced in Tnfrp55-/- mice, compared with WT ones. Supportingly, the administration of anti-TNF-α antibody or TNF-α inhibitor (etanercept) delayed the thrombus resolution in WT mice. Furthermore, TNF-α treatment enhanced gene expression of Plau, Mmp2, and Mmp9 in WT macrophages but not Tnfrp55-/- macrophages. These effects were significantly suppressed by ERK (extracellular signal regulated kinase) and NF-κB (nuclear factor-kappa B) inhibitors. Therefore, the lack of TNF-Rp55 has detrimental roles in the thrombus resolution by suppressing PLAU, MMP-2, and MMP-9 expression. In contrast, TNF-α administration accelerated thrombus resolution in WT but not Tnfrp55-/- mice. Conclusions- The TNF-α-TNF-Rp55 axis may have essential roles in the resolution of venous thrombus in mice.


Subject(s)
Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vena Cava, Inferior/metabolism , Venous Thrombosis/metabolism , Animals , Cells, Cultured , Disease Models, Animal , Macrophages, Peritoneal/metabolism , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice, Inbred BALB C , Mice, Knockout , Receptors, Tumor Necrosis Factor, Type I/deficiency , Receptors, Tumor Necrosis Factor, Type I/genetics , Signal Transduction , Time Factors , Urokinase-Type Plasminogen Activator/genetics , Urokinase-Type Plasminogen Activator/metabolism , Vena Cava, Inferior/pathology , Venous Thrombosis/blood , Venous Thrombosis/pathology
13.
Int J Mol Sci ; 20(24)2019 Dec 16.
Article in English | MEDLINE | ID: mdl-31888216

ABSTRACT

We previously reported that 4T1 murine breast cancer cells produce GM-CSF that up-regulates macrophage expression of several cancer promoting genes, including Mcp-1/Ccl2, Ccl17 and Rankl, suggesting a critical role of cancer cell-derived GM-CSF in cancer progression. Here, we attempted to define whether 4T1 cell-derived GM-CSF contributes to the expression of these genes by 4T1tumors, and their subsequent progression. Intraperitoneal injection of anti-GM-CSF neutralizing antibody did not decrease the expression of Mcp-1, Ccl17 or Rankl mRNA by 4T1 tumors. To further examine the role of cancer cell-derived GM-CSF, we generated GM-CSF-deficient 4T1 cells by using the Crisper-Cas9 system. As previously demonstrated, 4T1 cells are a mixture of cells and cloning of cells by itself significantly reduced tumor growth and lung metastasis. By contrast, GM-CSF-deficiency did not affect tumor growth, lung metastasis or the expression of these chemokine and cytokine genes in tumor tissues. By in-situ hybridization, the expression of Mcp-1 mRNA was detected in both F4/80-expressing and non-expressing cells in tumors of GM-CSF-deficient cells. These results indicate that cancer cell-derived GM-CSF is dispensable for the tuning of the 4T1 tumor microenvironment and the production of MCP-1, CCL17 or RANKL in the 4T1 tumor microenvironment is likely regulated by redundant mechanisms.


Subject(s)
Disease Progression , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Animals , Base Sequence , CRISPR-Cas Systems/genetics , Cell Line, Tumor , Cell Proliferation , Clone Cells , Female , Gene Expression Regulation, Neoplastic , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocytes/metabolism , Macrophages/metabolism , Mammary Neoplasms, Animal/genetics , Mice, Inbred BALB C , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Splenomegaly/pathology
14.
Blood ; 127(21): 2607-17, 2016 05 26.
Article in English | MEDLINE | ID: mdl-27006388

ABSTRACT

Basophilia is a frequently observed hematological abnormality in chronic myeloid leukemia (CML), but its pathophysiological roles are undefined. We previously demonstrated that an inflammatory chemokine, CCL3, preferentially acts on normal hematopoietic stem/progenitor cells and crucially contributes to the maintenance of leukemia initiating cells (LICs) in bone marrow (BM) during the initiation process of CML. However, the major cellular source of CCL3 in BM and the precise mechanism of CCL3-mediated maintenance of LICs remain to be investigated. To delineate the cellular process facilitating this CCL3-mediated crosstalk between normal and leukemic hematopoiesis, we precisely examined CCL3-expressing cells and their functions in both normal hematopoiesis and CML leukemogenesis. Herein, we demonstrate that basophils can constitutively express CCL3 to negatively regulate the normal hematopoietic process, especially hematopoietic reconstitution after BM transplantation. Moreover, CCL3-expressing basophil-like leukemia cells were found to accumulate in CML BM and supported the predominant expansion of LICs therein. These observations suggest that intra-BM basophil expansion can favor leukemia-tropic hematopoiesis in CML by providing CCL3, a potent inhibitor of normal hematopoiesis and that basophil-derived CCL3 may be a novel target molecule for the treatment of CML.


