Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
Add more filters










Publication year range
1.
J Physiol ; 597(4): 1023-1032, 2019 02.
Article in English | MEDLINE | ID: mdl-30022479

ABSTRACT

Both systemic (tracheal and bronchial) and pulmonary circulations perfuse the lung. However, documentation of angiogenesis of either is complicated by the presence of the other. Well-documented angiogenesis of the systemic circulations have been identified in asthma, cystic fibrosis, chronic thromboembolism and primary carcinomas. Angiogenesis of the vasa vasorum, which are branches of bronchial arteries, is seen in the walls of large pulmonary vessels after a period of chronic hypoxia. Documentation of increased pulmonary capillaries has been shown in models of chronic hypoxia, after pneumonectomy and in some carcinomas. Although endothelial cell proliferation may occur as part of the repair process in several pulmonary diseases, it is separate from the unique establishment of new functional perfusing networks defined as angiogenesis. Identification of the mechanisms driving the expansion of new vascular beds in the adult needs further investigation. Yet the growth factors and molecular mechanisms of lung angiogenesis remain difficult to separate from underlying disease sequelae.


Subject(s)
Lung/blood supply , Neovascularization, Physiologic , Animals , Blood Vessels/growth & development , Blood Vessels/physiology , Humans , Lung/physiology
2.
Physiol Rep ; 6(11): e13721, 2018 06.
Article in English | MEDLINE | ID: mdl-29894584

ABSTRACT

The importance of myeloid cells in promoting neovascularization has been shown in a number of pathological settings in several organs. However, the specific role of macrophages in promoting systemic angiogenesis during pulmonary ischemia is not fully determined. Our past work suggested that cells of monocytic lineage contributed to systemic angiogenesis in the lung since clodronate-induced depletion of all macrophages resulted in attenuated neovascularization. Our current goals were to define the population of macrophages important for systemic vessel growth into the lung after the onset of pulmonary ischemia in mice. Interstitial macrophages (CD64+ MerTK+ CD11b+ ) increased significantly as did the percent of CD45+ Ly6G+ neutrophils 1 day after the induction of left lung ischemia, despite the fact there was limited cell recruitment due to complete obstruction of the left pulmonary artery in this ischemia model. Since both interstitial macrophages and neutrophils express CD11b, we used CD11b+ DTR mice and showed the critical role for these cells since CD11b+ depleted mice showed no systemic angiogenesis 7 days after the onset of ischemia when compared to control mice. Coculture of mouse aortic endothelial cells with macrophages showed increased proliferation relative to endothelial cells in culture without inflammatory cells, or pulmonary artery endothelial cells. We conclude that CD11b+ leukocytes, trapped within the lung at the onset of ischemia, contribute to growth factor release, and are critical for new blood vessel proliferation.


Subject(s)
Ischemia/complications , Lung/blood supply , Lung/immunology , Macrophages/immunology , Neovascularization, Pathologic , Animals , CD11b Antigen/immunology , Cells, Cultured , Cytokines/immunology , Endothelial Cells/immunology , Inflammation/immunology , Leukocytes/immunology , Male , Mice, Inbred C57BL , Pulmonary Artery/immunology
3.
Theranostics ; 7(2): 377-389, 2017.
Article in English | MEDLINE | ID: mdl-28042341

