Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 39
1.
Oncol Rep ; 45(1): 291-298, 2021 01.
Article En | MEDLINE | ID: mdl-33155662

Cancer stem­like cells (CSCs; also referred to as tumor­initiating cells) play crucial roles in tumor progression and aggressiveness. Recent studies have demonstrated the antitumor activity of zoledronic acid (ZA), a third­generation bisphosphonate, in various types of human cancer. However, its effect on oral CSCs and the underlying mechanism remain obscure. The present study demonstrated that ZA suppresses the growth and stemness properties of oral/oropharyngeal squamous cell carcinoma (OSCC) cells. ZA inhibited the malignant characteristics of OSCC cells, such as anchorage­independent growth and epithelial thickening in organotypic raft cultures. Moreover, ZA treatment resulted in suppression of self­renewal capacity, a key feature of CSCs. ZA also inhibited important CSC properties, such as migration and chemo­radioresistance. Mechanistically, ZA exposure significantly decreased chemokine (C­C motif) ligand 3 (CCL3) expression in OSCC cells. It was further demonstrated that CCL3 signaling via its receptor is crucial for supporting the CSC phenotype in OSCC cells. Moreover, an antagonist of the CCL3 receptor reversed the effect of CCL3 on CSC properties, and exogenous CCL3 rescued the suppressaed CSC phenotype in ZA­treated OSCC cells. These results demonstrated that ZA suppresses the CSC phenotype in OSCC cells by reducing CCL3 expression, suggesting that ZA may be an effective therapeutic agent for oral cancer by targeting CSCs.


Chemokine CCL3/physiology , Mouth Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Squamous Cell Carcinoma of Head and Neck/drug therapy , Zoledronic Acid/pharmacology , Cell Line, Tumor , Chemokine CCL3/analysis , Humans , Mouth Neoplasms/immunology , Mouth Neoplasms/pathology , Neoplastic Stem Cells/chemistry , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/pathology
2.
Front Immunol ; 11: 306, 2020.
Article En | MEDLINE | ID: mdl-32194558

CCL3, a member of the CC-chemokine family, has been associated with macrophage recruitment to heart tissue and parasite control in the acute infection of mouse with Trypanosoma cruzi, the causative agent of Chagas disease. Here, we approached the participation of CCL3 in chronic chagasic cardiomyopathy (CCC), the main clinical form of Chagas disease. We induced CCC in C57BL/6 (ccl3+/+) and CCL3-deficient (ccl3-/-) mice by infection with the Colombian Type I strain. In ccl3+/+ mice, high levels of CCL3 mRNA and protein were detected in the heart tissue during the acute and chronic infection. Survival was not affected by CCL3 deficiency. In comparison with ccl3+/+, chronically infected ccl3-/- mice presented reduced cardiac parasitism and inflammation due to CD8+ cells and macrophages. Leukocytosis was decreased in infected ccl3-/- mice, paralleling the accumulation of CD8+ T cells devoid of activated CCR5+ LFA-1+ cells in the spleen. Further, T. cruzi-infected ccl3-/-mice presented reduced frequency of interferon-gamma (IFNγ)+ cells and numbers of parasite-specific IFNγ-producing cells, while the T. cruzi antigen-specific cytotoxic activity was increased. Stimulation of CCL3-deficient macrophages with IFNγ improved parasite control, in a milieu with reduced nitric oxide (NOx) and tumor necrosis factor (TNF), but similar interleukin-10 (IL-10), concentrations. In comparison with chronically T. cruzi-infected ccl3+/+ counterparts, ccl3-/- mice did not show enlarged heart, loss of left ventricular ejection fraction, QTc prolongation and elevated CK-MB activity. Compared with ccl3+/+, infected ccl3-/- mice showed reduced concentrations of TNF, while IL-10 levels were not affected, in the heart milieu. In spleen of ccl3+/+ NI controls, most of the CD8+ T-cells expressing the CCL3 receptors CCR1 or CCR5 were IL-10+, while in infected mice these cells were mainly TNF+. Lastly, selective blockage of CCR1/CCR5 (Met-RANTES therapy) in chronically infected ccl3+/+ mice reversed pivotal electrical abnormalities (bradycardia, prolonged PR, and QTc interval), in correlation with reduced TNF and, mainly, CCL3 levels in the heart tissue. Therefore, in the chronic T. cruzi infection CCL3 takes part in parasite persistence and contributes to form a CD8+ T-cell and macrophage-enriched cardiac inflammation. Further, increased levels of CCL3 create a scenario with abundant IFNγ and TNF, associated with cardiomyocyte injury, heart dysfunction and QTc prolongation, biomarkers of severity of Chagas' heart disease.


Chagas Cardiomyopathy/physiopathology , Chemokine CCL3/physiology , Interferon-gamma/physiology , Macrophages, Peritoneal/parasitology , Parasitemia/physiopathology , Trypanosoma cruzi/physiology , Tumor Necrosis Factor-alpha/physiology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cells, Cultured , Chagas Cardiomyopathy/parasitology , Chagas Cardiomyopathy/pathology , Chemokine CCL3/deficiency , Chemokine CCL3/pharmacology , Chemokine CCL5/pharmacology , Chemokine CCL5/therapeutic use , Chemotaxis, Leukocyte/drug effects , Cytokines/biosynthesis , Cytokines/genetics , Cytokines/pharmacology , Electrocardiography/drug effects , Female , Interferon-gamma/pharmacology , Macrophages, Peritoneal/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocarditis/etiology , Myocarditis/pathology , Myocarditis/physiopathology , RNA, Messenger/biosynthesis , Receptors, Chemokine/antagonists & inhibitors , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , Specific Pathogen-Free Organisms , Spleen/immunology , Spleen/metabolism , Stroke Volume , Trypanosoma cruzi/isolation & purification , Tumor Necrosis Factor-alpha/analysis
4.
Cancer Lett ; 389: 49-58, 2017 03 28.
Article En | MEDLINE | ID: mdl-28041977

The induction of localized pro-inflammatory niches in the periphery is instrumental in metastasis. In order to better understand how tumors engage distal sites and activate a pro-inflammatory response we utilized syngeneic breast cancers as a model and showed that soluble factors from the neoplastic epithelium activate the expression of the monocyte chemoattractive protein (MCP) chemokines of the mouse 11C cluster that include Ccl1, Ccl2, Ccl7, Ccl8, Ccl11 and Ccl12. Tissues such as the lungs and the brain, that are more prone to colonization by breast cancer cells, were more sensitive to MCP cluster chemokine induction than others such as the liver. Subsequent analyses involving chemokine arrays in breast cancer cells and media followed by functional validation assays in in vitro and in vivo identified the cytokine Ccl3 as the principle mediator of the communication between the neoplastic epithelium and the peripheral tissues in terms of MCP cluster chemokine induction. Our results show that MCP chemokines are activated in peripheral tissues of breast cancer-bearing mice, by a mechanism that involves breast cancer cell-derived Ccl3. Interference with the expression of cancer cell-derived Ccl3 may find application in the management of breast cancer metastases.


Chemokine CCL3/physiology , Mammary Neoplasms, Experimental/immunology , Monocyte Chemoattractant Proteins/biosynthesis , Animals , Female , Mammary Neoplasms, Experimental/mortality , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Inbred C57BL , NIH 3T3 Cells , Neoplasm Metastasis
5.
Proc Natl Acad Sci U S A ; 113(33): 9298-303, 2016 08 16.
Article En | MEDLINE | ID: mdl-27474165

Macrophages are key regulators of fibrosis development and resolution. Elucidating the mechanisms by which they mediate this process is crucial for establishing their therapeutic potential. Here, we use experimental models of liver fibrosis to show that deficiency of the scavenger receptor, stabilin-1, exacerbates fibrosis and delays resolution during the recovery phase. We detected a subset of stabilin-1(+) macrophages that were induced at sites of cellular injury close to the hepatic scar in mouse models of liver fibrosis and in human liver disease. Stabilin-1 deficiency abrogated malondialdehyde-LDL (MDA-LDL) uptake by hepatic macrophages and was associated with excess collagen III deposition. Mechanistically, the lack of stabilin-1 led to elevated intrahepatic levels of the profibrogenic chemokine CCL3 and an increase in GFAP(+) fibrogenic cells. Stabilin-1(-/-) macrophages demonstrated a proinflammatory phenotype during liver injury and the normal induction of Ly6C(lo) monocytes during resolution was absent in stabilin-1 knockouts leading to persistence of fibrosis. Human stabilin-1(+) monocytes efficiently internalized MDA-LDL and this suppressed their ability to secrete CCL3, suggesting that loss of stabilin-1 removes a brake to CCL3 secretion. Experiments with cell-lineage-specific knockouts revealed that stabilin-1 expression in myeloid cells is required for the induction of this subset of macrophages and that increased fibrosis occurs in their absence. This study demonstrates a previously unidentified regulatory pathway in fibrogenesis in which a macrophage scavenger receptor protects against organ fibrosis by removing fibrogenic products of lipid peroxidation. Thus, stabilin-1(+) macrophages shape the tissue microenvironment during liver injury and healing.