Subject(s)
Basophils/metabolism , Bone Marrow/metabolism , Chemokine CCL3/biosynthesis , Gene Expression Regulation, Leukemic , Hematopoiesis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Neoplasm Proteins/biosynthesis , Neoplastic Stem Cells/metabolism , Animals , Basophils/pathology , Bone Marrow/pathology , Chemokine CCL3/genetics , Female , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Proteins/genetics , Neoplastic Stem Cells/pathology
15.
Cytokine ; 103: 133-141, 2018 03.
Article in English | MEDLINE | ID: mdl-28982580

ABSTRACT

Stimulation of Ca2+ permeable TRPM3 (transient receptor potential melastatin-3) channels with the steroid ligand pregnenolone sulfate activates stimulus-responsive transcription factors, including the transcription factor AP-1 (activator protein-1). As part of a search for AP-1-regulated target genes we analyzed the gene encoding interleukin-8 (IL-8) in HEK293 cells expressing TRPM3 channels. Here, we show that stimulation of TRPM3 channels activated transcription of an IL-8 promoter-controlled reporter gene that was embedded into the chromatin of the cells. Mutational analysis of the IL-8 promoter revealed that the AP-1 binding site of the IL-8 promoter was essential to connect TRPM3 stimulation with the transcription of the IL-8 gene. Genetic experiments revealed that the basic region leucine zipper proteins c-Jun and ATF2 and the ternary complex factor Elk-1 are essential to couple TRPM3 channel stimulation with the IL-8 gene. Moreover, we identified extracellular signal-regulated protein kinase (ERK1/2) as signal transducer connecting TRPM3 stimulation with enhanced transcription of the IL-8 gene. Furthermore, we show that stimulation of TRPC6 (transient receptor potential canonical-6) channels with its ligand hyperforin also increased IL-8 promoter activity, involving the AP-1 binding site within the IL-8 gene, suggesting that activation of IL-8 gene transcription may be a common theme following TRP channel stimulation.


Subject(s)
Interleukin-8/biosynthesis , MAP Kinase Signaling System , Promoter Regions, Genetic , TRPM Cation Channels/metabolism , Transcription Factor AP-1/metabolism , Transcription, Genetic , HEK293 Cells , Humans , Interleukin-8/genetics , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , TRPM Cation Channels/genetics , Transcription Factor AP-1/genetics
16.
Pharmacol Res ; 134: 238-245, 2018 08.
Article in English | MEDLINE | ID: mdl-30018026

ABSTRACT

The polyphenol resveratrol activates stimulus-regulated transcription factors, including activator protein-1 (AP-1). As part of a search for resveratrol-regulated target genes we analyzed the gene encoding the chemokine interleukin-8 (IL-8) which is regulated by AP-1. Here, we show that treatment of HEK293 cells with resveratrol induced the expression of IL-8 and activated transcription of a chromatin-embedded IL-8 promoter-controlled reporter gene. Mutational analysis of the IL-8 promoter revealed that it was not the AP-1 binding site, but rather the NF-κB site that was essential to connect resveratrol stimulation with the transcriptional activation of the IL-8 gene. Thus, the NF-κB site of the IL-8 gene functions as resveratrol-responsive element. The analysis of an NF-κB-responsive reporter gene, controlled by the HIV-1 long terminal repeat (LTR), showed that resveratrol stimulation increased the transcriptional activity of NF-κB. These data were corroborated by an experiment showing that incubation of the cells with the NF-κB inhibitor JSH-23 attenuated resveratrol-induced activation of the IL-8 promoter and reduced the cellular NF-κB activity following stimulation of the cells with resveratrol. The protein kinase extracellular signal-regulated protein kinase ERK1/2 was identified to function as signal transducer connecting resveratrol stimulation with the activation of NF-κB and IL-8 promoter-controlled transcription. We conclude that resveratrol, proposed to exhibit anti-inflammatory activity, stimulates expression of the pro-inflammatory chemokine IL-8 via NF-κB, which is known as an important mediator of inflammatory processes.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Interleukin-8/metabolism , NF-kappa B/metabolism , Resveratrol/pharmacology , Transcription, Genetic/drug effects , Transcriptional Activation/drug effects , Binding Sites , Caco-2 Cells , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Hep G2 Cells , Humans , Inflammation/genetics , Inflammation/metabolism , Interleukin-8/genetics , NF-kappa B/genetics , Promoter Regions, Genetic , Signal Transduction/drug effects , Up-Regulation
17.
Int J Mol Sci ; 20(1)2018 Dec 29.
Article in English | MEDLINE | ID: mdl-30597969

ABSTRACT

Metastasis is responsible for most of the cancer-associated deaths and proceeds through multiple steps. Several lines of evidence have established an indispensable involvement of macrophages present at the primary tumor sites in various steps of metastasis, from primary tumor growth to its intravasation into circulation. The lungs encompass a large, dense vascular area and, therefore, are vulnerable to metastasis, particularly, hematogenous ones arising from various types of neoplasms. Lung tissues constitutively contain several types of tissue-resident macrophages and circulating monocytes to counteract potentially harmful exogenous materials, which directly reach through the airway. Recent advances have provided an insight into the ontogenetic, phenotypic, and functional heterogeneity of these lung macrophage and monocyte populations, under resting and inflammatory conditions. In this review, we discuss the ontogeny, trafficking dynamics, and functions of these pulmonary macrophages and monocytes and their potential roles in lung metastasis and measures to combat lung metastasis by targeting these populations.