ABSTRACT

Although angiogenesis is a hallmark feature of asthmatic inflammatory responses, therapeutic anti-angiogenesis interventions have received little attention. Objective: Assess the effectiveness of anti-angiogenic Sn2 lipase-labile prodrugs delivered via αvß3-micellar nanotherapy to suppress microvascular expansion, bronchial remodeling, and airway hyper-responsiveness in Brown Norway rats exposed to serial house dust mite (HDM) inhalation challenges. Results: Anti-neovascular effectiveness of αvß3-mixed micelles incorporating docetaxel-prodrug (Dxtl-PD) or fumagillin-prodrug (Fum-PD) were shown to robustly suppress neovascular expansion (p<0.01) in the upper airways/bronchi of HDM rats using simultaneous 19F/1H MR neovascular imaging, which was corroborated by adjunctive fluorescent microscopy. Micelles without a drug payload (αvß3-No-Drug) served as a carrier-only control. Morphometric measurements of HDM rat airway size (perimeter) and vessel number at 21d revealed classic vascular expansion in control rats but less vascularity (p<0.001) after the anti-angiogenic nanotherapies. CD31 RNA expression independently corroborated the decrease in airway microvasculature. Methacholine (MCh) induced respiratory system resistance (Rrs) was high in the HDM rats receiving αvß3-No-Drug micelles while αvß3-Dxtl-PD or αvß3-Fum-PD micelles markedly and equivalently attenuated airway hyper-responsiveness and improved airway compliance. Total inflammatory BAL cells among HDM challenged rats did not differ with treatment, but αvß3+ macrophages/monocytes were significantly reduced by both nanotherapies (p<0.001), most notably by the αvß3-Dxtl-PD micelles. Additionally, αvß3-Dxtl-PD decreased BAL eosinophil and αvß3+ CD45+ leukocytes relative to αvß3-No-Drug micelles, whereas αvß3-Fum-PD micelles did not. Conclusion: These results demonstrate the potential of targeted anti-angiogenesis nanotherapy to ameliorate the inflammatory hallmarks of asthma in a clinically relevant rodent model.


Subject(s)
Airway Remodeling , Angiogenesis Inhibitors/administration & dosage , Asthma/drug therapy , Asthma/pathology , Nanostructures/administration & dosage , Animals , Asthma/diagnostic imaging , Cyclohexanes/administration & dosage , Disease Models, Animal , Docetaxel , Drug Carriers/administration & dosage , Fatty Acids, Unsaturated/administration & dosage , Magnetic Resonance Imaging , Microscopy, Fluorescence , Prodrugs/administration & dosage , Pyroglyphidae/pathogenicity , Rats , Sesquiterpenes/administration & dosage , Taxoids/administration & dosage , Treatment Outcome
4.
Angiogenesis ; 20(1): 73-84, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27787629

ABSTRACT

Although bronchial angiogenesis has been well documented in allergic asthma, lymphangiogenesis has not been widely studied. Therefore, we evaluated changes in lung lymphatics in a rat model of allergen-induced asthma using house dust mite (Der p 1; 100 µg/challenge). Additionally, properties of isolated lung lymphatic endothelial cells (CD45-, CD141+, LYVE-1+, Prox-1+) were studied in vitro. Three weeks after the onset of intranasal allergen exposure (twice-weekly), an increase in the number of lung lymphatic vessels was measured (34% increase) by lung morphometry. New lymphatic structures were seen predominantly in the peribronchial and periarterial interstitial space but also surrounding large airways. Isolated lymphatic endothelial cells from sensitized lungs showed enhanced proliferation (% Ki67+), chemotaxis, and tube formation (number and length) compared to lymphatic endothelial cells isolated from naive rat lungs. This hyper-proliferative lymphangiogenic phenotype was preserved through multiple cell passages (2-8). Lymphatic endothelial cells isolated from naive and HDM-sensitized rats produced similar in vitro levels of VEGF-C, VEGF-D, and VEGFR3 protein, each recognized as critical lymphangiogenic factors. Inhibition with anti-VEGFR (axitinib, 0.1 µM) blocked proliferation and chemotaxis. Results suggest that in vivo sensitization causes fundamental changes to lymphatic endothelium, which are retained in vitro, and may relate to VEGFR downstream signaling.


Subject(s)
Asthma/physiopathology , Lymphangiogenesis , Animals , Asthma/parasitology , Asthma/pathology , Cell Proliferation , Disease Models, Animal , Endothelial Cells/metabolism , Frozen Sections , Lung/pathology , Male , Pyroglyphidae/physiology , Rats, Inbred BN , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor D/metabolism
5.
Cancer Res ; 76(20): 5962-5969, 2016 10 15.
Article in English | MEDLINE | ID: mdl-27569207

ABSTRACT

Angiogenesis is vital for tumor growth but in well-vascularized organs such as the lung its importance is unclear. This situation is complicated by the fact that the lung has two separate circulations, the pulmonary and the systemic bronchial circulation. There are few relevant animal models of non-small cell lung cancer, which can be used to study the lung's complex circulations, and mice, lacking a systemic bronchial circulation cannot be used. We report here a novel orthotopic model of non-small cell lung cancer in rats, where we have studied the separate contributions of each of the two circulations for lung tumor growth. Results show that bronchial artery perfusion, quantified by fluorescent microspheres (206% increase in large tumors) or high-resolution computed tomography scans (276% increase in large tumors), parallels the growth in tumor volume, whereas pulmonary artery perfusion remained unchanged. Ablation of the bronchial artery after the initiation of tumor growth resulted in a decrease in tumor volume over a subsequent course of 4 weeks. These results demonstrate that although the existing pulmonary circulation can supply the metabolic needs for tumor initiation, further growth of the tumor requires angiogenesis from the highly proliferative bronchial circulation. This model may be useful to investigate new therapeutic approaches that target specifically the bronchial circulation. Cancer Res; 76(20); 5962-9. ©2016 AACR.


Subject(s)
Adenocarcinoma/pathology , Bronchial Arteries/physiology , Lung Neoplasms/pathology , Neovascularization, Physiologic , Adenocarcinoma/blood supply , Adenocarcinoma of Lung , Animals , Cell Line, Tumor , Endothelial Cells/physiology , Humans , Lung Neoplasms/blood supply , Pulmonary Artery/physiology , Pulmonary Circulation , Rats , Tomography, X-Ray Computed , Tumor Burden
6.
Am J Respir Cell Mol Biol ; 54(3): 394-401, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26244419

ABSTRACT

Lymphocytes have been shown to modulate angiogenesis. Our previous work showed that T regulatory (Treg) cell depletion prevented angiogenesis. In the present study, we sought to examine T-cell populations during lung angiogenesis and subsequent angiostasis. In a mouse model of ischemia-induced systemic angiogenesis in the lung, we examined the time course (0-35 d) of neovascularization and T-cell phenotypes within the lung after left pulmonary artery ligation (LPAL). T cells increased and reached a maximum by 10 days after LPAL and then progressively decreased, suggestive of a modulatory role during the early phase of new vessel growth. Because others have shown IFN-γ to be angiostatic in tumor models, we focused on this effector T-cell cytokine to control the magnitude of angiogenesis. Results showed that IFN-γ protein is secreted at low levels after LPAL and that mice required Treg depletion to see the full effect of effector T cells. Using Foxp3(DTR) and diphtheria toxin to deplete T regulatory cells, increased numbers of effector T cells (CD8(+)) and/or increased capacity to secrete the prominent angiostatic cytokine IFN-γ (CD4(+)) were seen. In vitro culture of mouse systemic and pulmonary microvascular endothelial cells with IFN-γ showed increased endothelial cell apoptosis. CD8(-/-) mice and IFN-γR(-/-) mice showed enhanced angiogenesis compared with wild-type mice, confirming that, in this model, IFN-γ limits the extent of systemic neovascularization in the lung.


Subject(s)
Endothelial Cells/immunology , Ischemia/immunology , Lung/blood supply , Lung/immunology , Neovascularization, Physiologic , T-Lymphocytes, Regulatory/immunology , Animals , Apoptosis , CD4 Antigens/genetics , CD4 Antigens/immunology , CD4 Antigens/metabolism , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8 Antigens/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Endothelial Cells/pathology , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Ischemia/genetics , Ischemia/metabolism , Ischemia/pathology , Ischemia/physiopathology , Lung/metabolism , Lung/pathology , Lymphocyte Activation , Male , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Interferon/genetics , Receptors, Interferon/immunology , Receptors, Interferon/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Time Factors , Interferon gamma Receptor
7.
Am J Respir Cell Mol Biol ; 52(5): 603-10, 2015 May.
Article in English | MEDLINE | ID: mdl-25275926