Cell Adhesion Molecules, Neuronal/physiology , Chemical and Drug Induced Liver Injury/complications , Homeostasis , Liver Cirrhosis/prevention & control , Macrophages/physiology , Animals , Carbon Tetrachloride , Chemokine CCL3/physiology , Choline Deficiency/complications , Humans , Lipoproteins, LDL/metabolism , Malondialdehyde/analogs & derivatives , Malondialdehyde/metabolism , Mice
6.
J Indian Soc Pedod Prev Dent ; 34(3): 273-9, 2016.
Article En | MEDLINE | ID: mdl-27461813

AIMS AND OBJECTIVES: The study was conducted to detect the presence of macrophage inflammatory protein-1α (MIP-1α) and MIP-1ß and estimate their levels in gingival crevicular fluid (GCF) in children with dental caries and stainless steel crowns. MATERIALS AND METHODS: A total of 80 children with primary dentition were selected and categorized into four groups with twenty in each group; Group 1 - healthy subjects, Group 2 - dental caries, Group 3 - dental caries involving the pulp, and Group 4 - stainless steel crowns. GCF samples were collected by an extra-crevicular method with microcapillary pipettes. The GCF samples were quantified by ELISA and the levels of MIP-1α and MIP-1ß were determined. RESULTS: MIP-1α and MIP-1ß were detected in all the samples. Highest mean concentration in GCF was obtained for Group 3 followed by Groups 2 and 4 while the lowest concentration was seen in Group 1. This suggests that MIP-1α and MIP-1ß levels in GCF increased proportionately with the inflammation. CONCLUSIONS: GCF serves as a noninvasive diagnostic fluid to measure biomarkers released during dental caries initiation and progression. MIP-1α and MIP-1ß chemokines can be considered as novel biomarkers, in biological mechanism underlying the pathogenesis and inflammation in children with dental caries and stainless steel crowns.


Chemokine CCL3/analysis , Chemokine CCL4/analysis , Chemokines/analysis , Crowns , Dental Caries/metabolism , Gingival Crevicular Fluid/chemistry , Case-Control Studies , Chemokine CCL3/physiology , Chemokine CCL4/physiology , Chemokines/physiology , Child, Preschool , Enzyme-Linked Immunosorbent Assay , Female , Humans , Male , Stainless Steel
7.
Stem Cells Dev ; 25(8): 575-85, 2016 Apr 15.
Article En | MEDLINE | ID: mdl-26943017

Dipeptidyl peptidase 4 (DPP4)/CD26 truncates certain proteins, and this posttranslational modification can influence their activity. Truncated (T) colony-stimulating factors (CSFs) are decreased in potency for stimulating proliferation of hematopoietic progenitor cells (HPCs). T-CXCL12, a modified chemokine, is inactive as an HPC chemotactic, survival, and enhancing factor for replating or ex-vivo expansion of HPCs. Moreover, T-CSFs and T-CXCL12 specifically downmodulates the positively acting effects of their own full-length molecule. Other chemokines have DPP4 truncation sites. In the present study, we evaluated effects of DPP4 inhibition (by Diprotin A) or gene deletion of HPC on chemokine inhibition of multicytokine-stimulated HPC, and on chemokine-enhancing effects on single CSF-stimulated HPC proliferation, as well as effects of DPP4 treatment of a number of chemokines. Myelosuppressive effects of chemokines with, but not without, a DPP4 truncation site were greatly enhanced in inhibitory potency by pretreating target bone marrow (BM) cells with Diprotin A, or by assaying their activity on dpp4/cd26(-/-) BM cells. DPP4 treatment of myelosuppressive chemokines containing a DPP4 truncation site produced a nonmyelosuppressive molecule, but one which had the capacity to block suppression by that unmodified chemokine both in vitro and in vivo. Additionally, DPP4 treatment ablated the single cytokine-stimulated HPC-enhancing activity of CCL3/MIP-1α and CCL4/MIP-1ß, and blocked the enhancing activity of each unmodified molecule, in vitro and in vivo. These results highlight the functional posttranslational modulating effects of DPP4 on chemokine activities, and information offering additional biological insight into chemokine regulation of hematopoiesis.


Chemokine CCL3/physiology , Chemokine CCL4/physiology , Dipeptidyl Peptidase 4/physiology , Animals , Cell Proliferation , Chemokine CCL3/chemistry , Chemokine CCL4/chemistry , Dipeptidyl Peptidase 4/chemistry , Female , Hematopoiesis , Hematopoietic Stem Cells/physiology , Male , Mice, Inbred C57BL , Mice, Knockout , Protein Processing, Post-Translational , Proteolysis
8.
Mol Neurobiol ; 53(3): 1551-1564, 2016 Apr.
Article En | MEDLINE | ID: mdl-25663168

