Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
Add more filters










Publication year range
2.
Nature ; 603(7900): 328-334, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35197632

ABSTRACT

Effective antitumour immunity depends on the orchestration of potent T cell responses against malignancies1. Regression of human cancers has been induced by immune checkpoint inhibitors, T cell engagers or chimeric antigen receptor T cell therapies2-4. Although CD8 T cells function as key effectors of these responses, the role of CD4 T cells beyond their helper function has not been defined. Here we demonstrate that a trispecific antibody to HER2, CD3 and CD28 stimulates regression of breast cancers in a humanized mouse model through a mechanism involving CD4-dependent inhibition of tumour cell cycle progression. Although CD8 T cells directly mediated tumour lysis in vitro, CD4 T cells exerted antiproliferative effects by blocking cancer cell cycle progression at G1/S. Furthermore, when T cell subsets were adoptively transferred into a humanized breast cancer tumour mouse model, CD4 T cells alone inhibited HER2+ breast cancer growth in vivo. RNA microarray analysis revealed that CD4 T cells markedly decreased tumour cell cycle progression and proliferation, and also increased pro-inflammatory signalling pathways. Collectively, the trispecific antibody to HER2 induced T cell-dependent tumour regression through direct antitumour and indirect pro-inflammatory/immune effects driven by CD4 T cells.


Subject(s)
Breast Neoplasms , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , CD28 Antigens/metabolism , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Female , Humans , Mice , Receptor, ErbB-2/genetics
3.
Nat Cancer ; 1(1): 86-98, 2020 01.
Article in English | MEDLINE | ID: mdl-35121834

ABSTRACT

Despite the significant therapeutic advances provided by immune-checkpoint blockade and chimeric antigen receptor T cell treatments, many malignancies remain unresponsive to immunotherapy. Bispecific antibodies targeting tumor antigens and activating T cell receptor signaling have shown some clinical efficacy; however, providing co-stimulatory signals may improve T cell responses against tumors. Here, we developed a trispecific antibody that interacts with CD38, CD3 and CD28 to enhance both T cell activation and tumor targeting. The engagement of both CD3 and CD28 affords efficient T cell stimulation, whereas the anti-CD38 domain directs T cells to myeloma cells, as well as to certain lymphomas and leukemias. In vivo administration of this antibody suppressed myeloma growth in a humanized mouse model and also stimulated memory/effector T cell proliferation and reduced regulatory T cells in non-human primates at well-tolerated doses. Collectively, trispecific antibodies represent a promising platform for cancer immunotherapy.


Subject(s)
Antibodies, Bispecific , Multiple Myeloma , Animals , Antibodies, Bispecific/therapeutic use , CD28 Antigens , Mice , Multiple Myeloma/drug therapy , Receptors, Antigen, T-Cell , T-Lymphocytes
4.
Oncogenesis ; 8(4): 24, 2019 Apr 01.
Article in English | MEDLINE | ID: mdl-30936429

ABSTRACT

Senescence is an important p53-controlled tumor suppressor program that not only opposes the proliferation of cancer cells but also promotes their immune-mediated clearance in certain contexts. In hepatocellular cancer, p53 induction promotes an innate immune cell-mediated clearance of senescent cells wherein natural killer (NK) cells seem to play the primary sentinel role. Whether NK cells also surveil cancer cells in other tumor types when p53 is activated to promote a senescence response is unknown. To identify the role that NK and other innate immune cell types have on the surveillance and destruction of lung adenocarcinoma cells, we developed an orthotopic transplantation model where p53 gene function could be restored to induce senescence after successful engraftment of tumor cells in the mouse lung. Contrary to precedent, we found that NK cells actually limited the efficient clearance of tumor cells from the mouse lung after p53 restoration. Instead, activation of p53 induced the infiltration of monocytes, neutrophils, and interstitial macrophages. Loss of NK cells further promoted expansion of these inflammatory cell types and tumor clearance after p53 restoration. These observations suggest that NK cell responses to p53 activation in lung adenocarcinoma is distinct from those found in other tumor types and that diverse innate immune cell populations may play context-dependent roles during tumor immune surveillance. Further, our data provide an impetus to understand the broader mechanisms that regulate cancer cell destruction by multiple cell types of the innate immune system and distinct cancer contexts.