ABSTRACT

Angiogenesis in ischemic organs is modulated by immune cells. Systemic neovascularization of the ischemic lung requires macrophages, with chemokines playing a central role in new vessel growth. Because regulatory T (Treg) cells modulate tumor-induced neovascularization, we questioned whether this CD4(+) lymphocyte subset impacts blood vessel growth during ischemia. In a model of left lung ischemia, an increase in CD4(+) CD25(+) forkhead homeobox protein-3 (Foxp3)(+) cells was observed 3-5 days after the onset of ischemia in wild-type C57Bl/6 mice. Using transgenic mice where Foxp3(+) Treg cells can be depleted with diphtheria toxin (DT; Foxp3(DTR)), we unexpectedly found that Foxp3(+) Treg depletion led to markedly reduced lung angiogenesis (90% reduction from Foxp3(gfp) controls). Adoptive transfer studies using CD4(+) CD25(+) splenocytes from congenic CD45.1 mice into Foxp3(+) Treg-depleted mice showed an almost complete recovery of the angiogenic phenotype (80% of Foxp3(gfp) controls). A survey of lung gene expression of angiogenic (lipopolysaccharide-induced CXC chemokine [LIX], IL-6, IL-17) and angiostatic (IFN-γ, transforming growth factor-ß, IL-10) cytokines showed Treg-dependent differences only in LIX (CXCL5) and IL-6. Protein confirmation demonstrated a significant reduction in LIX in Treg-deficient mice compared with controls 5 days after the onset of ischemia. Phenotyping other inflammatory cells in the lung by multicolor flow cytometry demonstrated a significantly reduced number of macrophages (major histocombatibility complex class II [MHCII](int), CD11C(+)) in Treg-deficient lungs compared with Treg-sufficient lungs. Treg cells are essential for maximal systemic angiogenesis after pulmonary ischemia. One likely mechanism responsible for the decrease in angiogenesis in Treg-depleted mice was the decline in the essential CXC chemokine, LIX.


Subject(s)
Forkhead Transcription Factors/metabolism , Ischemia/metabolism , Lung/blood supply , Neovascularization, Physiologic , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Angiogenic Proteins/genetics , Angiogenic Proteins/metabolism , Animals , Chemokine CXCL5/genetics , Chemokine CXCL5/metabolism , Disease Models, Animal , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Ischemia/genetics , Ischemia/immunology , Ischemia/physiopathology , Macrophages/immunology , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/transplantation , Time Factors
8.
Angiogenesis ; 18(1): 1-11, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25149641

ABSTRACT

Expanded and aberrant bronchial vascularity, a prominent feature of the chronic asthmatic airway, might explain persistent airway wall edema and sustained leukocyte recruitment. Since it is well established that there are causal relationships between exposure to house dust mite (HDM) and the development of asthma, determining the effects of HDM in rats, mammals with a bronchial vasculature similar to humans, provides an opportunity to study the effects of bronchial angiogenesis on airway function directly. We studied rats exposed bi-weekly to HDM (Der p 1; 50 µg/challenge by intranasal aspiration, 1, 2, 3 weeks) and measured the time course of appearance of increased blood vessels within the airway wall. Results demonstrated that within 3 weeks of HDM exposure, the number of vessels counted within airway walls of bronchial airways (0.5-3 mm perimeter) increased significantly. These vascular changes were accompanied by increased airway responsiveness to methacholine. A shorter exposure regimen (2 weeks of bi-weekly exposure) was insufficient to cause a significant increase in functional vessels or reactivity. Yet, 19F/1H MR imaging at 3T following αvß3-targeted perfluorocarbon nanoparticle infusion revealed a significant increase in 19F signal in rat airways after 2 weeks of bi-weekly HDM, suggesting earlier activation of the process of neovascularization. Although many antigen-induced mouse models exist, mice lack a bronchial vasculature and consequently lack the requisite human parallels to study bronchial edema. Overall, our results provide an important new model to study the impact of bronchial angiogenesis on chronic inflammation and airways hyperreactivity.


Subject(s)
Asthma/physiopathology , Bronchial Hyperreactivity/physiopathology , Disease Models, Animal , Neovascularization, Pathologic/parasitology , Pyroglyphidae/pathogenicity , Airway Resistance/physiology , Analysis of Variance , Animals , Bronchial Arteries/pathology , Bronchial Hyperreactivity/parasitology , DNA Primers/genetics , Fluorocarbons , Lung/pathology , Magnetic Resonance Imaging , Methacholine Chloride , Nanoparticles , Rats , Real-Time Polymerase Chain Reaction , Silicone Elastomers , Time Factors
9.
PLoS One ; 9(9): e106092, 2014.
Article in English | MEDLINE | ID: mdl-25181540

ABSTRACT

The process of leukocyte recruitment to the airways in real time has not been extensively studied, yet airway inflammation persists as a major contributor to lung pathology. We showed previously in vivo, that neutrophils are recruited acutely to the large airways after periods of airway distension imposed by the application of positive end-expiratory pressure (PEEP). Given extensive literature implicating products of nitric oxide synthase (NOS) in lung injury after ventilatory over-distension, we questioned whether similar mechanisms exist in airway post-capillary venules. Yet, endothelial nitric oxide has been shown to be largely anti-inflammatory in other systemic venules. Using intravital microscopy to visualize post-capillary tracheal venules in anesthetized, ventilated mice, the number of adherent leukocytes was significantly decreased in eNOS-/- mice under baseline conditions (2±1 cell/60 min observation) vs wild type (WT) C57BL/6 mice (7±2 cells). After exposure to PEEP (8 cmH2O for 1 min; 5 times), adherent cells increased significantly (29±5 cells) in WT mice while eNOS-/- mice demonstrated a significantly decreased number of adherent cells (11±4 cells) after PEEP. A similar response was seen when thrombin was used as the pro-inflammatory stimulus. In addition, mouse tracheal venular endothelial cells studied in vitro after exposure to cyclic stretch (18% elongation) or thrombin both demonstrated increased p-selectin expression that was significantly attenuated by NG-nitro-L-arginine methyl ester, N-acetylcysteine amide (NACA) and excess BH4. In vivo treatment with the ROS inhibitor NACA or co-factor BH4 abolished completely the PEEP-induced leukocyte adherence. These results suggest that pro-inflammatory stimuli cause leukocyte recruitment to tracheal endothelium in part due to eNOS uncoupling.


Subject(s)
Leukocytes/enzymology , Leukocytes/pathology , Nitric Oxide Synthase Type III/metabolism , Stress, Mechanical , Trachea/pathology , Venules/pathology , Animals , Cell Adhesion/drug effects , Leukocytes/drug effects , Male , Mice, Inbred C57BL , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/deficiency , P-Selectin/metabolism , Phosphorylation/drug effects , Positive-Pressure Respiration , Reactive Oxygen Species/metabolism , Thrombin/pharmacology
10.
Pulm Circ ; 4(2): 260-8, 2014 Jun.
Article in English | MEDLINE | ID: mdl-25006445

ABSTRACT

Abnormal lung microvascular endothelial vascular barrier function may contribute to pulmonary inflammation, such as that occurring during inhalation of cigarette smoke (CS). Cystic fibrosis transmembrane conductance regulator (CFTR), an anion channel expressed in both epithelial and endothelial cells, regulates the organization of tight junctions between epithelial cells and has also been implicated in the transport of sphingosine-1 phosphate (S1P), a vascular barrier-enhancing sphingolipid. Because CS has been shown to affect CFTR function, we hypothesized that CFTR function contributes to lung endothelial cell barrier and that CFTR dysfunction worsens CS-induced injury. CFTR inhibitors GlyH-101 or CFTRinh172 caused a dose-dependent increase in pulmonary or bronchial endothelial monolayer permeability, which peaked after 4 hours. CFTR inhibition was associated with both intercellular gaps and actin stress fiber formation compared with vehicle-treated cells. Increasing endothelial S1P, either by exogenous treatment or by inhibition of its degradation, significantly improved the barrier function in CFTR-inhibited monolayers. Both cultured lung endothelia and the lung microcirculation visualized in vivo with intravital two-photon imaging of transgenic mice deficient in CFTR showed that CFTR dysfunction increased susceptibility to CS-induced permeability. These results suggested that CFTR function might be required for lung endothelial barrier, including adherence junction stability. Loss of CFTR function, especially concomitant to CS exposure, might promote lung inflammation by increasing endothelial cell permeability, which could be ameliorated by S1P.

11.
Angiogenesis ; 17(1): 51-60, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23918207

ABSTRACT

Angiogenesis is an important constituent of many inflammatory pulmonary diseases, which has been unappreciated until recently. Early neovascular expansion in the lungs in preclinical models and patients is very difficult to assess noninvasively, particularly quantitatively. The present study demonstrated that (19)F/(1)H MR molecular imaging with αvß3-targeted perfluorocarbon nanoparticles can be used to directly measure neovascularity in a rat left pulmonary artery ligation (LPAL) model, which was employed to create pulmonary ischemia and induce angiogenesis. In rats 3 days after LPAL, simultaneous (19)F/(1)H MR imaging at 3T revealed a marked (19)F signal in animals 2 h following αvß3-targeted perfluorocarbon nanoparticles [(19)F signal (normalized to background) = 0.80 ± 0.2] that was greater (p = 0.007) than the non-targeted (0.30 ± 0.04) and the sham-operated (0.07 ± 0.09) control groups. Almost no (19)F signal was found in control right lung with any treatment. Competitive blockade of the integrin-targeted particles greatly decreased the (19)F signal (p = 0.002) and was equivalent to the non-targeted control group. Fluorescent and light microscopy illustrated heavy decorating of vessel walls in and around large bronchi and large pulmonary vessels. Focal segmental regions of neovessel expansion were also noted in the lung periphery. Our results demonstrate that (19)F/(1)H MR molecular imaging with αvß3-targeted perfluorocarbon nanoparticles provides a means to assess the extent of systemic neovascularization in the lung.


Subject(s)
Contrast Media/pharmacology , Fluorocarbons/pharmacology , Ischemia , Lung Diseases , Magnetic Resonance Angiography/methods , Nanoparticles , Neovascularization, Physiologic , Animals , Integrin alphaVbeta3/metabolism , Ischemia/diagnostic imaging , Ischemia/metabolism , Isotopes/pharmacology , Lung Diseases/diagnostic imaging , Lung Diseases/metabolism , Male , Radiography , Rats , Rats, Sprague-Dawley
12.
PLoS One ; 8(6): e66432, 2013.
Article in English | MEDLINE | ID: mdl-23776670

ABSTRACT

Angiogenesis in the lung involves the systemic bronchial vasculature and becomes prominent when chronic inflammation prevails. Mechanisms for neovascularization following pulmonary ischemia include growth factor transit from ischemic parenchyma to upstream bronchial arteries, inflammatory cell migration/recruitment through the perfusing artery, and paracrine effects of lung cells within the left bronchus, the niche where arteriogenesis takes place. We analyzed left lung bronchoalveolar lavage (BAL) fluid and left bronchus homogenates after left pulmonary artery ligation (LPAL) in rats, immediately after the onset of ischemia (0 h), 6 h and 24 h later. Additionally, we tested the effectiveness of dexamethasone on decreasing inflammation (0-24 h LPAL) and angiogenesis at early (3 d LPAL; bronchial endothelial proliferation) and late (14 d LPAL; blood flow) stages. After LPAL (6 h), BAL protein, total inflammatory cells, macrophages, and polymorphonuclear cells increased significantly. In parallel, pro-angiogenic CXC chemokines increased in BAL and the left main-stem bronchus (CXCL1) or only within the bronchus (CXCL2). Dexamethasone treatment reduced total BAL protein, inflammatory cells (total and polymorphonuclear cells), and CXCL1 but not CXCL2 in BAL. By contrast, no decrease was seen in either chemokine within the bronchial tissue, in proliferating bronchial endothelial cells, or in systemic perfusion of the left lung. Our results confirm the presence of CXC chemokines within BAL fluid as well as within the left mainstem bronchus. Despite significant reduction in lung injury and inflammation with dexamethasone treatment, chemokine expression within the bronchial tissue as well as angiogenesis were not affected. Our results suggest that early changes within the bronchial niche contribute to subsequent neovascularization during pulmonary ischemia.