A broad spectrum of diseases is characterized by myelin abnormalities, oligodendrocyte pathology, and concomitant glia activation, among multiple sclerosis (MS). Our knowledge regarding the factors triggering gliosis and demyelination is scanty. Chemokines are pivotal for microglia and astrocyte activation and orchestrate critical steps during the formation of central nervous system (CNS) demyelinating lesions. Redundant functions of chemokines complicate, however, the study of their functional relevance. We used the cuprizone model to study redundant functions of two chemokines, CCL2/MCP1 and CCL3/MIP1α, which are critically involved in the pathological process of cuprizone-induced demyelination. First, we generated a mutant mouse strain lacking functional genes of both chemokines and demonstrated that double-mutant animals are viable, fertile, and do not present with gross abnormalities. Astrocytes and peritoneal macrophages, cultured form tissues of these animals did neither express CCL2 nor CCL3. Exposure to cuprizone resulted in increased CCL2 and CCL3 brain levels in wild-type but not mutant animals. Cuprizone-induced demyelination, oligodendrocyte loss, and astrogliosis were significantly ameliorated in the cortex but not corpus callosum of chemokine-deficient animals. In summary, we provide a novel powerful model to study the redundant function of two important chemokines. Our study reveals that chemokine function in the CNS redounds to region-specific pathophysiological events.


Blood-Brain Barrier , Chemokine CCL2/deficiency , Chemokine CCL3/deficiency , Demyelinating Diseases/pathology , Gray Matter/pathology , White Matter/pathology , Animals , Astrocytes/metabolism , Brain/pathology , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/physiology , Chemokine CCL3/genetics , Chemokine CCL3/physiology , Cuprizone/toxicity , Demyelinating Diseases/chemically induced , Disease Models, Animal , Female , Gliosis/chemically induced , Gliosis/pathology , Macrophages, Peritoneal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Viscera/pathology
9.
Oncotarget ; 6(27): 24218-29, 2015 Sep 15.
Article En | MEDLINE | ID: mdl-26155942

We previously showed that macrophages (MΦs) infiltrate the bone marrow (BM) of patients with myeloma and may play a role in drug resistance. This study analyzed chemokines expressed by myeloma BM that are responsible for recruiting monocytes to the tumor bed. We found that chemokines CCL3, CCL14, and CCL2 were highly expressed by myeloma and BM cells, and the levels of CCL14 and CCL3 in myeloma BM positively correlated with the percentage of BM-infiltrating MΦs. In vitro, these chemokines were responsible for chemoattracting human monocytes to tumor sites and in vivo for MΦ infiltration into myeloma-bearing BM in the 5TGM1 mouse model. Surprisingly, we also found that these chemokines stimulated MΦ in vitro proliferation induced by myeloma cells and in vivo in a human myeloma xenograft SCID mouse model. The chemokines also activated normal MΦ polarization and differentiation into myeloma-associated MΦs. Western blot analysis revealed that these chemokines promoted growth and survival signaling in MΦs via activating the PI3K/Akt and ERK MAPK pathways and c-myc expression. Thus, this study provides novel insight into the mechanism of MΦ infiltration of BM and also potential targets for improving the efficacy of chemotherapy in myeloma.


Bone Marrow Cells/cytology , Chemokine CCL2/physiology , Chemokine CCL3/physiology , Chemokines, CC/physiology , Macrophages/cytology , Multiple Myeloma/metabolism , Animals , Cell Movement , Cell Proliferation , Chemotaxis , Enzyme-Linked Immunosorbent Assay , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mice , Mice, Inbred C57BL , Mice, SCID , Monocytes/cytology , Monocytes/metabolism , Neoplasm Transplantation , Signal Transduction , Tumor Microenvironment
10.
Leuk Res ; 38(11): 1327-31, 2014 Nov.
Article En | MEDLINE | ID: mdl-25245399

The aim of this study is to evaluate the expression of macrophage inflammatory protein-1α (MIP-1α) in Jurkat cells and its effect on transendothelial migration. In the present study, human acute lymphoblastic leukemia Jurkat cells (Jurkat cells) were used as a model of T cells in human T-cell acute lymphoblastic leukemia (T-ALL), which demonstrated significantly higher MIP-1α expression compared with that in normal T-cell controls. The ability of Jurkat cells to cross a human brain microvascular endothelial cell (HBMEC) monolayer was almost completely abrogated by MIP-1α siRNA. In addition, the overexpression of MIP-1α resulted in the up-regulated expression of endothelial adhesion molecules, which enhanced the migration of Jurkat cells through a monolayer of HBMEC. MIP-1α levels in Jurkat cells appeared to be an important factor for its transendothelial migration, which may provide the theoretical basis to understand the mechanisms of brain metastases of T-ALL at cellular and molecular levels.


Chemokine CCL3/physiology , Intercellular Adhesion Molecule-1/physiology , Transendothelial and Transepithelial Migration/physiology , Up-Regulation , Vascular Cell Adhesion Molecule-1/physiology , Base Sequence , Chemokine CCL3/genetics , DNA Primers , Humans , Jurkat Cells , RNA, Small Interfering , Real-Time Polymerase Chain Reaction
11.
Clin Calcium ; 24(6): 871-8, 2014 Jun.
Article Ja | MEDLINE | ID: mdl-24870838

Multiple myeloma (MM) develops and expands almost exclusively in the bone marrow, and generates devastating bone destruction. MM cells produce a variety of cytokines to stimulate RANK ligand-mediated osteoclastogenesis and suppress osteoblastic differentiation from bone marrow stromal cells, leading to extensive bone destruction with rapid loss of bone. MM cells alter through bone destruction the microenvironment in bone where they colonize, which in turn favors tumor growth and survival, thereby forming a progressive vicious cycle between tumor expansion and bone destruction in MM.


Cytokines/physiology , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Animals , Bone Marrow/pathology , Bone Marrow Cells/pathology , Bone and Bones/pathology , Cell Differentiation , Cellular Microenvironment , Chemokine CCL3/physiology , Chemokine CCL4/physiology , Cytokines/biosynthesis , Disease Progression , Humans , Intercellular Signaling Peptides and Proteins/physiology , Interleukin-17/physiology , Membrane Proteins/physiology , Mice , Multiple Myeloma/metabolism , Osteoblasts/cytology , Osteoclasts/cytology , RANK Ligand/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
12.
J Med Food ; 17(5): 550-7, 2014 May.
Article En | MEDLINE | ID: mdl-24325454

Obesity-induced inflammation is characterized by recruitment of adipose tissue macrophages that release inflammatory cytokines and chemokines. MIP-1α (macrophage inflammatory protein 1α)/CCL3, a CC chemokine, induces monocyte/macrophage infiltration and thus is implicated in obesity-induced adipose inflammation. Quercetin has been shown to modulate obesity-induced inflammation, but the mechanism of its action remains unclear. Here we demonstrate that quercetin decreases MIP-1α release from adipocytes and macrophages and from cocultured adipocytes/macrophages; it also opposes MIP-1α-induced macrophage infiltration and activation. The inhibitory action of quercetin on the MIP-1α-induced inflammatory responses of macrophages is mediated by downregulation of CCR1/CCR5, and inhibition of activation of JNK, p38 mitogen-activated-protein kinase (MAPK), and IKK as well as IκBα degradation. These findings suggest that quercetin may be a useful agent against obesity-induced adipose tissue inflammation.


Adipose Tissue , Chemokine CCL3/antagonists & inhibitors , Inflammation/prevention & control , Quercetin/pharmacology , Receptors, CCR/genetics , 3T3-L1 Cells , Adipocytes/metabolism , Adipose Tissue/chemistry , Animals , Cell Line , Chemokine CCL3/genetics , Chemokine CCL3/physiology , Chemotaxis/drug effects , Coculture Techniques , Culture Media, Conditioned , Down-Regulation/drug effects , Enzyme Activation/drug effects , Inflammation/etiology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , Obesity/complications , RNA, Messenger/analysis , Receptors, CCR1/genetics , Receptors, CCR5/genetics , Signal Transduction/drug effects , p38 Mitogen-Activated Protein Kinases
13.
J Immunol ; 192(3): 886-96, 2014 Feb 01.
Article En | MEDLINE | ID: mdl-24367031

Systemic lupus erythematosus is a chronic inflammatory autoimmune disease, the development of which is characterized by a progressive loss of renal function. Such dysfunction is associated with leukocyte infiltration in the glomerular and tubulointerstitial compartments in both human and experimental lupus nephritis. In this study, we investigated the role of the Ccr1 chemokine receptor in this infiltration process during the progression of nephritis in the lupus-prone New Zealand Black/New Zealand White (NZB/W) mouse model. We found that peripheral T cells, mononuclear phagocytes, and neutrophils, but not B cells, from nephritic NZB/W mice were more responsive to Ccr1 ligands than the leukocytes from younger prenephritic NZB/W mice. Short-term treatment of nephritic NZB/W mice with the orally available Ccr1 antagonist BL5923 decreased renal infiltration by T cells and macrophages. Longer Ccr1 blockade decreased kidney accumulation of effector/memory CD4(+) T cells, Ly6C(+) monocytes, and both M1 and M2 macrophages; reduced tubulointerstitial and glomerular injuries; delayed fatal proteinuria; and prolonged animal lifespan. In contrast, renal humoral immunity was unaffected in BL5923-treated mice, which reflected the unchanged numbers of infiltrated B cells in the kidneys. Altogether, these findings define a pivotal role for Ccr1 in the recruitment of T and mononuclear phagocyte cells to inflamed kidneys of NZB/W mice, which in turn contribute to the progression of renal injury.


Lupus Nephritis/therapy , Myeloid Cells/immunology , Neutrophil Infiltration , Receptors, CCR1/antagonists & inhibitors , T-Lymphocyte Subsets/immunology , Age Factors , Animals , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Chemokine CCL3/biosynthesis , Chemokine CCL3/deficiency , Chemokine CCL3/genetics , Chemokine CCL3/physiology , Chemokine CCL5/biosynthesis , Chemokine CCL5/genetics , Chemokine CCL5/physiology , Chemotaxis, Leukocyte , Disease Progression , Drug Evaluation, Preclinical , Humans , Kidney/immunology , Kidney/pathology , Ligands , Lupus Nephritis/immunology , Lupus Nephritis/pathology , Mice , Mice, Inbred NZB , Myeloid Cells/drug effects , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neutrophil Infiltration/drug effects , RNA, Messenger/biosynthesis , Random Allocation , Receptors, CCR1/biosynthesis , Receptors, CCR1/genetics , Receptors, CCR1/physiology , Spleen/immunology , Spleen/pathology , Splenomegaly/etiology , Splenomegaly/immunology , Splenomegaly/pathology , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
14.
Cell Death Dis ; 4: e764, 2013 Aug 08.
Article En | MEDLINE | ID: mdl-23928703

Despite androgen deprivation therapy (ADT) suppression of prostate cancer (PCa) growth, its overall effects on PCa metastasis remain unclear. Using human (C4-2B/THP1) and mouse (TRAMP-C1/RAW264.7) PCa cells-macrophages co-culture systems, we found currently used anti-androgens, MDV3100 (enzalutamide) or Casodex (bicalutamide), promoted macrophage migration to PCa cells that consequently led to enhanced PCa cell invasion. In contrast, the AR degradation enhancer, ASC-J9, suppressed both macrophage migration and subsequent PCa cell invasion. Mechanism dissection showed that Casodex/MDV3100 reduced the AR-mediated PIAS3 expression and enhanced the pSTAT3-CCL2 pathway. Addition of CCR2 antagonist reversed the Casodex/MDV3100-induced macrophage migration and PCa cell invasion. In contrast, ASC-J9 could regulate pSTAT3-CCL2 signaling using two pathways: an AR-dependent pathway via inhibiting PIAS3 expression and an AR-independent pathway via direct inhibition of the STAT3 phosphorylation/activation. These findings were confirmed in the in vivo mouse model with orthotopically injected TRAMP-C1 cells. Together, these results may raise the potential concern about the currently used ADT with anti-androgens that promotes PCa metastasis and may provide some new and better therapeutic strategies using ASC-J9 alone or a combinational therapy that simultaneously targets androgens/AR signaling and PIAS3-pSTAT3-CCL2 signaling to better battle PCa growth and metastasis at castration-resistant stage.


Androgen Antagonists/pharmacology , Androgen Receptor Antagonists/pharmacology , Anilides/pharmacology , Antineoplastic Agents/pharmacology , Chemokine CCL3/physiology , Curcumin/analogs & derivatives , Nitriles/pharmacology , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms/pathology , STAT3 Transcription Factor/physiology , Tosyl Compounds/pharmacology , Animals , Benzamides , Cell Movement/drug effects , Chemokine CCL3/metabolism , Coculture Techniques , Curcumin/pharmacology , Humans , Macrophages/drug effects , Male , Mice , Neoplasm Metastasis/pathology , Phenylthiohydantoin/pharmacology , Prostatic Neoplasms/drug therapy , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects
15.
PLoS One ; 8(6): e66106, 2013.
Article En | MEDLINE | ID: mdl-23799074

Liver fibrosis is associated with infiltrating immune cells and activation of hepatic stellate cells. We here aimed to investigate the effects of the CC chemokine CCL3, also known as macrophage inflammatory protein-1α, in two different fibrosis models. To this end, we treated mice either with carbon tetrachloride or with a methionine- and choline-deficient diet to induce fibrosis in CCL3 deficient and wild-type mice. The results show that the protein expression of CCL3 is increased in wild-type mice after chronic liver injury. Deletion of CCL3 exhibited reduced liver fibrosis compared to their wild-type counterparts. We could validate these results by treating the two mouse groups with either carbon tetrachloride or by feeding a methionine- and choline-deficient diet. In these models, lack of CCL3 is functionally associated with reduced stellate cell activation and liver immune cell infiltration. In vitro, we show that CCL3 leads to increased proliferation and migration of hepatic stellate cells. In conclusion, our results define the chemokine CCL3 as a mediator of experimental liver fibrosis. Thus, therapeutic modulation of CCL3 might be a promising target for chronic liver diseases.