5.
Science ; 358(6367)2017 12 01.
Article in English | MEDLINE | ID: mdl-29191879

ABSTRACT

Bone marrow-derived myeloid cells can accumulate within tumors and foster cancer outgrowth. Local immune-neoplastic interactions have been intensively investigated, but the contribution of the systemic host environment to tumor growth remains poorly understood. Here, we show in mice and cancer patients (n = 70) that lung adenocarcinomas increase bone stromal activity in the absence of bone metastasis. Animal studies reveal that the cancer-induced bone phenotype involves bone-resident osteocalcin-expressing (Ocn+) osteoblastic cells. These cells promote cancer by remotely supplying a distinct subset of tumor-infiltrating SiglecFhigh neutrophils, which exhibit cancer-promoting properties. Experimentally reducing Ocn+ cell numbers suppresses the neutrophil response and lung tumor outgrowth. These observations posit osteoblasts as remote regulators of lung cancer and identify SiglecFhigh neutrophils as myeloid cell effectors of the osteoblast-driven protumoral response.


Subject(s)
Adenocarcinoma/pathology , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Bone and Bones/pathology , Lectins/metabolism , Lung Neoplasms/pathology , Neutrophil Infiltration , Neutrophils/metabolism , Neutrophils/pathology , Osteoblasts/pathology , Adenocarcinoma of Lung , Animals , Bone Density , Bone Marrow Cells/pathology , Bone and Bones/metabolism , Cell Line, Tumor , Humans , Mice , Mice, Inbred C57BL , Myeloid Cells/pathology , Neoplasms, Experimental/pathology , Osteocalcin/metabolism , Receptor for Advanced Glycation End Products/metabolism
6.
Cancer Discov ; 6(6): 630-49, 2016 06.
Article in English | MEDLINE | ID: mdl-27072748

ABSTRACT

UNLABELLED: Immune cells promote the initial metastatic dissemination of carcinoma cells from primary tumors. In contrast to their well-studied functions in the initial stages of metastasis, the specific roles of immunocytes in facilitating progression through the critical later steps of the invasion-metastasis cascade remain poorly understood. Here, we define novel functions of neutrophils in promoting intraluminal survival and extravasation at sites of metastatic dissemination. We show that CD11b(+)/Ly6G(+) neutrophils enhance metastasis formation via two distinct mechanisms. First, neutrophils inhibit natural killer cell function, which leads to a significant increase in the intraluminal survival time of tumor cells. Thereafter, neutrophils operate to facilitate extravasation of tumor cells through the secretion of IL1ß and matrix metalloproteinases. These results identify neutrophils as key regulators of intraluminal survival and extravasation through their cross-talk with host cells and disseminating carcinoma cells. SIGNIFICANCE: This study provides important insights into the systemic contributions of neutrophils to cancer metastasis by identifying how neutrophils facilitate intermediate steps of the invasion-metastasis cascade. We demonstrate that neutrophils suppress natural killer cell activity and increase extravasation of tumor cells. Cancer Discov; 6(6); 630-49. ©2016 AACR.This article is highlighted in the In This Issue feature, p. 561.


Subject(s)
Carcinoma/immunology , Carcinoma/pathology , Killer Cells, Natural/immunology , Neutrophils/immunology , Adoptive Transfer , Animals , Biomarkers , Carcinoma/genetics , Carcinoma/metabolism , Cell Communication , Cell Line, Tumor , Cell Movement , Cell Survival , Cytokines/biosynthesis , Disease Models, Animal , Endothelial Cells/metabolism , Heterografts , Humans , Immunity, Innate , Immunophenotyping , Killer Cells, Natural/metabolism , Matrix Metalloproteinases/metabolism , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Neutrophils/metabolism , Phenotype
7.
Immunity ; 44(2): 343-54, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26872698