Subject(s)
Bronchi/metabolism , Bronchi/pathology , Chemokines/metabolism , Animals , Bronchi/drug effects , Bronchial Diseases/metabolism , Cell Proliferation , Chemokine CXCL1/metabolism , Chemokine CXCL2/metabolism , Dexamethasone/therapeutic use , Endothelial Cells/cytology , Lung/drug effects , Lung/metabolism , Lung/pathology , Male , Neovascularization, Pathologic/drug therapy , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction
13.
Respir Res ; 13: 93, 2012 Oct 12.
Article in English | MEDLINE | ID: mdl-23061826

ABSTRACT

BACKGROUND: Systemic neovascularization of the lung during chronic ischemia has been observed in all mammals studied. However, the proteins that orchestrate the complex interaction of new vessel growth and tunneling through lung tissue matrix have not been described. Although previous work has demonstrated the CXC chemokines are essential growth factors in the process of angiogenesis in mice and rats, key matrix proteins have not been identified. METHODS: Since the degradation of chemokines has been shown to be dependent on metalloproteinases (MMP), we first surveyed gene expression patterns (real time RT-PCR) of several lung matrix proteins in DBA/J (D2) mice and C57Bl/6 (B6) mice, strains known to have divergent parenchymal responses in other lung disease models. We studied changes in the time course of MMP-12 activity in D2 and B6 mice. Functional angiogenesis was determined 14 days after the onset of complete left lung ischemia induced by left pulmonary artery ligation (LPAL), using fluorescent microspheres. RESULTS: Our results confirmed higher levels of MMP-12 gene expression in D2 mice relative to B6, which corresponded to a phenotype of minimal systemic angiogenesis in D2 mice and more robust angiogenesis in B6 mice (p < 0.01). MMP-12 activity decreased over the course of 14 days in B6 mice whereas it increased in D2 mice (p < 0.05). MMP-12 was associated largely with cells expressing the macrophage marker F4/80. Genetic deficiency of MMP-12 resulted in significantly enhanced neovascularization (p < 0.01 from B6). CONCLUSION: Taken together, our results suggest macrophage-derived MMP-12 contributes to angiostasis in the ischemic lung.


Subject(s)
Ischemia/enzymology , Lung/blood supply , Lung/enzymology , Matrix Metalloproteinase 12/metabolism , Neovascularization, Physiologic , Animals , Antigens, Differentiation/metabolism , Biomarkers/metabolism , Disease Models, Animal , Gene Expression Profiling/methods , Gene Expression Regulation, Enzymologic , Ischemia/etiology , Ischemia/genetics , Ischemia/immunology , Ligation , Lung/immunology , Macrophages/enzymology , Macrophages/immunology , Matrix Metalloproteinase 12/deficiency , Matrix Metalloproteinase 12/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Pulmonary Artery/surgery , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Species Specificity , Time Factors
14.
PLoS One ; 6(11): e26716, 2011.
Article in English | MEDLINE | ID: mdl-22110592

