Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 387
Filter
Add more filters

Publication year range
1.
Nature ; 566(7744): 383-387, 2019 02.
Article in English | MEDLINE | ID: mdl-30760925

ABSTRACT

Sleep is integral to life1. Although insufficient or disrupted sleep increases the risk of multiple pathological conditions, including cardiovascular disease2, we know little about the cellular and molecular mechanisms by which sleep maintains cardiovascular health. Here we report that sleep regulates haematopoiesis and protects against atherosclerosis in mice. We show that mice subjected to sleep fragmentation produce more Ly-6Chigh monocytes, develop larger atherosclerotic lesions and produce less hypocretin-a stimulatory and wake-promoting neuropeptide-in the lateral hypothalamus. Hypocretin controls myelopoiesis by restricting the production of CSF1 by hypocretin-receptor-expressing pre-neutrophils in the bone marrow. Whereas hypocretin-null and haematopoietic hypocretin-receptor-null mice develop monocytosis and accelerated atherosclerosis, sleep-fragmented mice with either haematopoietic CSF1 deficiency or hypocretin supplementation have reduced numbers of circulating monocytes and smaller atherosclerotic lesions. Together, these results identify a neuro-immune axis that links sleep to haematopoiesis and atherosclerosis.


Subject(s)
Atherosclerosis/prevention & control , Hematopoiesis/physiology , Sleep/physiology , Animals , Antigens, Ly/metabolism , Atherosclerosis/metabolism , Atherosclerosis/pathology , Bone Marrow Cells/metabolism , Female , Hematopoiesis/drug effects , Hypothalamic Area, Lateral/metabolism , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/deficiency , Macrophage Colony-Stimulating Factor/metabolism , Male , Mice , Monocytes/drug effects , Monocytes/metabolism , Myelopoiesis/drug effects , Neutrophils/metabolism , Orexin Receptors/deficiency , Orexin Receptors/metabolism , Orexins/biosynthesis , Orexins/deficiency , Orexins/metabolism , Orexins/pharmacology , Sleep/drug effects , Sleep Deprivation/metabolism , Sleep Deprivation/physiopathology , Sleep Deprivation/prevention & control
2.
PLoS Pathog ; 12(12): e1006046, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27923070

ABSTRACT

Dynamic regulation of leukocyte population size and activation state is crucial for an effective immune response. In malaria, Plasmodium parasites elicit robust host expansion of macrophages and monocytes, but the underlying mechanisms remain unclear. Here we show that myeloid expansion during P. chabaudi infection is dependent upon both CD4+ T cells and the cytokine Macrophage Colony Stimulating Factor (MCSF). Single-cell RNA-Seq analysis on antigen-experienced T cells revealed robust expression of Csf1, the gene encoding MCSF, in a sub-population of CD4+ T cells with distinct transcriptional and surface phenotypes. Selective deletion of Csf1 in CD4+ cells during P. chabaudi infection diminished proliferation and activation of certain myeloid subsets, most notably lymph node-resident CD169+ macrophages, and resulted in increased parasite burden and impaired recovery of infected mice. Depletion of CD169+ macrophages during infection also led to increased parasitemia and significant host mortality, confirming a previously unappreciated role for these cells in control of P. chabaudi. This work establishes the CD4+ T cell as a physiologically relevant source of MCSF in vivo; probes the complexity of the CD4+ T cell response during type 1 infection; and delineates a novel mechanism by which T helper cells regulate myeloid cells to limit growth of a blood-borne intracellular pathogen.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/immunology , Malaria/immunology , Animals , CD4-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Fluorescent Antibody Technique , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Plasmodium chabaudi/immunology , Polymerase Chain Reaction
3.
Blood ; 127(9): 1202-5, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26738538

ABSTRACT

Natural killer (NK) cells exist as subsets based on expression of inhibitory receptors that recognize major histocompatibility complex I (MHCI) molecules. NK cell subsets bearing MHCI binding receptors for self-MHCI have been termed as "licensed" and exhibit a higher ability to respond to stimuli. In the context of bone marrow transplantation (BMT), host licensed-NK (L-NK) cells have also been demonstrated to be responsible for the acute rejection of allogeneic and MHCI-deficient BM cells (BMCs) in mice after lethal irradiation. However, the role of recipient unlicensed-NK (U-NK) cells has not been well established with regard to allogeneic BMC resistance. After NK cell stimulation, the prior depletion of host L-NK cells resulted in a marked increase of donor engraftment compared with the untreated group. Surprisingly, this increased donor engraftment was reduced after total host NK cell depletion, indicating that U-NK cells can actually promote donor allogeneic BMC engraftment. Furthermore, direct coculture of U-NK cells with allogeneic but not syngeneic BMCs resulted in increased colony-forming unit cell growth in vitro, which was at least partially mediated by granulocyte macrophage colony-stimulating factor (GM-CSF) production. These data demonstrate that host NK cell subsets exert markedly different roles in allogeneic BMC engraftment where host L- and U-NK cells reject or promote donor allogeneic BMC engraftment, respectively.


Subject(s)
Bone Marrow Transplantation , Killer Cells, Natural/immunology , Animals , Female , Histocompatibility Antigens Class I/metabolism , Macrophage Colony-Stimulating Factor/biosynthesis , Mice, Inbred BALB C , Mice, Inbred C57BL , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Transplantation, Homologous
4.
Eur J Immunol ; 46(3): 552-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26634935

ABSTRACT

IL-34 and colony-stimulating factor 1 (CSF1) are two alternative ligands for the CSF1 receptor that play nonredundant roles in the development, survival, and function of tissue macrophages and Langerhans cells (LCs). In this study, we investigated the spatio-temporal production of IL-34 and its impact on skin LCs in the developing embryo and adult mice in the steady state and during inflammation using Il34(LacZ) reporter mice and newly generated inducible Il34-knockout mice. We found that IL-34 is produced in the developing skin epidermis of the embryo, where it promotes the final differentiation of LC precursors. In adult life, LCs required IL-34 to continually self-renew in the steady state. However, during UV-induced skin damage, LC regeneration depended on neutrophils infiltrating the skin, which produced large amounts of CSF1. We conclude that LCs require IL-34 when residing in fully differentiated and anatomically intact skin epidermis, but rely on neutrophil-derived CSF1 during inflammation. Our demonstration that neutrophils are an important source of CSF1 during skin inflammation may exemplify a mechanism through which neutrophils promote their subsequent replacement with mononuclear phagocytes.


Subject(s)
Inflammation/immunology , Interleukins/biosynthesis , Interleukins/immunology , Keratinocytes/immunology , Langerhans Cells/physiology , Macrophage Colony-Stimulating Factor/immunology , Neutrophils/immunology , Animals , Cell Differentiation , Embryo, Mammalian/immunology , Epidermal Cells , Epidermis/embryology , Epidermis/immunology , Epidermis/physiology , Interleukins/genetics , Interleukins/physiology , Langerhans Cells/immunology , Langerhans Cells/pathology , Macrophage Colony-Stimulating Factor/biosynthesis , Mice , Mice, Knockout , Neutrophils/physiology , Ultraviolet Rays
5.
Am J Pathol ; 186(5): 1258-66, 2016 05.
Article in English | MEDLINE | ID: mdl-26970370

ABSTRACT

During human pregnancy, immune tolerance of the fetal semiallograft occurs in the presence of abundant maternal leukocytes. At the implantation site, macrophages comprise approximately 20% of the leukocyte population and act as primary mediators of tissue remodeling. Decidual macrophages display a balance between anti-inflammatory and proinflammatory phenotypes. However, a shift to an M1 subtype is reported in preeclampsia. Granulocyte-macrophage colony-stimulating-factor (GM-CSF) and macrophage colony-stimulating factor (M-CSF) are major differentiating factors that mediate M1 and M2 polarization, respectively. Previously, we observed the following: i) the preeclamptic decidua contains an excess of both macrophages and GM-CSF, ii) the preeclampsia-associated proinflammatory cytokines, IL-1ß and tumor necrosis factor-α, markedly enhance GM-CSF and M-CSF expression in cultured leukocyte-free first-trimester decidual cells (FTDCs), iii) FTDC-secreted GM-CSF polarizes macrophages toward an M1 subtype. The microenvironment is a key determinant of macrophage phenotype. Thus, we examined proinflammatory stimulation of FTDC-secreted M-CSF and its role in macrophage development. Immunofluorescence staining demonstrated elevated M-CSF-positive decidual cell numbers in preeclamptic decidua. In FTDCs, IL-1ß and tumor necrosis factor-α signal through the NF-κB pathway to induce M-CSF production, which does the following: i) enhances differentiation of and elevates CD163 expression in macrophages, ii) increases macrophage phagocytic capacity, and iii) inhibits signal-regulatory protein α expression by macrophages. These findings suggest that FTDC-secreted M-CSF modulates the decidual immune balance by inducing M2 macrophage polarization and phagocytic capacity in response to proinflammatory stimuli.


Subject(s)
Decidua/immunology , Macrophage Colony-Stimulating Factor/physiology , Pre-Eclampsia/immunology , Antigens, Differentiation/metabolism , Cell Differentiation/immunology , Cells, Cultured , Decidua/metabolism , Female , Humans , Interleukin-1beta/physiology , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/metabolism , NF-kappa B/metabolism , Phagocytosis/immunology , Pregnancy , Pregnancy Trimester, First , Receptors, Immunologic/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/physiology
6.
J Immunol ; 194(11): 5211-22, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25917103

ABSTRACT

Cannabidiol (CBD) is a natural nonpsychotropic cannabinoid from marijuana (Cannabis sativa) with anti-epileptic and anti-inflammatory properties. Effect of CBD on naive immune system is not precisely understood. In this study, we observed that administering CBD into naive mice triggers robust induction of CD11b(+)Gr-1(+) myeloid-derived suppressor cells (MDSC) in the peritoneum, which expressed functional arginase 1, and potently suppressed T cell proliferation ex vivo. Furthermore, CBD-MDSC suppressed LPS-induced acute inflammatory response upon adoptive transfer in vivo. CBD-induced suppressor cells were comprised of CD11b(+)Ly6-G(+)Ly6-C(+) granulocytic and CD11b(+)Ly6-G(-)Ly6-C(+) monocytic subtypes, with monocytic MDSC exhibiting higher T cell-suppressive function. Induction of MDSC by CBD was markedly attenuated in Kit-mutant (Kit(W/W-v)) mast cell-deficient mice. MDSC response was reconstituted upon transfer of wild-type bone marrow-derived mast cells in Kit(W/W-v) mice, suggesting the key role of cKit (CD117) as well as mast cells. Moreover, mast cell activator compound 48/80 induced significant levels of MDSC in vivo. CBD administration in mice induced G-CSF, CXCL1, and M-CSF, but not GM-CSF. G-CSF was found to play a key role in MDSC mobilization inasmuch as neutralizing G-CSF caused a significant decrease in MDSC. Lastly, CBD enhanced the transcriptional activity of peroxisome proliferator-activated receptor γ in luciferase reporter assay, and PPAR-γ selective antagonist completely inhibited MDSC induction in vivo, suggesting its critical role. Together, the results suggest that CBD may induce activation of PPAR-γ in mast cells leading to secretion of G-CSF and consequent MDSC mobilization. CBD being a major component of Cannabis, our study indicates that marijuana may modulate or dysregulate the immune system by mobilizing MDSC.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cannabidiol/pharmacology , Cannabis/metabolism , Myeloid Cells/immunology , PPAR gamma/genetics , Animals , Arginase/biosynthesis , CD11b Antigen/metabolism , Cell Proliferation/drug effects , Chemokine CXCL1/biosynthesis , Female , Granulocyte Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/biosynthesis , Mast Cells , Mice , Mice, Inbred C57BL , Mice, Knockout , Myeloid Cells/metabolism , PPAR gamma/antagonists & inhibitors , Receptors, Chemokine/metabolism , T-Lymphocytes/immunology , Transcriptional Activation/genetics , p-Methoxy-N-methylphenethylamine/pharmacology
7.
Anaesthesia ; 72(7): 879-888, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28407211

ABSTRACT

Optimal immunological homoeostasis determines the long-term recovery of patients in the postoperative period. The functional adaptability of monocytes plays a pivotal role in adjusting the host's response to an insult, immunostasis and long-term health, and may help to determine successful recovery. We undertook a longitudinal analysis of the functional adaptability of monocytes in 20 patients undergoing heart surgery with cardiopulmonary bypass, as a model of severe stress. Using each patient's pre-cardiopulmonary bypass data as a baseline, we investigated the characteristics of peripheral blood monocytes' functional plasticity in-vitro before elective bypass, and three months afterwards. Approximately 30% of subjects showed diminished monocyte plasticity, as demonstrated by decreased monocyte differentiation into dendritic cells three months after bypass. Diminished monocyte functional plasticity was related to over-production of macrophage colony-stimulating factor. Adding a neutralising antibody to macrophage colony-stimulating factor corrected the monocytes' differentiation defect. Finally, patients with reduced monocyte plasticity had significantly elevated serum C-reactive protein, with a concomitant increase in cytomegalovirus IgG antibody titres, suggestive of the acquisition of immuno-suppressive traits. Our study shows that severe surgical stress resulted in a lasting immunological defect in individuals who had seemingly recovered.


Subject(s)
Cardiac Surgical Procedures/adverse effects , Elective Surgical Procedures/adverse effects , Monocytes/physiology , Aged , Aged, 80 and over , Female , Humans , Lectins, C-Type/analysis , Macrophage Colony-Stimulating Factor/biosynthesis , Male , Mannose Receptor , Mannose-Binding Lectins/analysis , Middle Aged , Receptors, Cell Surface/analysis
8.
J Neurovirol ; 21(3): 249-56, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25060299

ABSTRACT

Combination antiretroviral therapy (cART) has improved the longevity and quality of life for people living with HIV; however, it does not target virus that persists in long-lived cells, such as macrophages (MΦs). This allows for the development of viral reservoirs in various anatomical compartments where these cells reside, including the central nervous system (CNS), where perivascular MΦs and resident microglia constitute the principle cellular reservoir of HIV. How HIV persists in MΦs/microglia is not completely understood; however, prosurvival signaling that protects infected MΦs/microglia from apoptosis is likely important to viral persistence. Macrophage colony-stimulating factor (M-CSF) is an important factor in MΦ survival and has been implicated in HIV neuropathogenesis through its ability to enhance the susceptibility of MΦs to infection and promote virus production. While M-CSF has been detected in cerebrospinal fluid of HIV-infected patients, the cellular source of M-CSF in the CNS is unknown. Here, we demonstrate, for the first time, that MΦs comprising perivascular cuffs and nodular lesions in SIV encephalitis (SIVE) brain are the principle source of M-CSF. These cells also serve as the primary reservoir of productive SIV infection in the brain. We further demonstrate that M-CSF and IL-34, which signal through the same receptor, cFMS, enhance HIV-1 production by microglia in vitro. This is attenuated by the addition of a receptor tyrosine kinase inhibitor with high specificity for cFMS, GW2580. Together, these data suggest that cFMS signaling may be an attractive target for eliminating long-lived MΦ reservoirs of HIV in the brain and other tissues.


Subject(s)
AIDS Dementia Complex/virology , Brain/virology , Disease Reservoirs/virology , Microglia/virology , Simian Acquired Immunodeficiency Syndrome/virology , AIDS Dementia Complex/immunology , Animals , Brain/immunology , HIV-1/immunology , Humans , Immunohistochemistry , In Situ Hybridization , Interleukins/immunology , Macaca mulatta , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/immunology , Macrophages/immunology , Macrophages/virology , Microglia/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Signal Transduction/physiology , Simian Acquired Immunodeficiency Syndrome/immunology
9.
Tumour Biol ; 36(7): 5361-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25854167

ABSTRACT

Pigmented villonodular synovitis (PVNS) is a benign, translocation-derived neoplasm. Because of its high local recurrence rate after surgery and occurrence of osteochondral destruction, a novel therapeutic target is required. The present study aimed to evaluate the significance of protein expression possibly associated with the pathogenesis during the clinical course of PVNS. In 40 cases of PVNS, positivity of colony-stimulated factor 1 (CSF1), its receptor (CSF1R), and receptor activator of nuclear factor kappa-B ligand (RANKL) were immunohistochemically determined. The relationship between the positivity and clinical outcomes was investigated. High positivity of CSF1 staining intensity was associated with an increased incidence of osteochondral lesions (bone erosion and osteoarthritis) (p = 0.009), but not with the rate of local recurrence. Positivity of CSF1R and RANKL staining was not associated with any clinical variables. The number of giant cells was not correlated with positivity of any of the three proteins, or with the clinical outcome. Focusing on knee cases, CSF1 positivity was also associated with the incidence of osteochondal change (p = 0.02). CSF1R positivity was high in cases which had local recurrence, but not significantly so (p = 0.129). Determination of CSF1 and CSF1R expression may be useful as a prognosticator of the clinical course and/or outcomes of PVNS.


Subject(s)
Macrophage Colony-Stimulating Factor/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Synovitis, Pigmented Villonodular/genetics , Adolescent , Adult , Child , Female , Gene Expression Regulation, Neoplastic , Humans , Macrophage Colony-Stimulating Factor/genetics , Male , Middle Aged , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteoarthritis/surgery , Prognosis , RANK Ligand/biosynthesis , RANK Ligand/genetics , Receptor, Macrophage Colony-Stimulating Factor/genetics , Synovitis, Pigmented Villonodular/pathology , Synovitis, Pigmented Villonodular/surgery , Treatment Outcome
10.
J Biol Chem ; 288(34): 24503-17, 2013 Aug 23.
Article in English | MEDLINE | ID: mdl-23821550

ABSTRACT

Skeletal remodeling consists of timely formation and resorption of bone by osteoblasts and osteoclasts in a quantitative manner. Patients with chronic myeloid leukemia receiving inhibitors of c-Abl tyrosine kinase often show reduced bone remodeling due to impaired osteoblast and osteoclast function. BMP-2 plays a significant role in bone generation and resorption by contributing to the formation of mature osteoblasts and osteoclasts. The effects of c-Abl on BMP-2-induced bone remodeling and the underlying mechanisms are not well studied. Using a pharmacological inhibitor and expression of a dominant negative mutant of c-Abl, we show an essential role of this tyrosine kinase in the development of bone nodules containing mature osteoblasts and formation of multinucleated osteoclasts in response to BMP-2. Calvarial osteoblasts prepared from c-Abl null mice showed the absolute requirement of this tyrosine kinase in maturation of osteoblasts and osteoclasts. Activation of phosphatidylinositol 3-kinase (PI 3-kinase)/Akt signaling by BMP-2 leads to osteoblast differentiation. Remarkably, inhibition of c-Abl significantly suppressed BMP-2-stimulated PI 3-kinase activity and its downstream Akt phosphorylation. Interestingly, c-Abl regulated BMP-2-induced osteoclastogenic CSF-1 expression. More importantly, we identified the requirements of c-Abl in BMP-2 autoregulation and the expressions of alkaline phosphatase and osterix that are necessary for osteoblast differentiation. c-Abl contributed to BMP receptor-specific Smad-dependent transcription of CSF-1, osterix, and BMP-2. Finally, c-Abl associates with BMP receptor IA and regulates phosphorylation of Smad in response to BMP-2. We propose that activation of c-Abl is an important step, which induces into two signaling pathways involving noncanonical PI 3-kinase and canonical Smads to integrate BMP-2-induced osteogenesis.


Subject(s)
Bone Morphogenetic Protein 2/metabolism , Osteoblasts/metabolism , Osteogenesis/physiology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Smad5 Protein/metabolism , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/genetics , Animals , Bone Morphogenetic Protein 2/genetics , Cell Line , Gene Expression Regulation/physiology , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/genetics , Mice , Osteoblasts/cytology , Phosphatidylinositol 3-Kinases/genetics , Proto-Oncogene Proteins c-abl/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/physiology , Skull/cytology , Skull/metabolism , Smad5 Protein/genetics , Sp7 Transcription Factor , Transcription Factors/biosynthesis , Transcription Factors/genetics
11.
J Cell Biochem ; 115(8): 1412-9, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24700654

ABSTRACT

Human skeletal aging is characterized as a gradual loss of bone mass due to an excess of bone resorption not balanced by new bone formation. Using human marrow cells, we tested the hypothesis that there is an age-dependent increase in osteoclastogenesis due to intrinsic changes in regulatory factors [macrophage-colony stimulating factor (M-CSF), receptor activator of NF-κB ligand (RANKL), and osteoprotegerin (OPG)] and their receptors [c-fms and RANK]. In bone marrow cells (BMCs), c-fms (r = 0.61, P = 0.006) and RANK expression (r = 0.59, P = 0.008) were increased with age (27-82 years, n = 19). In vitro generation of osteoclasts was increased with age (r = 0.89, P = 0.007). In enriched marrow stromal cells (MSCs), constitutive expression of RANKL was increased with age (r = 0.41, P = 0.049) and expression of OPG was inversely correlated with age (r = -0.43, P = 0.039). Accordingly, there was an age-related increase in RANKL/OPG (r = 0.56, P = 0.005). These data indicate an age-related increase in human osteoclastogenesis that is associated with an intrinsic increase in expression of c-fms and RANK in osteoclast progenitors, and, in the supporting MSCs, an increase in pro-osteoclastogenic RANKL expression and a decrease in anti-osteoclastogenic OPG. These findings support the hypothesis that human marrow cells and their products can contribute to skeletal aging by increasing the generation of bone-resorbing osteoclasts. These findings help to explain underlying molecular mechanisms of progressive bone loss with advancing age in humans.


Subject(s)
Aging/metabolism , Cell Differentiation/genetics , Gene Expression Regulation, Developmental/genetics , Osteoclasts/metabolism , Aging/pathology , Bone Marrow Cells/metabolism , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Humans , Macrophage Colony-Stimulating Factor/biosynthesis , Osteoclasts/pathology , Osteogenesis/genetics , Osteoprotegerin/biosynthesis , RANK Ligand/biosynthesis , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/biosynthesis , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Stromal Cells/metabolism
12.
Breast Cancer Res ; 16(3): R54, 2014 May 29.
Article in English | MEDLINE | ID: mdl-24886617

ABSTRACT

INTRODUCTION: Although C-X-C motif chemokine 12 (CXCL12) has been shown to bind to C-X-C chemokine receptor type 7 (CXCR7), the exact molecular mechanism regulations by CXCL12/CXCR7 axis in breast tumor growth and metastasis are not well understood. CXCR7 expression has been shown to be upregulated during pathological processes such as inflammation and cancer. METHODS: Breast cancer cell lines were genetically silenced or pharmacologically inhibited for CXCR7 and/or its downstream target signal transducer and activator of transcription 3 (STAT3). 4T1 or 4T1 downregulated for CXCR7 and 4T1.2 breast cancer cell lines were injected in mammary gland of BALB/c mice to form tumors, and the molecular pathways regulating tumor growth and metastasis were assessed. RESULTS: In this study, we observed that CXCL12 enhances CXCR7-mediated breast cancer migration. Furthermore, genetic silencing or pharmacologic inhibition of CXCR7 reduced breast tumor growth and metastasis. Further elucidation of mechanisms revealed that CXCR7 mediates tumor growth and metastasis by activating proinflammatory STAT3 signaling and angiogenic markers. Furthermore, enhanced breast tumorigenicity and invasiveness were associated with macrophage infiltration. CXCR7 recruits tumor-promoting macrophages (M2) to the tumor site through regulation of the macrophage colony-stimulating factor (M-CSF)/macrophage colony-stimulating factor receptor (MCSF-R) signaling pathway. In addition, CXCR7 regulated breast cancer metastasis by enhancing expression of metalloproteinases (MMP-9, MMP-2) and vascular cell-adhesion molecule-1 (VCAM-1). We also observed that CXCR7 is highly expressed in invasive ductal carcinoma (IDC) and metastatic breast tissue in human patient samples. In addition, high CXCR7 expression in tumors correlates with worse prognosis for both overall survival and lung metastasis-free survival in IDC patients. CONCLUSION: These observations reveal that CXCR7 enhances breast cancer growth and metastasis via a novel pathway by modulating the tumor microenvironment. These findings identify CXCR7-mediated STAT3 activation and modulation of the tumor microenvironment as novel regulation of breast cancer growth and metastasis. These studies indicate that new strategies using CXCR7 inhibitors could be developed for antimetastatic therapy.


Subject(s)
Breast Neoplasms/pathology , Chemokine CXCL12/metabolism , Lung Neoplasms/secondary , Receptors, CXCR/metabolism , STAT3 Transcription Factor/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Macrophage Activation/genetics , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophages/immunology , Matrix Metalloproteinase 2/biosynthesis , Matrix Metalloproteinase 9/biosynthesis , Mice , Mice, Inbred BALB C , Neoplasm Invasiveness/genetics , Neoplasm Transplantation , Protein Binding , RNA Interference , RNA, Small Interfering , Receptor, Macrophage Colony-Stimulating Factor/biosynthesis , Receptors, CXCR/antagonists & inhibitors , Receptors, CXCR/biosynthesis , Receptors, CXCR/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , Tumor Microenvironment , Vascular Cell Adhesion Molecule-1/biosynthesis
13.
FASEB J ; 27(3): 1223-35, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23233531

ABSTRACT

Breast cancer is often fatal during its metastatic dissemination. To unravel the role of microRNAs (miRs) during malignancy, we analyzed miR expression in 77 primary breast carcinomas and identified 16 relapse-associated miRs that correlate with survival and/or distinguish tumor subtypes in different datasets. Among them, miR-148b, down-regulated in aggressive breast tumors, was found to be a major coordinator of malignancy. In fact, it is able to oppose various steps of tumor progression when overexpressed in cell lines by influencing invasion, survival to anoikis, extravasation, lung metastasis formation, and chemotherapy response. miR-148b controls malignancy by coordinating a novel pathway involving over 130 genes and, in particular, it directly targets players of the integrin signaling, such as ITGA5, ROCK1, PIK3CA/p110α, and NRAS, as well as CSF1, a growth factor for stroma cells. Our findings reveal the importance of the identified 16 miRs for disease outcome predictions and suggest a critical role for miR-148b in the control of breast cancer progression.


Subject(s)
Breast Neoplasms/metabolism , Integrin alpha5/biosynthesis , Macrophage Colony-Stimulating Factor/biosynthesis , MicroRNAs/metabolism , Oncogene Protein p21(ras)/biosynthesis , Phosphatidylinositol 3-Kinases/biosynthesis , RNA, Neoplasm/metabolism , rho-Associated Kinases/biosynthesis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Class I Phosphatidylinositol 3-Kinases , Disease Progression , Female , Humans , Integrin alpha5/genetics , Macrophage Colony-Stimulating Factor/genetics , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Oncogene Protein p21(ras)/genetics , Phosphatidylinositol 3-Kinases/genetics , RNA, Neoplasm/genetics , rho-Associated Kinases/genetics
14.
J Cell Sci ; 124(Pt 13): 2120-31, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21670198

ABSTRACT

We have shown previously that distinct Mena isoforms are expressed in invasive and migratory tumor cells in vivo and that the invasion isoform (Mena(INV)) potentiates carcinoma cell metastasis in murine models of breast cancer. However, the specific step of metastatic progression affected by this isoform and the effects on metastasis of the Mena11a isoform, expressed in primary tumor cells, are largely unknown. Here, we provide evidence that elevated Mena(INV) increases coordinated streaming motility, and enhances transendothelial migration and intravasation of tumor cells. We demonstrate that promotion of these early stages of metastasis by Mena(INV) is dependent on a macrophage-tumor cell paracrine loop. Our studies also show that increased Mena11a expression correlates with decreased expression of colony-stimulating factor 1 and a dramatically decreased ability to participate in paracrine-mediated invasion and intravasation. Our results illustrate the importance of paracrine-mediated cell streaming and intravasation on tumor cell dissemination, and demonstrate that the relative abundance of Mena(INV) and Mena11a helps to regulate these key stages of metastatic progression in breast cancer cells.


Subject(s)
Breast Neoplasms/metabolism , Cell Movement , Cytoskeletal Proteins/metabolism , Transendothelial and Transepithelial Migration , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophages/metabolism , Mice , Mice, SCID , Microfilament Proteins , Neoplasm Invasiveness , Neoplasm Metastasis , Protein Isoforms/metabolism , Rats
15.
Protein Expr Purif ; 88(1): 93-7, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23246714

ABSTRACT

Colony Stimulating Factor-1 (CSF-1) is involved in proliferation, differentiation, and survival of the mononuclear lineage, in development of the female reproductive system and mammary glands during pregnancy and lactation. It is also implicated in the biology of breast cancer and promotion of its metastasis to bones. Therefore, CSF-1 is required for many applications in cellular and molecular biology studies. Commercial products, usually expressed in prokaryotic systems, are costly, with the likelihood of endotoxin contamination and also lack posttranslational modifications. These considerations provide the rationale to express growth factors in eukaryotic systems. In this study, the biologically active and soluble fragment (residues 33-182) of human (CSF-1) was cloned from K562 cell line and expressed in Pichia pastoris. The expression level of the active CSF-1 was about 100 µg/ml of the P. pastoris culture medium. Protein analysis revealed that the expressed CSF-1 appears in three bands with apparent molecular weight of 30, 26 and 20 kDa constituting 44%, 25% and 13% of all proteins in the culture medium, respectively. The expressed protein was partially purified and concentrated (10x) by ultrafiltration, then filter sterilized. The product was confirmed to be biologically active by stimulation of its receptor (FMS) autophosphorylation in THP-1 cells and also growth promotion of factor dependent FDC-P1 cells expressing human wild-type FMS (FD-FMS-WT). Therefore, P. pastoris is a highly efficient and cost-effective expression system for production of endotoxin-free CSF-1 for research and potentially for therapeutic applications.


Subject(s)
Gene Expression , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/isolation & purification , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Bioreactors , Cell Line , Cloning, Molecular , Humans , Macrophage Colony-Stimulating Factor/genetics , Phosphorylation , Pichia
16.
Mol Cancer ; 11: 58, 2012 Aug 21.
Article in English | MEDLINE | ID: mdl-22909061

ABSTRACT

BACKGROUND: Colony stimulating factor-1 (CSF-1) plays an important role in ovarian cancer biology and as a prognostic factor in ovarian cancer. Elevated levels of CSF-1 promote progression of ovarian cancer, by binding to CSF-1R (the tyrosine kinase receptor encoded by c-fms proto-oncogene).Post-transcriptional regulation of CSF-1 mRNA by its 3' untranslated region (3'UTR) has been studied previously. Several cis-acting elements in 3'UTR are involved in post-transcriptional regulation of CSF-1 mRNA. These include conserved protein-binding motifs as well as miRNA targets. miRNAs are 21-23nt single strand RNA which bind the complementary sequences in mRNAs, suppressing translation and enhancing mRNA degradation. RESULTS: In this report, we investigate the effect of miRNAs on post-transcriptional regulation of CSF-1 mRNA in human ovarian cancer. Bioinformatics analysis predicts at least 14 miRNAs targeting CSF-1 mRNA 3'UTR. By mutations in putative miRNA targets in CSF-1 mRNA 3'UTR, we identified a common target for both miR-128 and miR-152. We have also found that both miR-128 and miR-152 down-regulate CSF-1 mRNA and protein expression in ovarian cancer cells leading to decreased cell motility and adhesion in vitro, two major aspects of the metastatic potential of cancer cells. CONCLUSION: The major CSF-1 mRNA 3'UTR contains a common miRNA target which is involved in post-transcriptional regulation of CSF-1. Our results provide the evidence for a mechanism by which miR-128 and miR-152 down-regulate CSF-1, an important regulator of ovarian cancer.


Subject(s)
Gene Expression Regulation, Neoplastic , Macrophage Colony-Stimulating Factor/biosynthesis , MicroRNAs/genetics , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , 3' Untranslated Regions , Cell Adhesion/drug effects , Cell Movement/genetics , Computational Biology , Female , Humans , Luciferases/genetics , Luciferases/metabolism , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/metabolism , MicroRNAs/metabolism , Ovarian Neoplasms/pathology , Proto-Oncogene Mas , RNA, Messenger/genetics , RNA, Messenger/metabolism
17.
Biol Reprod ; 86(5): 155, 1-10, 2012 May.
Article in English | MEDLINE | ID: mdl-22262689

ABSTRACT

In the seminiferous epithelium, spermatogonial stem cells (SSCs) are located in a particular environment called the "niche" that is controlled by the basement membrane, key testis somatic cells, and factors originating from the vascular network. However, the role of Leydig cells (LCs) as a niche component is not yet clearly elucidated. Recent studies showed that peccaries (Tayassu tajacu) present a peculiar LC cytoarchitecture in which these cells are located around the seminiferous tubule lobes, making the peccary a unique model for investigating the SSC niche. This peculiarity allowed us to subdivide the seminiferous tubule cross-sections in three different testis parenchyma regions (tubule-tubule, tubule-interstitium, and tubule-LC contact). Our aims were to characterize the different spermatogonial cell types and to determine the location and/or distribution of the SSCs along the seminiferous tubules. Compared to differentiating spermatogonia, undifferentiated spermatogonia (A(und)) presented a noticeably higher nuclear volume (P < 0.05), allowing an accurate evaluation of their distribution. Immunostaining analysis demonstrated that approximately 93% of A(und) were GDNF receptor alpha 1 positive (GFRA1(+)), and these cells were preferentially located adjacent to the interstitial compartment without LCs (P < 0.05). The expression of colony-stimulating factor 1 was observed in LCs and peritubular myoid cells (PMCs), whereas its receptor was present in LCs and in GFRA1(+) A(und). Taken together, our findings strongly suggest that LCs, different from PMCs, might play a minor role in the SSC niche and physiology and that these steroidogenic cells are probably involved in the differentiation of A(und) toward type A(1) spermatogonia.


Subject(s)
Spermatogonia/metabolism , Stem Cell Niche/physiology , Animals , Artiodactyla/physiology , Glial Cell Line-Derived Neurotrophic Factor Receptors/analysis , Leydig Cells/cytology , Leydig Cells/metabolism , Macrophage Colony-Stimulating Factor/biosynthesis , Male , Receptor, Macrophage Colony-Stimulating Factor/analysis , Seminiferous Tubules/cytology , Spermatogenesis/physiology , Spermatogonia/cytology , Stem Cells/cytology , Stem Cells/metabolism
18.
Gynecol Oncol ; 124(2): 325-34, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22005523

ABSTRACT

OBJECTIVE: The purpose of this study was to investigate the role of miR-130b in the development of multidrug-resistant ovarian cancer. METHODS: The expression of miR-130b was assessed in ovarian tissues and cell lines by qRT-PCR. In vitro, miR-130b level was manipulated by transfection with mimics or inhibitors. Methylation level of miR-130b was evaluated by quantitative methylation-specific PCR (qMSP). CSF-1 expression in ovarian tissues and cells was determined by qRT-PCR, immunohistochemistry and ELISA, respectively. CSF-1 regulated by miR-130b was detected using Dual Luciferase Reporter system. RESULTS: Down-regulation of miR-130b in ovarian cancer was associated with FIGO III-IV clinical stages and poorer histological differentiation. MiR-130b was downregulated in multidrug resistant ovarian cancer cells. Restoration of miR-130b expression could sensitize these cells to anticancer drugs. MiR-130b hypermethylation was found in ovarian cancer tissues as well as in drug resistant cell lines and the methylation level was negatively correlated with its expression. Demethylation with 5-aza-CdR led to reactivation of miR-130b expression in drug resistant ovarian cancer cell lines concomitant with increase of sensibility to cisplatin and taxol. CSF-1 expression was negatively associated with miR-130b level in ovarian tissues and cell lines. Luciferase assay validated CSF-1 is a direct target of miR-130b. Knock-down of CSF-1 sensitized ovarian cancer cells to anticancer drugs and could partially attenuate the resistance inducing effect of miR-130b inhibitors. CONCLUSIONS: Downregulation of miR-130b promotes the development of multidrug resistant ovarian cancer partially by targeting the 3'-UTR of CSF-1, and the silencing of miR-130b may be mediated by DNA methylation.


Subject(s)
Macrophage Colony-Stimulating Factor/genetics , MicroRNAs/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Cell Line, Tumor , CpG Islands , Cytosine/metabolism , DNA Methylation , Down-Regulation , Drug Resistance, Multiple , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor , Epigenomics , Female , Gene Silencing , Humans , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophage Colony-Stimulating Factor/metabolism , MicroRNAs/administration & dosage , MicroRNAs/biosynthesis , Ovarian Neoplasms/metabolism , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Transfection
19.
Int J Immunopathol Pharmacol ; 25(4): 1131-6, 2012.
Article in English | MEDLINE | ID: mdl-23298503

ABSTRACT

Pigmented villonodular synovitis (PVNS) is a rare condition, most commonly involving the knee joint. PVNS is locally aggressive and can invade and destroy surrounding soft tissue and bone, leading to anatomical and functional deterioration of the affected joint. Localized PVNS is an unusual presentation of the disease, generally consisting of a nodular lesion protruding into the articular cavity. Localized PVNS of the knee can mimic other joint disorders which may pose a challenge for a correct diagnosis. Given the locally aggressive behavior of PVNS, prompt identification and excision of the lesion are instrumental to avoid complications. Here, we report a rare case of localized cystic PVNS involving the anterior cruciate ligament of the knee in a 32-year-old woman with persistent knee pain, in whom magnetic resonance imaging was inconclusive. The diagnosis was achieved via arthroscopy and histology. We also present a concise review of the literature on this pathological entity as well as a discussion on the differential diagnosis between localized PVNS and other intra-articular cystic lesions.


Subject(s)
Anterior Cruciate Ligament/pathology , Synovitis, Pigmented Villonodular/pathology , Adult , Diagnosis, Differential , Female , Humans , Inflammation/diagnosis , Inflammation/pathology , Macrophage Colony-Stimulating Factor/biosynthesis , Magnetic Resonance Imaging , Neoplasms/diagnosis , Neoplasms/pathology , Rare Diseases/diagnosis , Rare Diseases/pathology , Synovitis, Pigmented Villonodular/diagnosis
20.
Curr Opin Oncol ; 23(4): 361-6, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21577109

ABSTRACT

PURPOSE OF REVIEW: To review recent developments in the molecular pathogenesis of tenosynovial giant cell tumor (TGCT) or pigmented villonodular synovitis (PVNS) and its therapeutic implications. RECENT FINDINGS: TGCT or PVNS is a benign clonal neoplastic proliferation arising from the synovium characterized by a minor population of intratumoral cells that harbor a recurrent translocation. These cells overexpress CSF1, resulting in recruitment of CSF1R-bearing macrophages that are polyclonal and make up the bulk of the tumor. Inhibition of CSF1R using small molecule inhibitors such as imatinib, nilotinib or sunitinib can result in clinical, radiological and functional improvement in the affected joint. SUMMARY: Currently, surgery remains the treatment of choice for patients with TGCT/PVNS. Localized TGCT/PVNS is managed by marginal excision. Recurrences occur in 8-20% of patients and are easily managed by re-excision. Diffuse TGCT/PVNS tends to recur more often (33-50%) and has a much more aggressive clinical course. Patients are often symptomatic and require multiple surgical procedures during their lifetime. For patients with unresectable disease or multiple recurrences, systemic therapy using CSF1R inhibitors may help delay or avoid surgical procedures and improve functional outcomes.


Subject(s)
Antineoplastic Agents/therapeutic use , Giant Cell Tumors/therapy , Soft Tissue Neoplasms/therapy , Synovitis, Pigmented Villonodular/therapy , Antineoplastic Agents/pharmacology , Benzamides , Giant Cell Tumors/drug therapy , Giant Cell Tumors/pathology , Giant Cell Tumors/surgery , Humans , Imatinib Mesylate , Indoles/pharmacology , Indoles/therapeutic use , Macrophage Colony-Stimulating Factor/biosynthesis , Macrophages/metabolism , Piperazines/pharmacology , Piperazines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Pyrroles/pharmacology , Pyrroles/therapeutic use , Receptor, Macrophage Colony-Stimulating Factor/antagonists & inhibitors , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Soft Tissue Neoplasms/drug therapy , Soft Tissue Neoplasms/pathology , Soft Tissue Neoplasms/surgery , Sunitinib , Synovial Membrane/pathology , Synovitis, Pigmented Villonodular/drug therapy , Synovitis, Pigmented Villonodular/pathology , Synovitis, Pigmented Villonodular/surgery , Tendons/pathology
SELECTION OF CITATIONS
SEARCH DETAIL