Chemokine CCL3/physiology , Liver Cirrhosis/physiopathology , Animals , Carbon Tetrachloride/administration & dosage , Cell Proliferation , Enzyme-Linked Immunosorbent Assay , Lipogenesis , Liver Cirrhosis/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Knockout , T-Lymphocytes/cytology
16.
Bone ; 52(1): 259-67, 2013 Jan.
Article En | MEDLINE | ID: mdl-23059626

Bone remodeling is affected by mechanical loading and inflammatory mediators, including chemokines. The chemokine (C-C motif) ligand 3 (CCL3) is involved in bone remodeling by binding to C-C chemokine receptors 1 and 5 (CCR1 and CCR5) expressed on osteoclasts and osteoblasts. Our group has previously demonstrated that CCR5 down-regulates mechanical loading-induced bone resorption. Thus, the present study aimed to investigate the role of CCR1 and CCL3 in bone remodeling induced by mechanical loading during orthodontic tooth movement in mice. Our results showed that bone remodeling was significantly decreased in CCL3(-/-) and CCR1(-/-) mice and in animals treated with Met-RANTES (an antagonist of CCR5 and CCR1). mRNA levels of receptor activator of nuclear factor kappa-B (RANK), its ligand RANKL, tumor necrosis factor alpha (TNF-α) and RANKL/osteoprotegerin (OPG) ratio were diminished in the periodontium of CCL3(-/-) mice and in the group treated with Met-RANTES. Met-RANTES treatment also reduced the levels of cathepsin K and metalloproteinase 13 (MMP13). The expression of the osteoblast markers runt-related transcription factor 2 (RUNX2) and periostin was decreased, while osteocalcin (OCN) was augmented in CCL3(-/-) and Met-RANTES-treated mice. Altogether, these findings show that CCR1 is pivotal for bone remodeling induced by mechanical loading during orthodontic tooth movement and these actions depend, at least in part, on CCL3.


Bone Remodeling/physiology , Chemokine CCL3/physiology , Receptors, CCR1/physiology , Tooth Movement Techniques , Animals , Base Sequence , Biomechanical Phenomena , Chemokine CCL3/genetics , DNA Primers , Mice , Mice, Inbred C57BL , Mice, Knockout , Real-Time Polymerase Chain Reaction , Receptors, CCR1/genetics
17.
Retrovirology ; 9: 101, 2012 Dec 05.
Article En | MEDLINE | ID: mdl-23217137

BACKGROUND: Despite readily detectable levels of the HIV-1 (co)-receptors CD4, CCR5 and DC-SIGN on placental macrophages (Hofbauer Cells [HCs]), the rate of HIV-1 infection in utero in the absence of interventions is only 7% of exposed infants. Here, we examine the replication kinetics of human HCs to the primary isolate HIV-1BaL. We also determined the infectivity of HIV-1-exposed HCs by co-culturing with isolated cord and peripheral blood mononuclear cells [CBMCs, PBMCs]. To understand the limiting nature of HCs to HIV-1 replication, we examined the effect of endogenously secreted cytokines on replication kinetics. RESULTS: HCs have reduced ability to replicate HIV-1 in vitro (p < 0.01) and to transmit virus to CBMCs and PBMCs (p < 0.001 for both) compared to standard infections of MDMs. HCs were shown to release HIV-1 particles at levels comparable to MDMs, however exhibit significant decreases in viral transcription (gag and env), which may account for lower levels of HIV-1 replication. Un-stimulated HCs constitutively express significantly higher levels of regulatory cytokines, IL-10 and TGF-ß, compared to MDMs (p < 0.01), which may contribute to immunoregulatory predominance at the placenta and possibly account for down-regulation of HIV-1 replication and infectivity by HCs. We further demonstrate that these regulatory cytokines inhibit HIV-1 replication within HCs in vitro. CONCLUSION: HCs have reduced ability to replicate and disseminate R5-tropic HIV-1BaLin vitro and potentially offset mother to child transmission (MTCT) of HIV-1 by the induction of immunoregulatory cytokines. Despite the potential for migration and infectivity, HCs are not present in the neighboring fetal circulation. These results implicate HCs as important mediators of protection at the feto-maternal interface during ongoing HIV-1 exposure.


Acquired Immunodeficiency Syndrome/transmission , Cytokines/biosynthesis , HIV-1/physiology , Infectious Disease Transmission, Vertical/prevention & control , Macrophages/physiology , Placenta/virology , Virus Replication , Cell Membrane/virology , Chemokine CCL3/physiology , Chemokine CCL4/physiology , Female , Humans , Placenta/cytology , Pregnancy , Receptors, CCR5/physiology , Virus Assembly
18.
Blood ; 120(4): 880-90, 2012 Jul 26.
Article En | MEDLINE | ID: mdl-22674804