Subject(s)
Lung Neoplasms/pathology , Macrophages/pathology , Animals , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Macrophages/metabolism
18.
Int J Mol Sci ; 18(8)2017 Aug 22.
Article in English | MEDLINE | ID: mdl-28829353

ABSTRACT

All blood lineage cells are generated from hematopoietic stem cells (HSCs), which reside in bone marrow after birth. HSCs self-renew, proliferate, and differentiate into mature progeny under the control of local microenvironments including hematopoietic niche, which can deliver regulatory signals in the form of bound or secreted molecules and from physical cues such as oxygen tension and shear stress. Among these mediators, accumulating evidence indicates the potential involvement of several chemokines, particularly CXCL12, in the interaction between HSCs and bone marrow microenvironments. Fusion between breakpoint cluster region (BCR) and Abelson murine leukemia viral oncogene homolog (ABL)-1 gene gives rise to BCR-ABL protein with a constitutive tyrosine kinase activity and transforms HSCs and/or hematopoietic progenitor cells (HPCs) into disease-propagating leukemia stem cells (LSCs) in chronic myeloid leukemia (CML). LSCs can self-renew, proliferate, and differentiate under the influence of the signals delivered by bone marrow microenvironments including niche, as HSCs can. Thus, the interaction with bone marrow microenvironments is indispensable for the initiation, maintenance, and progression of CML. Moreover, the crosstalk between LSCs and bone marrow microenvironments can contribute to some instances of therapeutic resistance. Furthermore, evidence is accumulating to indicate the important roles of bone marrow microenvironment-derived chemokines. Hence, we will herein discuss the roles of chemokines in CML with a focus on bone marrow microenvironments.


Subject(s)
Bone Marrow/metabolism , Chemokines/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Tumor Microenvironment , Animals , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Neoplastic Stem Cells/metabolism , Protein Binding , Receptors, Chemokine/metabolism , Signal Transduction , Stem Cell Niche
19.
Biochem Biophys Res Commun ; 477(3): 515-9, 2016 08 26.
Article in English | MEDLINE | ID: mdl-27311854

ABSTRACT

Patients with pancreatic ductal adenocarcinoma (PDAC) commonly require chemotherapy because they frequently develop metastatic disease or locally advanced tumors. Gemcitabine, an analogue of cytosine arabinoside, is commonly used for PDAC treatment. We observed that gemcitabine induced senescence phenotypes characterized by enhanced senescence-associated ß-galactosidase (SA ß-Gal) staining and increased expression of senescence-associated molecules in two human pancreatic cancer cell lines, Miapaca-2 and Panc-1, which exhibit resistance to gemcitabine but not L3.pl cells with a high sensitivity to gemcitabine. Gemcitabine-induced cell senescence can be inhibited by reactive oxygen species inhibitor, N-acetyl cysteine. Although gemcitabine also enhanced CXCL8 expression, anti-CXCL8 antibody failed to reduce gemcitabine-induced increases in SA ß-Gal-positive cell numbers. These observations would indicate that cell senescence can proceed independently of CXCL8 expression, a characteristic feature of senescence-associated secretion phenotype.


Subject(s)
Cellular Senescence/drug effects , Deoxycytidine/analogs & derivatives , Pancreatic Neoplasms/pathology , Deoxycytidine/pharmacology , Humans , Gemcitabine
20.
Rinsho Ketsueki ; 57(2): 129-36, 2016 Feb.
Article in Japanese | MEDLINE | ID: mdl-26935630

ABSTRACT

Several tyrosine kinase inhibitors have been developed to target the BCR-ABL fusion gene, a pathognomonic genetic change in chronic myeloid leukemia (CML), and have dramatically improved the outcomes of CML patients. BCR-ABL-expressing CML cells compete with normal hematopoietic cells over the limited space in the bone marrow to proliferate. Moreover, CML cells can gain resistance to tyrosine kinase inhibitors by utilizing bone marrow microenvironments. Thus, in order to develop a novel treatment strategy for CML, it is necessary to elucidate the cellular and molecular basis underlying the interactions between CML and normal hematopoietic cells. Herein, we discuss the roles of chemokines, particularly CXCL12 and CCL3, in reconstruction processes of bone marrow microenvironments by CML cells and the possibility of novel treatment modalities against CML, based on targeting these chemokines.


Subject(s)
Chemokines/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Bone Marrow/metabolism , Cellular Microenvironment , Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL