Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
2.
Atherosclerosis ; 239(1): 232-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25618031

ABSTRACT

OBJECTIVE: Homozygosity for a 1.7 kb intragenic duplication of the Haptoglobin (Hp) gene (Hp 2-2 genotype), present in 36% of the population, has been associated with a 2-3 fold increased incidence of atherothrombosis in individuals with Diabetes (DM) in 10 longitudinal studies compared to DM individuals not homozygous for this duplication (Hp 1-1/2-1). The increased CVD risk associated with the Hp 2-2 genotype has been shown to be prevented with vitamin E supplementation in man. We sought to determine if there was an interaction between the Hp genotype and vitamin E on atherosclerotic plaque growth and stability in a transgenic model of the Hp polymorphism. METHODS AND RESULTS: Brachiocephalic artery atherosclerotic plaque volume was serially assessed by high resolution ultrasound in 28 Hp 1-1 and 26 Hp 2-2 mice in a C57Bl/6 ApoE(-/-) background. Hp 2-2 mice had more rapid plaque growth and an increased incidence of plaque hemorrhage and rupture. Vitamin E significantly reduced plaque growth in Hp 2-2 but not in Hp 1-1 mice with a significant pharmacogenomic interaction between the Hp genotype and vitamin E on plaque growth. CONCLUSIONS: These results may help explain why vitamin E supplementation in man can prevent CVD in Hp 2-2 DM but not in non Hp 2-2 DM individuals.


Subject(s)
Genotype , Haptoglobins/genetics , Plaque, Atherosclerotic/genetics , Vitamin E/metabolism , Alleles , Animals , Antioxidants/metabolism , Apolipoproteins E/genetics , Brachiocephalic Trunk/pathology , Dietary Supplements , Disease Progression , Homozygote , Mice , Mice, Inbred C57BL , Mice, Transgenic , Oxygen/chemistry
3.
Eur Heart J ; 36(23): 1478-88, 2015 Jun 14.
Article in English | MEDLINE | ID: mdl-24950695

ABSTRACT

BACKGROUND: Myocarditis is characterized by inflammatory cell infiltration of the heart and subsequent deterioration of cardiac function. Monocytes are the most prominent population of accumulating leucocytes. We investigated whether in vivo administration of nanoparticle-encapsulated siRNA targeting chemokine (C-C motif) receptor 2 (CCR2)-a chemokine receptor crucial for leucocyte migration in humans and mice--reduces inflammation in autoimmune myocarditis. METHODS AND RESULTS: In myocardium of patients with myocarditis, CCL2 mRNA levels and CCR2(+) cells increased (P < 0.05), motivating us to pursue CCR2 silencing. Flow cytometric analysis showed that siRNA silencing of CCR2 (siCCR2) reduced the number of Ly6C(high) monocytes in hearts of mice with acute autoimmune myocarditis by 69% (P < 0.05), corroborated by histological assessment. The nanoparticle-delivered siRNA was not only active in monocytes but also in bone marrow haematopoietic progenitor cells. Treatment with siCCR2 reduced the migration of bone marrow granulocyte macrophage progenitors into the blood. Cellular magnetic resonance imaging (MRI) after injection of macrophage-avid magnetic nanoparticles detected myocarditis and therapeutic effects of RNAi non-invasively. Mice with acute myocarditis showed enhanced macrophage MRI contrast, which was prevented by siCCR2 (P < 0.05). Follow-up MRI volumetry revealed that siCCR2 treatment improved ejection fraction (P < 0.05 vs. control siRNA-treated mice). CONCLUSION: This study highlights the importance of CCR2 in the pathogenesis of myocarditis. In addition, we show that siCCR2 affects leucocyte progenitor trafficking. The data also point to a novel therapeutic strategy for the treatment of myocarditis.