ABSTRACT

Checkpoint blockade immunotherapies can be extraordinarily effective, but might benefit only the minority of patients whose tumors are pre-infiltrated by T cells. Here, using lung adenocarcinoma mouse models, including genetic models, we show that autochthonous tumors that lacked T cell infiltration and resisted current treatment options could be successfully sensitized to host antitumor T cell immunity when appropriately selected immunogenic drugs (e.g., oxaliplatin combined with cyclophosphamide for treatment against tumors expressing oncogenic Kras and lacking Trp53) were used. The antitumor response was triggered by direct drug actions on tumor cells, relied on innate immune sensing through toll-like receptor 4 signaling, and ultimately depended on CD8(+) T cell antitumor immunity. Furthermore, instigating tumor infiltration by T cells sensitized tumors to checkpoint inhibition and controlled cancer durably. These findings indicate that the proportion of cancers responding to checkpoint therapy can be feasibly and substantially expanded by combining checkpoint blockade with immunogenic drugs.


Subject(s)
Adenocarcinoma/therapy , CD8-Positive T-Lymphocytes/drug effects , Immunotherapy/methods , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/drug effects , Adenocarcinoma/immunology , Animals , Cell Line, Tumor , Central Nervous System Sensitization/drug effects , Cyclophosphamide/administration & dosage , Disease Models, Animal , Drug Therapy/methods , Genes, cdc/drug effects , Humans , Immunity, Innate , Lung Neoplasms/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Toll-Like Receptor 4/metabolism
8.
PLoS One ; 10(3): e0121419, 2015.
Article in English | MEDLINE | ID: mdl-25799465

ABSTRACT

Uncontrolled inflammation is one of the leading causes of kidney failure. Pro-inflammatory responses can occur in the absence of infection, a process called sterile inflammation. Here we show that the purinergic receptor P2Y14 (GPR105) is specifically and highly expressed in collecting duct intercalated cells (ICs) and mediates sterile inflammation in the kidney. P2Y14 is activated by UDP-glucose, a damage-associated molecular pattern molecule (DAMP) released by injured cells. We found that UDP-glucose increases pro-inflammatory chemokine expression in ICs as well as MDCK-C11 cells, and UDP-glucose activates the MEK1/2-ERK1/2 pathway in MDCK-C11 cells. These effects were prevented following inhibition of P2Y14 with the small molecule PPTN. Tail vein injection of mice with UDP-glucose induced the recruitment of neutrophils to the renal medulla. This study identifies ICs as novel sensors, mediators and effectors of inflammation in the kidney via P2Y14.


Subject(s)
Inflammation/metabolism , Kidney Tubules, Collecting/pathology , Receptors, Purinergic P2Y/metabolism , Uridine Diphosphate Glucose/pharmacology , Animals , Cells, Cultured , Dogs , Inflammation/immunology , Inflammation/pathology , Kidney Tubules, Collecting/cytology , Kidney Tubules, Collecting/metabolism , MAP Kinase Signaling System/drug effects , Madin Darby Canine Kidney Cells , Male , Mice , Neutrophils/metabolism
9.
Biol Reprod ; 90(5): 90, 2014 May.
Article in English | MEDLINE | ID: mdl-24648397

ABSTRACT

The epithelium that lines the epididymal duct establishes the optimal milieu in which spermatozoa mature, acquire motility, and are stored. This finely tuned environment also protects antigenic sperm against pathogens and autoimmunity, which are potential causes of transient or permanent infertility. The epididymal epithelium is pseudostratified and contains basal cells (BCs) that are located beneath other epithelial cells. Previous studies showed that in the mouse epididymis, BCs possess macrophage-like characteristics. However, we previously identified a dense population of cells belonging to the mononuclear phagocyte (MP) system (comprised of macrophages and dendritic cells) in the basal compartment of the mouse epididymis and showed that a subset of MPs express the macrophage marker F4/80. In the present study, we evaluate the distribution of BCs and MPs in the epididymis of transgenic CD11c-EYFP mice, in which EYFP is expressed exclusively in MPs, using antibodies against the BC marker keratin 5 (KRT5) and the macrophage marker F4/80. Immunofluorescence labeling for laminin, a basement membrane marker, showed that BCs and most MPs are located in the basal region of the epithelium. Confocal microscopy showed that in the initial segment, both BCs and MPs project intraepithelial extensions and establish a very intricate network. Flow cytometry experiments demonstrated that epididymal MPs and BCs are phenotypically distinct. BCs do not express F4/80, and MPs do not express KRT5. Therefore, despite their proximity and some morphological similarities with peritubular macrophages and dendritic cells, BCs do not belong to the MP system.


Subject(s)
Dendritic Cells/immunology , Epididymis/immunology , Epithelium/immunology , Macrophages/immunology , Animals , Antigens, Differentiation/immunology , CD11 Antigens/immunology , Epididymis/cytology , Epithelial Cells/immunology , Flow Cytometry , Male , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence
10.
Oncoimmunology ; 2(5): e24183, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23762799

ABSTRACT

The communication between tumor and host cells involves signals that act across extended distances in the body. Recent evidence indicates that the hormone angiotensin II is overproduced by lung adenocarcinoma to remotely expand bone marrow-derived hematopoietic stem cells. This process amplifies the supply of tumor-associated macrophages, which promote disease progression.

11.
Immunity ; 38(2): 296-308, 2013 Feb 21.
Article in English | MEDLINE | ID: mdl-23333075

ABSTRACT

Macrophages frequently infiltrate tumors and can enhance cancer growth, yet the origins of the macrophage response are not well understood. Here we address molecular mechanisms of macrophage production in a conditional mouse model of lung adenocarcinoma. We report that overproduction of the peptide hormone Angiotensin II (AngII) in tumor-bearing mice amplifies self-renewing hematopoietic stem cells (HSCs) and macrophage progenitors. The process occurred in the spleen but not the bone marrow, and was independent of hemodynamic changes. The effects of AngII required direct hormone ligation on HSCs, depended on S1P(1) signaling, and allowed the extramedullary tissue to supply new tumor-associated macrophages throughout cancer progression. Conversely, blocking AngII production prevented cancer-induced HSC and macrophage progenitor amplification and thus restrained the macrophage response at its source. These findings indicate that AngII acts upstream of a potent macrophage amplification program and that tumors can remotely exploit the hormone's pathway to stimulate cancer-promoting immunity.


Subject(s)
Adenocarcinoma/metabolism , Angiotensin II/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Macrophages/metabolism , Spleen/metabolism , Adenocarcinoma/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Angiotensin II/metabolism , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Communication , Cell Movement , Cell Proliferation , Gene Expression , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lysophospholipids/metabolism , Macrophages/pathology , Mice , Mice, Transgenic , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/metabolism , Spleen/pathology , Tumor Burden
12.
PLoS Biol ; 10(9): e1001393, 2012.
Article in English | MEDLINE | ID: mdl-23055830

ABSTRACT

Intestinal epithelial cells exist within a complex environment that affects how they interpret and respond to stimuli. We have applied a multi-scale in vivo systems approach to understand how intestinal immune cells communicate with epithelial cells to regulate responses to inflammatory signals. Multivariate modeling analysis of a large dataset composed of phospho-signals, cytokines, and immune cell populations within the intestine revealed an intimate relationship between immune cells and the epithelial response to TNF-α. Ablation of lymphocytes in the intestine prompted a decrease in the expression of MCP-1, which in turn increased the steady state number of intestinal plasmacytoid dendritic cells (pDCs). This change in the immune compartment affected the intestinal cytokine milieu and subsequent epithelial cell signaling network, with cells becoming hypersensitive to TNF-α-induced apoptosis in a way that could be predicted by mathematical modeling. In summary, we have uncovered a novel cellular network that regulates the response of intestinal epithelial cells to inflammatory stimuli in an in vivo setting.


Subject(s)
Apoptosis/drug effects , Intestinal Mucosa/cytology , Intestinal Mucosa/drug effects , Lymphocytes/cytology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cell Movement/drug effects , Chemokine CCL2/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Interferon-gamma/metabolism , Intestinal Mucosa/metabolism , Mice , Mice, Inbred C57BL , Models, Biological , Neutralization Tests , Phenotype , Signal Transduction/drug effects
13.
Cell Rep ; 1(4): 317-24, 2012 Apr 19.
Article in English | MEDLINE | ID: mdl-22545247

ABSTRACT

Monocytes serve as a central defense system against infection and injury but can also promote pathological inflammatory responses. Considering the evidence that monocytes exist in at least two subsets committed to divergent functions, we investigated whether distinct factors regulate the balance between monocyte subset responses in vivo. We identified a microRNA (miRNA), miR-146a, which is differentially regulated both in mouse (Ly-6C(hi)/Ly-6C(lo)) and human (CD14(hi)/CD14(lo)CD16(+)) monocyte subsets. The single miRNA controlled the amplitude of the Ly-6C(hi) monocyte response during inflammatory challenge whereas it did not affect Ly-6C(lo) cells. miR-146a-mediated regulation was cell-intrinsic and depended on Relb, a member of the noncanonical NF-κB/Rel family, which we identified as a direct miR-146a target. These observations not only provide mechanistic insights into the molecular events that regulate responses mediated by committed monocyte precursor populations but also identify targets for manipulating Ly-6C(hi) monocyte responses while sparing Ly-6Clo monocyte activity.


Subject(s)
MicroRNAs/physiology , Monocytes/physiology , Transcription Factor RelB/physiology , Animals , Cell Movement , Cell Proliferation , Gene Expression Regulation , Humans , Mice , Monocytes/immunology , Monocytes/metabolism , Transcription Factor RelB/metabolism
14.
J Innate Immun ; 4(5-6): 411-23, 2012.
Article in English | MEDLINE | ID: mdl-22433183

ABSTRACT

Tissue macrophages (Mø) and dendritic cells (DC) are thought to derive from hematopoietic stem cells, which exist in the bone marrow and generate intermediate precursor populations with increasingly restricted lineage potentials. There exists several precursors committed to the Mø and DC lineages; these cells exhibit distinct tropism and function and respond differentially in pathophysiologic conditions. In this review, we consider experimental contexts in which Mø and DC responses in tissue are not only dictated by the local environment, but also by the quantity and quality of newly recruited lineage precursor cells. Consequently, we discuss whether therapeutic control of Mø and DC responses in tissue may be achieved through manipulation of their lineage precursors.


Subject(s)
Cell Lineage/immunology , Dendritic Cells/immunology , Macrophages/immunology , Myeloid Progenitor Cells/immunology , Animals , Cell Differentiation , Dendritic Cells/cytology , Humans , Macrophages/cytology , Mice , Myeloid Progenitor Cells/cytology
15.
Proc Natl Acad Sci U S A ; 109(7): 2491-6, 2012 Feb 14.
Article in English | MEDLINE | ID: mdl-22308361

ABSTRACT

Tumor-associated macrophages (TAMs) and tumor-associated neutrophils (TANs) can control cancer growth and exist in almost all solid neoplasms. The cells are known to descend from immature monocytic and granulocytic cells, respectively, which are produced in the bone marrow. However, the spleen is also a recently identified reservoir of monocytes, which can play a significant role in the inflammatory response that follows acute injury. Here, we evaluated the role of the splenic reservoir in a genetic mouse model of lung adenocarcinoma driven by activation of oncogenic Kras and inactivation of p53. We found that high numbers of TAM and TAN precursors physically relocated from the spleen to the tumor stroma, and that recruitment of tumor-promoting spleen-derived TAMs required signaling of the chemokine receptor CCR2. Also, removal of the spleen, either before or after tumor initiation, reduced TAM and TAN responses significantly and delayed tumor growth. The mechanism by which the spleen was able to maintain its reservoir capacity throughout tumor progression involved, in part, local accumulation in the splenic red pulp of typically rare extramedullary hematopoietic stem and progenitor cells, notably granulocyte and macrophage progenitors, which produced CD11b(+) Ly-6C(hi) monocytic and CD11b(+) Ly-6G(hi) granulocytic cells locally. Splenic granulocyte and macrophage progenitors and their descendants were likewise identified in clinical specimens. The present study sheds light on the origins of TAMs and TANs, and positions the spleen as an important extramedullary site, which can continuously supply growing tumors with these cells.


Subject(s)
Macrophages/immunology , Neoplasms/pathology , Neutrophils/immunology , Animals , Humans , Mice , Neoplasms/immunology , Spleen/immunology , Spleen/pathology
16.
Nat Biotechnol ; 29(11): 1005-10, 2011 Oct 09.
Article in English | MEDLINE | ID: mdl-21983520

ABSTRACT

Excessive and prolonged activity of inflammatory monocytes is a hallmark of many diseases with an inflammatory component. In such conditions, precise targeting of these cells could be therapeutically beneficial while sparing many essential functions of the innate immune system, thus limiting unwanted effects. Inflammatory monocytes-but not the noninflammatory subset-depend on the chemokine receptor CCR2 for localization to injured tissue. Here we present an optimized lipid nanoparticle and a CCR2-silencing short interfering RNA that, when administered systemically in mice, show rapid blood clearance, accumulate in spleen and bone marrow, and localize to monocytes. Efficient degradation of CCR2 mRNA in monocytes prevents their accumulation in sites of inflammation. Specifically, the treatment attenuates their number in atherosclerotic plaques, reduces infarct size after coronary artery occlusion, prolongs normoglycemia in diabetic mice after pancreatic islet transplantation, and results in reduced tumor volumes and lower numbers of tumor-associated macrophages.


Subject(s)
Gene Silencing , Inflammation/therapy , Macrophages/drug effects , Nanoparticles , RNA, Small Interfering/therapeutic use , Receptors, CCR2/antagonists & inhibitors , Animals , Atherosclerosis/therapy , Blood Glucose , Diabetes Mellitus/surgery , Diabetes Mellitus/therapy , Disease Models, Animal , Graft Survival/genetics , Humans , Islets of Langerhans Transplantation , Mice , Mice, Inbred C57BL , Monocytes/metabolism , Myocardial Infarction/prevention & control , Myocardial Infarction/therapy , Nanoparticles/chemistry , Receptors, CCR2/genetics
17.
Reproduction ; 141(5): 653-63, 2011 May.
Article in English | MEDLINE | ID: mdl-21310816

ABSTRACT

One of the most intriguing aspects of male reproductive physiology is the ability to generate spermatogenic cells - which are 'foreign' to the host - without triggering immune activation. After leaving the testis, spermatozoa enter the epididymis where they mature and are stored. In this study, we report a previously unrecognized dense network of dendritic cells (DCs) located at the base of the epididymal epithelium. This network was detected in transgenic mice expressing CD11c-EYFP and CX3CR1-GFP reporters. Epididymal DCs (eDCs) establish intimate interactions with the epithelium and project long dendrites between epithelial cells toward the lumen. We show that isolated eDCs express numerous leukocyte markers described previously in other organs that are in contact with the external environment, and present and cross-present ovalbumin to T cells in vitro. eDCs are, therefore, strategically positioned to regulate the complex interplay between immune tolerance and activation, a balance that is fundamental to male fertility.


Subject(s)
Dendritic Cells/immunology , Epididymis/immunology , Immune Tolerance , Animals , Antigen Presentation , Bacterial Proteins/biosynthesis , Bacterial Proteins/genetics , Biomarkers/metabolism , CD11c Antigen/biosynthesis , CD11c Antigen/genetics , Cells, Cultured , Coculture Techniques , Dendritic Cells/metabolism , Epididymis/cytology , Epididymis/metabolism , Fertility , Genes, Reporter , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Immunophenotyping , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Ovalbumin/immunology , Phenotype , Receptors, Chemokine/biosynthesis , Receptors, Chemokine/genetics , T-Lymphocytes/immunology
18.
J Immunol ; 186(3): 1369-76, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21187450

ABSTRACT

Repulsive guidance molecule (RGM) family members RGMa, RGMb/Dragon, and RGMc/hemojuvelin were found recently to act as bone morphogenetic protein (BMP) coreceptors that enhance BMP signaling activity. Although our previous studies have shown that hemojuvelin regulates hepcidin expression and iron metabolism through the BMP pathway, the role of the BMP signaling mediated by Dragon remains largely unknown. We have shown previously that Dragon is expressed in neural cells, germ cells, and renal epithelial cells. In this study, we demonstrate that Dragon is highly expressed in macrophages. Studies with RAW264.7 and J774 macrophage cell lines reveal that Dragon negatively regulates IL-6 expression in a BMP ligand-dependent manner via the p38 MAPK and Erk1/2 pathways but not the Smad1/5/8 pathway. We also generated Dragon knockout mice and found that IL-6 is upregulated in macrophages and dendritic cells derived from whole lung tissue of these mice compared with that in respective cells derived from wild-type littermates. These results indicate that Dragon is an important negative regulator of IL-6 expression in immune cells and that Dragon-deficient mice may be a useful model for studying immune and inflammatory disorders.


Subject(s)
Gene Expression Regulation/immunology , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Macrophages/immunology , Macrophages/metabolism , Nerve Tissue Proteins/physiology , Animals , Cell Adhesion Molecules, Neuronal , Cell Line , Disease Models, Animal , Down-Regulation/immunology , GPI-Linked Proteins , Inflammation Mediators/metabolism , Inflammation Mediators/physiology , Interleukin-6/biosynthesis , MAP Kinase Signaling System/immunology , Macrophages/pathology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/physiology , Mitogen-Activated Protein Kinase 3/physiology , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , p38 Mitogen-Activated Protein Kinases/physiology
19.
Circ Res ; 107(11): 1364-73, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20930148

ABSTRACT

RATIONALE: Monocytes recruited to ischemic myocardium originate from a reservoir in the spleen, and the release from their splenic niche relies on angiotensin (Ang) II signaling. OBJECTIVE: Because monocytes are centrally involved in tissue repair after ischemia, we hypothesized that early angiotensin-converting enzyme (ACE) inhibitor therapy impacts healing after myocardial infarction partly via effects on monocyte traffic. METHODS AND RESULTS: In a mouse model of permanent coronary ligation, enalapril arrested the release of monocytes from the splenic reservoir and consequently reduced their recruitment into the healing infarct by 45%, as quantified by flow cytometry of digested infarcts. Time-lapse intravital microscopy revealed that enalapril reduces monocyte motility in the spleen. In vitro migration assays and Western blotting showed that this was caused by reduced signaling through the Ang II type 1 receptor. We then studied the long-term consequences of blocked splenic monocyte release in atherosclerotic apolipoprotein (apo)E(-/-) mice, in which infarct healing is impaired because of excessive inflammation in the cardiac wound. Enalapril improved histologic healing biomarkers and reduced inflammation in infarcts measured by FMT-CT (fluorescence molecular tomography in conjunction with x-ray computed tomography) of proteolytic activity. ACE inhibition improved MRI-derived ejection fraction by 14% on day 21, despite initially comparable infarct size. In apoE(-/-) mice, ischemia/reperfusion injury resulted in larger infarct size and enhanced monocyte recruitment and was reversible by enalapril treatment. Splenectomy reproduced antiinflammatory effects of enalapril. CONCLUSION: This study suggests that benefits of early ACE inhibition after myocardial infarction can partially be attributed to its potent antiinflammatory impact on the splenic monocyte reservoir.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/pharmacology , Cell Movement/drug effects , Monocytes/enzymology , Monocytes/pathology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Spleen/enzymology , Spleen/pathology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Animals , Cell Movement/physiology , Enalapril/pharmacology , Enalapril/therapeutic use , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Monocytes/drug effects , Myocardial Infarction/drug therapy , Spleen/drug effects
20.
Opt Lett ; 35(7): 1088-90, 2010 Apr 01.
Article in English | MEDLINE | ID: mdl-20364226

ABSTRACT

Optical projection tomography is a new ex vivo imaging technique that allows imaging of whole organs in three dimensions at high spatial resolutions. In this Letter we demonstrate its capability to tomographically visualize molecular activity in whole organs of mice. In particular, eosinophil activity in asthmatic lungs is resolved using a Born-normalized fluorescence optical projection tomography and employing a near-IR molecular probe. The possibility to achieve molecularly sensitive imaging contrast in optical projection tomography by means of targeted and activatable imaging reporter agents adds a new range of capabilities for investigating molecular signatures of pathophysiological processes and a wide variety of diseases and their development.


Subject(s)
Microscopy, Fluorescence/instrumentation , Molecular Probe Techniques/instrumentation , Tomography, Optical/instrumentation , Computer-Aided Design , Equipment Design , Equipment Failure Analysis , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...