Initial observations suggested that C-C motif chemokines exclusively mediate chemotaxis of mononuclear cells. In addition, recent studies also implicated these chemotactic cytokines in the recruitment of neutrophils. The underlying mechanisms remained largely unknown. Using in vivo microscopy on the mouse cremaster muscle, intravascular adherence and subsequent paracellular transmigration of neutrophils elicited by the chemokine (C-C motif) ligand 3 (CCL3, synonym MIP-1α) were significantly diminished in mice with a deficiency of the chemokine (C-C motif) receptor 1 (Ccr1(-/-)) or 5 (Ccr5(-/-)). Using cell-transfer techniques, neutrophil responses required leukocyte CCR1 and nonleukocyte CCR5. Furthermore, neutrophil extravasation elicited by CCL3 was almost completely abolished on inhibition of G protein-receptor coupling and PI3Kγ-dependent signaling, while neutrophil recruitment induced by the canonical neutrophil attractants chemokine (C-X-C motif) ligand 1 (CXCL1, synonym KC) or the lipid mediator platetelet-activating factor (PAF) was only partially reduced. Moreover, Ab blockade of ß(2) integrins, of α(4) integrins, or of their putative counter receptors ICAM-1 and VCAM-1 significantly attenuated CCL3-, CXCL1-, or PAF-elicited intravascular adherence and paracellular transmigration of neutrophils. These data indicate that the C-C motif chemokine CCL3 and canonical neutrophil attractants exhibit both common and distinct mechanisms for the regulation of intravascular adherence and transmigration of neutrophils.


Cell Movement , Chemokine CCL3/physiology , Chemotaxis, Leukocyte/physiology , Neutrophils/metabolism , Animals , Carrier Proteins/metabolism , Cells, Cultured , Chemokine CCL2/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Flow Cytometry , Integrins/metabolism , Intercellular Adhesion Molecule-1/metabolism , Male , Mice , Mice, Inbred BALB C , Neutrophils/cytology , Receptors, CCR1/metabolism , Receptors, CCR5/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
19.
Cancer Metastasis Rev ; 31(3-4): 569-78, 2012 Dec.
Article En | MEDLINE | ID: mdl-22706844

Bone is one of the most frequent sites of metastasis in patients with malignancies. Up to 90 % of patients with multiple myeloma, and 60 % to 75 % patients with prostate cancer and breast cancer develop bone metastasis at the later stages of their diseases. Bone metastases are responsible for tremendous morbidity in patients with cancer, including severe bone pain, pathologic fractures, spinal cord and nerve compression syndromes, life-threatening hypercalcemia, and increased mortality. Multiple factors produced by tumor cells or produced by the bone marrow microenvironment in response to tumor cells play important roles in activation of osteoclastic bone resorption and modulation of osteoblastic activity in patients with bone metastasis. In this chapter, we will review the genes that play important roles in bone destruction, tumor growth, and osteoblast activity in bone metastasis and discuss the potential therapies targeting the products of these genes to block both bone destruction and tumor growth.


Bone Neoplasms/secondary , Activins/physiology , Animals , Annexin A2/physiology , Bone Neoplasms/therapy , Chemokine CCL3/physiology , Humans , Interleukin-6/physiology , Osteoblasts/physiology , Osteolysis , Osteoprotegerin/physiology , Parathyroid Hormone-Related Protein/physiology , RANK Ligand/physiology
20.
PLoS One ; 7(2): e31508, 2012.
Article En | MEDLINE | ID: mdl-22359597

Macrophage inflammatory protein-1α (CCL3) plays a well-known role in infectious and viral diseases; however, its contribution to atherosclerotic lesion formation and lipid metabolism has not been determined. Low density lipoprotein receptor deficient (LDLR(-/-)) mice were transplanted with bone marrow from CCL3(-/-) or C57BL/6 wild type donors. After 6 and 12 weeks on western diet (WD), recipients of CCL3(-/-) marrow demonstrated lower plasma cholesterol and triglyceride concentrations compared to recipients of C57BL/6 marrow. Atherosclerotic lesion area was significantly lower in female CCL3(-/-) recipients after 6 weeks and in male CCL3(-/-) recipients after 12 weeks of WD feeding (P<0.05). Surprisingly, male CCL3(-/-) recipients had a 50% decrease in adipose tissue mass after WD-feeding, and plasma insulin, and leptin levels were also significantly lower. These results were specific to CCL3, as LDLR(-/-) recipients of monocyte chemoattractant protein(-/-) (CCL2) marrow were not protected from the metabolic consequences of high fat feeding. Despite these improvements in LDLR(-/-) recipients of CCL3(-/-) marrow in the bone marrow transplantation (BMT) model, double knockout mice, globally deficient in both proteins, did not have decreased body weight, plasma lipids, or atherosclerosis compared with LDLR(-/-) controls. Finally, there were no differences in myeloid progenitors or leukocyte populations, indicating that changes in body weight and plasma lipids in CCL3(-/-) recipients was not due to differences in hematopoiesis. Taken together, these data implicate a role for CCL3 in lipid metabolism in hyperlipidemic mice following hematopoietic reconstitution.


Adipose Tissue/pathology , Atherosclerosis/pathology , Chemokine CCL3/deficiency , Fatty Liver/etiology , Animals , Atherosclerosis/etiology , Body Weight , Bone Marrow Transplantation , Chemokine CCL3/physiology , Diet , Female , Hyperlipidemias , Lipid Metabolism , Lipids/blood , Male , Mice , Mice, Knockout
...