Subject(s)
Autoimmune Diseases/therapy , Chemokine CCL2/genetics , Myocarditis/therapy , RNA, Small Interfering/pharmacology , Adult , Animals , Cell Movement , Chemokine CCL2/metabolism , Female , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/physiology , Humans , Magnetic Resonance Angiography , Male , Mice , Monocytes/metabolism , Nanoparticles , RNA Interference/physiology
4.
Nat Biotechnol ; 31(10): 898-907, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24013197

ABSTRACT

In a cell-free approach to regenerative therapeutics, transient application of paracrine factors in vivo could be used to alter the behavior and fate of progenitor cells to achieve sustained clinical benefits. Here we show that intramyocardial injection of synthetic modified RNA (modRNA) encoding human vascular endothelial growth factor-A (VEGF-A) results in the expansion and directed differentiation of endogenous heart progenitors in a mouse myocardial infarction model. VEGF-A modRNA markedly improved heart function and enhanced long-term survival of recipients. This improvement was in part due to mobilization of epicardial progenitor cells and redirection of their differentiation toward cardiovascular cell types. Direct in vivo comparison with DNA vectors and temporal control with VEGF inhibitors revealed the greatly increased efficacy of pulse-like delivery of VEGF-A. Our results suggest that modRNA is a versatile approach for expressing paracrine factors as cell fate switches to control progenitor cell fate and thereby enhance long-term organ repair.


Subject(s)
Cell Lineage , Myocardial Infarction/therapy , Myocardium/pathology , RNA, Messenger/metabolism , Regeneration , Stem Cells/cytology , Stem Cells/metabolism , Animals , Apoptosis , Biomarkers/metabolism , Cell Differentiation , Cell Proliferation , Disease Models, Animal , Endothelial Cells/pathology , Gene Transfer Techniques , Humans , Kinetics , Luciferases/metabolism , Mice , Models, Biological , Muscle, Skeletal/metabolism , Myocardial Infarction/physiopathology , Myocardium/metabolism , RNA, Messenger/genetics , Stem Cell Transplantation , Survival Analysis , Treatment Outcome , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism
5.
Nat Med ; 19(9): 1166-72, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23933982

ABSTRACT

During the inflammatory response that drives atherogenesis, macrophages accumulate progressively in the expanding arterial wall. The observation that circulating monocytes give rise to lesional macrophages has reinforced the concept that monocyte infiltration dictates macrophage buildup. Recent work has indicated, however, that macrophage accumulation does not depend on monocyte recruitment in some inflammatory contexts. We therefore revisited the mechanism underlying macrophage accumulation in atherosclerosis. In murine atherosclerotic lesions, we found that macrophages turn over rapidly, after 4 weeks. Replenishment of macrophages in these experimental atheromata depends predominantly on local macrophage proliferation rather than monocyte influx. The microenvironment orchestrates macrophage proliferation through the involvement of scavenger receptor A (SR-A). Our study reveals macrophage proliferation as a key event in atherosclerosis and identifies macrophage self-renewal as a therapeutic target for cardiovascular disease.


Subject(s)
Atherosclerosis/immunology , Atherosclerosis/pathology , Cell Proliferation , Macrophages/physiology , Animals , Cells, Cultured , Inflammation , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/physiology
6.
Circulation ; 127(20): 2038-46, 2013 May 21.
Article in English | MEDLINE | ID: mdl-23616627

ABSTRACT

BACKGROUND: Exaggerated and prolonged inflammation after myocardial infarction (MI) accelerates left ventricular remodeling. Inflammatory pathways may present a therapeutic target to prevent post-MI heart failure. However, the appropriate magnitude and timing of interventions are largely unknown, in part because noninvasive monitoring tools are lacking. Here, we used nanoparticle-facilitated silencing of CCR2, the chemokine receptor that governs inflammatory Ly-6C(high) monocyte subset traffic, to reduce infarct inflammation in apolipoprotein E-deficient (apoE(-/-)) mice after MI. We used dual-target positron emission tomography/magnetic resonance imaging of transglutaminase factor XIII (FXIII) and myeloperoxidase (MPO) activity to monitor how monocyte subset-targeted RNAi altered infarct inflammation and healing. METHODS AND RESULTS: Flow cytometry, gene expression analysis, and histology revealed reduced monocyte numbers and enhanced resolution of inflammation in infarcted hearts of apoE(-/-) mice that were treated with nanoparticle-encapsulated siRNA. To follow extracellular matrix cross-linking noninvasively, we developed a fluorine-18-labeled positron emission tomography agent ((18)F-FXIII). Recruitment of MPO-rich inflammatory leukocytes was imaged with a molecular magnetic resonance imaging sensor of MPO activity (MPO-Gd). Positron emission tomography/magnetic resonance imaging detected anti-inflammatory effects of intravenous nanoparticle-facilitated siRNA therapy (75% decrease of MPO-Gd signal; P<0.05), whereas (18)F-FXIII positron emission tomography reflected unimpeded matrix cross-linking in the infarct. Silencing of CCR2 during the first week after MI improved ejection fraction on day 21 after MI from 29% to 35% (P<0.05). CONCLUSION: CCR2-targeted RNAi reduced recruitment of Ly-6C(high) monocytes, attenuated infarct inflammation, and curbed post-MI left ventricular remodeling.


Subject(s)
Atherosclerosis/therapy , Gene Targeting/methods , Monocytes/metabolism , Myocardial Infarction/genetics , Myocardial Infarction/therapy , RNA Interference/physiology , Receptors, CCR2/genetics , Wound Healing/genetics , Amino Acid Sequence , Animals , Atherosclerosis/genetics , Atherosclerosis/pathology , Female , Genetic Predisposition to Disease , Genetic Therapy/methods , Mice , Mice, Inbred C57BL , Mice, Knockout , Molecular Sequence Data , Monocytes/pathology , Myocardial Infarction/pathology , Random Allocation , Receptors, CCR2/antagonists & inhibitors , Receptors, CCR2/metabolism
7.
Circ Res ; 112(6): 891-9, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23392842

ABSTRACT

RATIONALE: High-resolution imaging of the heart in vivo is challenging owing to the difficulty in accessing the heart and the tissue motion caused by the heartbeat. OBJECTIVE: Here, we describe a suction-assisted endoscope for visualizing fluorescently labeled cells and vessels in the beating heart tissue through a small incision made in the intercostal space. METHODS AND RESULTS: A suction tube with a diameter of 2 to 3 mm stabilizes the local tissue motion safely and effectively at a suction pressure of 50 mm Hg. Using a minimally invasive endoscope integrated into a confocal microscope, we performed fluorescence cellular imaging in both normal and diseased hearts in live mice for an hour per session repeatedly over a few weeks. Real-time imaging revealed the surprisingly rapid infiltration of CX3CR1(+) monocytes into the injured site within several minutes after acute myocardial infarction. CONCLUSIONS: The time-lapse analysis of flowing and rolling (patrolling) monocytes in the heart and the peripheral circulation provides evidence that the massively recruited monocytes come first from the vascular reservoir and later from the spleen. The imaging method requires minimal surgical preparation and can be implemented into standard intravital microscopes. Our results demonstrate the applicability of our imaging method for a wide range of cardiovascular research.


Subject(s)
Cell Movement/physiology , Endoscopy/methods , Monocytes/physiology , Myocardial Infarction/pathology , Time-Lapse Imaging/methods , Animals , Cell Count/methods , Endoscopes , Heart Rate/physiology , Immobilization/instrumentation , Immobilization/methods , Immunity, Cellular/physiology , Leukocyte Rolling/physiology , Mice , Microscopy, Confocal/instrumentation , Monocytes/cytology , Myocardial Contraction/physiology , Myocardial Infarction/immunology , Optical Imaging/methods , Suction/instrumentation , Suction/methods
8.
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
9.
Proc Natl Acad Sci U S A ; 109(52): 21444-9, 2012 Dec 26.
Article in English | MEDLINE | ID: mdl-23236189

ABSTRACT

The treatment of diseased vasculature remains challenging, in part because of the difficulty in implanting drug-eluting devices without subjecting vessels to damaging mechanical forces. Implanting materials using adhesive forces could overcome this challenge, but materials have previously not been shown to durably adhere to intact endothelium under blood flow. Marine mussels secrete strong underwater adhesives that have been mimicked in synthetic systems. Here we develop a drug-eluting bioadhesive gel that can be locally and durably glued onto the inside surface of blood vessels. In a mouse model of atherosclerosis, inflamed plaques treated with steroid-eluting adhesive gels had reduced macrophage content and developed protective fibrous caps covering the plaque core. Treatment also lowered plasma cytokine levels and biomarkers of inflammation in the plaque. The drug-eluting devices developed here provide a general strategy for implanting therapeutics in the vasculature using adhesive forces and could potentially be used to stabilize rupture-prone plaques.


Subject(s)
Adhesives/chemistry , Blood Vessels/pathology , Dexamethasone/therapeutic use , Plaque, Atherosclerotic/drug therapy , Plaque, Atherosclerotic/pathology , Adhesiveness/drug effects , Animals , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Arteries/drug effects , Arteries/pathology , Blood Vessels/drug effects , Catechols/chemistry , Dexamethasone/pharmacology , Drug Delivery Systems , Female , Gels/chemistry , Human Umbilical Vein Endothelial Cells/drug effects , Implants, Experimental , Inflammation/pathology , Mice , Mice, Inbred C57BL , Solubility , Stress, Mechanical , Stress, Physiological/drug effects
10.
Proc Natl Acad Sci U S A ; 109(48): 19638-43, 2012 Nov 27.
Article in English | MEDLINE | ID: mdl-23150542

ABSTRACT

The environments that harbor hematopoietic stem and progenitor cells are critical to explore for a better understanding of hematopoiesis during health and disease. These compartments often are inaccessible for controlled and rapid experimentation, thus limiting studies to the evaluation of conventional cell culture and transgenic animal models. Here we describe the manufacture and image-guided monitoring of an engineered microenvironment with user-defined properties that recruits hematopoietic progenitors into the implant. Using intravital imaging and fluorescence molecular tomography, we show in real time that the cell homing and retention process is efficient and durable for short- and long-term engraftment studies. Our results indicate that bone marrow stromal cells, precoated on the implant, accelerate the formation of new sinusoidal blood vessels with vascular integrity at the microcapillary level that enhances the recruitment hematopoietic progenitor cells to the site. This implantable construct can serve as a tool enabling the study of hematopoiesis.


Subject(s)
Hematopoietic Stem Cells/cytology , Neoplasms/pathology , Stem Cell Niche , Tissue Scaffolds , Tumor Microenvironment , Animals , Extracellular Matrix , Humans , Hydrogels , Mice , Mice, Inbred NOD , Mice, Nude , Microscopy, Confocal , Tomography/methods
11.
Nat Commun ; 3: 1054, 2012.
Article in English | MEDLINE | ID: mdl-22968700

ABSTRACT

Real-time imaging of moving organs and tissues at microscopic resolutions represents a major challenge in studying the complex biology of live animals. Here we present a technique based on a novel stabilizer setup combined with a gating acquisition algorithm for the imaging of a beating murine heart at the single-cell level. The method allows serial in vivo fluorescence imaging of the beating heart in live mice in both confocal and nonlinear modes over the course of several hours. We demonstrate the utility of this technique for in vivo optical sectioning and dual-channel time-lapse fluorescence imaging of cardiac ischaemia. The generic method could be adapted to other moving organs and thus broadly facilitate in vivo microscopic investigations.


Subject(s)
Microscopy, Confocal/methods , Microscopy, Fluorescence, Multiphoton/methods , Myocardial Contraction/physiology , Algorithms , Animals , Heart , Mice
12.
Nature ; 487(7407): 325-9, 2012 Jul 19.
Article in English | MEDLINE | ID: mdl-22763456

ABSTRACT

During progression of atherosclerosis, myeloid cells destabilize lipid-rich plaques in the arterial wall and cause their rupture, thus triggering myocardial infarction and stroke. Survivors of acute coronary syndromes have a high risk of recurrent events for unknown reasons. Here we show that the systemic response to ischaemic injury aggravates chronic atherosclerosis. After myocardial infarction or stroke, Apoe-/- mice developed larger atherosclerotic lesions with a more advanced morphology. This disease acceleration persisted over many weeks and was associated with markedly increased monocyte recruitment. Seeking the source of surplus monocytes in plaques, we found that myocardial infarction liberated haematopoietic stem and progenitor cells from bone marrow niches via sympathetic nervous system signalling. The progenitors then seeded the spleen, yielding a sustained boost in monocyte production. These observations provide new mechanistic insight into atherogenesis and provide a novel therapeutic opportunity to mitigate disease progression.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/pathology , Myocardial Infarction/complications , Myocardial Infarction/pathology , Animals , Apolipoproteins E/genetics , Hematopoietic Stem Cells/cytology , Inflammation/complications , Mice , Mice, Inbred C57BL , Monocytes/cytology , Spleen/cytology , Stem Cells/cytology
13.
Neoplasia ; 14(5): 388-95, 2012 May.
Article in English | MEDLINE | ID: mdl-22745585

ABSTRACT

Identifying circulating tumor cells (CTCs) with greater sensitivity could facilitate early detection of cancer and rapid assessment of treatment response. Most current technologies use EpCAM expression as a CTC identifier. However, given that a significant fraction of cancer patients have low or even absent EpCAM levels, there is a need for better detection methods. Here, we hypothesize that a multimarker strategy combined with direct sensing of CTC in whole blood would increase the detection of CTC in patients. Accordingly, molecular profiling of biopsies from a patient cohort revealed a four-marker set (EpCAM, HER-2, EGFR, and MUC-1) capable of effectively differentiating cancer cells from normal host cells. Using a point-of-care micro-nuclear magnetic resonance (µNMR) system, we consequently show that this multimarker combination readily detects individual CTC directly in whole blood without the need for primary purification. We also confirm these results in a comparative trial of patients with ovarian cancer. This platform could potentially benefit a broad range of applications in clinical oncology.


Subject(s)
Biomarkers, Tumor/metabolism , Magnetic Resonance Spectroscopy/methods , Neoplasms/diagnosis , Neoplasms/metabolism , Neoplastic Cells, Circulating/metabolism , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Female , Humans , Magnetic Resonance Spectroscopy/instrumentation , Male , Middle Aged , Sensitivity and Specificity
14.
Eur J Immunol ; 42(9): 2343-53, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22733595

ABSTRACT

Ligands of the B7 family provide both positive and negative costimulatory signals to the CD28 family of receptors on T lymphocytes, the balance of which determines the immune response. B7-H3 is a member of the B7 family whose function in T-cell activation has been the subject of some controversy: in autoimmunity and tumor immunity, it has been described as both costimulatory and coinhibitory, while in transplantation, B7-H3 signaling is thought to contribute to graft rejection. However, we now demonstrate results to the contrary. Signaling through a putative B7-H3 receptor prolonged allograft survival in a fully MHC-mismatched cardiac model and promoted a shift toward a Th2 milieu; conversely, B7-H3 blockade, achieved by use of a blocking antibody, resulted in accelerated rejection, an effect associated with enhanced IFN-γ production. Finally, graft prolongation achieved by CTLA4 Ig was shortened both by B7-H3 blockade and the absence of recipient B7-H3. These findings suggest a coinhibitory role for B7-H3. However, experience with other CD28/B7 family members suggests that immune redundancy plays a crucial role in determining the functions of various pathways. Given the abundance of conflicting data, it is plausible that, under differing conditions, B7-H3 may have both positive and negative costimulatory functions.


Subject(s)
B7 Antigens/immunology , Heart Transplantation/immunology , Signal Transduction/immunology , Th1 Cells/immunology , Transplantation, Homologous/immunology , Abatacept , Animals , B7 Antigens/metabolism , CD28 Antigens/immunology , CD28 Antigens/metabolism , Graft Rejection/immunology , Graft Rejection/metabolism , Immunoconjugates/immunology , Immunoconjugates/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Kinetics , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th1 Cells/metabolism , Th2 Cells/immunology , Th2 Cells/metabolism , Transplantation Immunology
15.
Cancer Res ; 72(12): 2949-56, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22505651

ABSTRACT

Observing drug responses in the tumor microenvironment in vivo can be technically challenging. As a result, cellular responses to molecularly targeted cancer drugs are often studied in cell culture, which does not accurately represent the behavior of cancer cells growing in vivo. Using high-resolution microscopy and fluorescently labeled genetic reporters for apoptosis, we developed an approach to visualize drug-induced cell death at single-cell resolution in vivo. Stable expression of the mitochondrial intermembrane protein IMS-RP was established in human breast and pancreatic cancer cells. Image analysis was then used to quantify release of IMS-RP into the cytoplasm upon apoptosis and irreversible mitochondrial permeabilization. Both breast and pancreatic cancer cells showed higher basal apoptotic rates in vivo than in culture. To study drug-induced apoptosis, we exposed tumor cells to navitoclax (ABT-263), an inhibitor of Bcl-2, Bcl-xL, and Bcl-w, both in vitro and in vivo. Although the tumors responded to Bcl-2 inhibition in vivo, inducing apoptosis in around 20% of cancer cells, the observed response was much higher in cell culture. Together, our findings show an imaging technique that can be used to directly visualize cell death within the tumor microenvironment in response to drug treatment.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis , Breast Neoplasms/physiopathology , Mitochondrial Membranes/physiology , Molecular Imaging/methods , Pancreatic Neoplasms/physiopathology , Sulfonamides/pharmacology , Animals , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Green Fluorescent Proteins , Humans , Image Processing, Computer-Assisted , Mice , Mice, Nude , Mitochondrial Membranes/drug effects , Mitochondrial Proteins/metabolism , Pancreatic Neoplasms/drug therapy , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Recombinant Fusion Proteins/metabolism , Single-Cell Analysis , Tumor Microenvironment
16.
PLoS One ; 7(4): e34427, 2012.
Article in English | MEDLINE | ID: mdl-22509302

ABSTRACT

To date there is a lack of tools to map the spatio-temporal dynamics of diverse cells in experimental heart models. Conventional histology is labor intensive with limited coverage, whereas many imaging techniques do not have sufficiently high enough spatial resolution to map cell distributions. We have designed and built a high resolution, dual channel Born-normalized near-infrared fluorescence optical projection tomography system to quantitatively and spatially resolve molecular agents distribution within whole murine heart. We validated the use of the system in a mouse model of monocytes/macrophages recruitment during myocardial infarction. While acquired, data were processed and reconstructed in real time. Tomographic analysis and visualization of the key inflammatory components were obtained via a mathematical formalism based on left ventricular modeling. We observed extensive monocyte recruitment within and around the infarcted areas and discovered that monocytes were also extensively recruited into non-ischemic myocardium, beyond that of injured tissue, such as the septum.


Subject(s)
Molecular Probes/metabolism , Myocardium/metabolism , Tomography, Optical/methods , Animals , Biocompatible Materials/chemistry , Biocompatible Materials/metabolism , Biological Transport , Cathepsin B/metabolism , Dextrans/chemistry , Image Processing, Computer-Assisted , Mice , Molecular Probes/chemistry , Myocardial Infarction/metabolism , Nanoparticles/chemistry , Time Factors
17.
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
18.
Science ; 335(6068): 597-601, 2012 Feb 03.
Article in English | MEDLINE | ID: mdl-22245738

ABSTRACT

Recognition and clearance of a bacterial infection are a fundamental properties of innate immunity. Here, we describe an effector B cell population that protects against microbial sepsis. Innate response activator (IRA) B cells are phenotypically and functionally distinct, develop and diverge from B1a B cells, depend on pattern-recognition receptors, and produce granulocyte-macrophage colony-stimulating factor. Specific deletion of IRA B cell activity impairs bacterial clearance, elicits a cytokine storm, and precipitates septic shock. These observations enrich our understanding of innate immunity, position IRA B cells as gatekeepers of bacterial infection, and identify new treatment avenues for infectious diseases.


Subject(s)
B-Lymphocyte Subsets/immunology , Escherichia coli Infections/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunity, Innate , Peritonitis/immunology , Sepsis/immunology , Animals , B-Lymphocyte Subsets/metabolism , Cell Lineage , Cell Separation , Female , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Immunoglobulin M/metabolism , Immunophenotyping , Integrin alpha4beta1/immunology , Integrin alpha4beta1/metabolism , Lipopolysaccharides , Lymphocyte Activation , Lymphocyte Function-Associated Antigen-1/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Mice, Inbred C57BL , Parabiosis , Shock, Septic/immunology , Spleen/immunology , Toll-Like Receptor 4/immunology
19.
J Exp Med ; 209(1): 123-37, 2012 Jan 16.
Article in English | MEDLINE | ID: mdl-22213805

ABSTRACT

Monocytes (Mo) and macrophages (MΦ) are emerging therapeutic targets in malignant, cardiovascular, and autoimmune disorders. Targeting of Mo/MΦ and their effector functions without compromising innate immunity's critical defense mechanisms first requires addressing gaps in knowledge about the life cycle of these cells. Here we studied the source, tissue kinetics, and clearance of Mo/MΦ in murine myocardial infarction, a model of acute inflammation after ischemic injury. We found that a) Mo tissue residence time was surprisingly short (20 h); b) Mo recruitment rates were consistently high even days after initiation of inflammation; c) the sustained need of newly made Mo was fostered by extramedullary monocytopoiesis in the spleen; d) splenic monocytopoiesis was regulated by IL-1ß; and e) the balance of cell recruitment and local death shifted during resolution of inflammation. Depending on the experimental approach, we measured a 24 h Mo/MΦ exit rate from infarct tissue between 5 and 13% of the tissue cell population. Exited cells were most numerous in the blood, liver, and spleen. Abrogation of extramedullary monocytopoiesis proved deleterious for infarct healing and accelerated the evolution of heart failure. We also detected rapid Mo kinetics in mice with stroke. These findings expand our knowledge of Mo/MΦ flux in acute inflammation and provide the groundwork for novel anti-inflammatory strategies for treating heart failure.


Subject(s)
Hematopoiesis, Extramedullary , Macrophages/physiology , Monocytes/cytology , Monocytes/physiology , Myocardial Infarction/physiopathology , Adoptive Transfer , Animals , Biomarkers/metabolism , Cell Death/genetics , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Kinetics , Macrophages/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Biological , Myeloid Cells/metabolism , Myocardial Infarction/immunology , Myocardial Infarction/pathology , Signal Transduction , Spleen/physiology , Stroke/immunology , Stroke/metabolism , Wound Healing/physiology
20.
J Am Coll Cardiol ; 59(2): 153-63, 2012 Jan 10.
Article in English | MEDLINE | ID: mdl-22222080

ABSTRACT

OBJECTIVES: The aim of this study was to explore post-myocardial infarction (MI) myocardial inflammation. BACKGROUND: Innate immune cells are centrally involved in infarct healing and are emerging therapeutic targets in cardiovascular disease; however, clinical tools to assess their presence in tissue are scarce. Furthermore, it is currently not known if the nonischemic remote zone recruits monocytes. METHODS: Acute inflammation was followed in mice with coronary ligation by 18-fluorodeoxyglucose ((18)FDG) positron emission tomography/magnetic resonance imaging, fluorescence-activated cell sorting, polymerase chain reaction, and histology. RESULTS: Gd-DTPA-enhanced infarcts showed high (18)FDG uptake on day 5 after MI. Cell depletion and isolation data confirmed that this largely reflected inflammation; CD11b(+) cells had 4-fold higher (18)FDG uptake than the infarct tissue from which they were isolated (p < 0.01). Surprisingly, there was considerable monocyte recruitment in the remote myocardium (approximately 10(4)/mg of myocardium, 5.6-fold increase; p < 0.01), a finding mirrored by macrophage infiltration in the remote myocardium of patients with acute MI. Temporal kinetics of cell recruitment were slower than in the infarct, with peak numbers on day 10 after ischemia. Quantitative polymerase chain reaction showed a robust increase of recruiting adhesion molecules and chemokines in the remote myocardium (e.g., 12-fold increase of monocyte chemoattractant protein-1), although levels were always lower than in the infarct. Finally, matrix metalloproteinase activity was significantly increased in noninfarcted myocardium, suggesting that monocyte recruitment to the remote zone may contribute to post-MI dilation. CONCLUSIONS: This study shed light on the innate inflammatory response in remote myocardium after MI.


Subject(s)
Macrophages/physiology , Monocytes/physiology , Myocardial Infarction/diagnostic imaging , Myocarditis/diagnostic imaging , Aged , Animals , Case-Control Studies , Female , Fluorodeoxyglucose F18 , Humans , Magnetic Resonance Imaging , Male , Matrix Metalloproteinase 2/metabolism , Matrix Metalloproteinase 9/metabolism , Mice , Middle Aged , Myocardial Infarction/enzymology , Myocardial Infarction/immunology , Myocarditis/enzymology , Myocarditis/immunology , Positron-Emission Tomography , Radiopharmaceuticals
SELECTION OF CITATIONS
SEARCH DETAIL