ABSTRACT

Angiogenesis after pulmonary ischemia is initiated by reactive O(2) species and is dependent on CXC chemokine growth factors, and its magnitude is correlated with the number of lavaged macrophages. After complete obstruction of the left pulmonary artery in mice, the left lung is isolated from the peripheral circulation until 5-7 days later, when a new systemic vasculature invades the lung parenchyma. Consequently, this model offers a unique opportunity to study the differentiation and/or proliferation of monocyte-derived cells within the lung. In this study, we questioned whether macrophage subpopulations were differentially expressed and which subset contributed to growth factor release. We characterized the change in number of all macrophages (MHCII(int), CD11C+), alveolar macrophages (MHCII(int), CD11C+, CD11B-) and mature lung macrophages (MHCII(int), CD11C+, CD11B+) in left lungs from mice immediately (0 h) or 24 h after left pulmonary artery ligation (LPAL). In left lung homogenates, only lung macrophages increased 24 h after LPAL (vs. 0 h; p<0.05). No changes in proliferation were seen in any subset by PCNA expression (0 h vs. 24 h lungs). When the number of monocytic cells was reduced with clodronate liposomes, systemic blood flow to the left lung 14 days after LPAL decreased by 42% (p<0.01) compared to vehicle controls. Furthermore, when alveolar macrophages and lung macrophages were sorted and studied in vitro, only lung macrophages secreted the chemokine MIP-2α (ELISA). These data suggest that ischemic stress within the lung contributes to the differentiation of immature monocytes to lung macrophages within the first 24 h after LPAL. Lung macrophages but not alveolar macrophages increase and secrete the proangiogenic chemokine MIP-2α. Overall, an increase in the number of lung macrophages appears to be critical for neovascularization in the lung, since clodronate treatment decreased their number and attenuated functional angiogenesis.


Subject(s)
Ischemia , Lung/blood supply , Macrophages/cytology , Animals , Cell Differentiation , Cell Proliferation , In Vitro Techniques , Lung/physiopathology , Male , Mice , Mice, Inbred C57BL , Monocytes/cytology , Neovascularization, Physiologic , Phenotype
15.
Am J Physiol Lung Cell Mol Physiol ; 301(5): L782-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21821727

ABSTRACT

Hyaluronan (HA), a glycosaminoglycan critical to the lung extracellular matrix, has been shown to dissociate into low-molecular-weight (LMW) HA fragments following exposure to injurious stimuli. In the present study we questioned whether lung HA changed during ischemia and whether changes had an effect on subsequent angiogenesis. After left pulmonary artery ligation (LPAL) in mice, we analyzed left lung homogenates immediately after the onset of ischemia (0 h) and intermittently for 14 days. The relative expression of HA synthase (HAS)1, HAS2, and HAS3 was determined by real-time RT-PCR, total HA in the lung was measured by an ELISA-like assay, gel electrophoresis was performed to determine changes in HA size distribution, and the activity of hyaluronidases was determined by zymography. A 50% increase in total HA was measured 16 h after the onset of ischemia and remained elevated for up to 7 days. Furthermore, a fourfold increase in LMW HA fragments (495-30 kDa) was observed by 4 h after LPAL. Both HAS1 and HAS2 showed increased expression 4-16 h after LPAL, yet no changes were seen in hyaluronidase activity. These results suggest that both HA fragmentation and activation of HA synthesis contribute to increased HA levels during lung ischemia. Delivery of LMW HA fragments in an in vitro tube formation assay or directly to the ischemic mouse lung in vivo both resulted in increased angiogenesis. We conclude that ischemic injury results in matrix fragmentation, which leads to stimulation of neovascularization.


Subject(s)
Glucuronosyltransferase/metabolism , Hyaluronic Acid/metabolism , Ischemia/metabolism , Lung/metabolism , Neovascularization, Pathologic/metabolism , Pulmonary Artery/physiopathology , Animals , Collagen , Drug Combinations , Electrophoresis, Agar Gel , Enzyme-Linked Immunosorbent Assay , Gene Expression/drug effects , Glucuronosyltransferase/genetics , Hyaluronan Synthases , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Ischemia/physiopathology , Laminin , Lung/drug effects , Lung/pathology , Lung/physiopathology , Mice , Mice, Inbred C57BL , Molecular Weight , Proteoglycans , Pulmonary Artery/surgery , Real-Time Polymerase Chain Reaction
16.
J Appl Physiol (1985) ; 110(2): 538-44, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21148340

ABSTRACT

Bronchial vascular angiogenesis takes place in a variety of lung inflammatory conditions such as asthma, cystic fibrosis, lung cancer, and chronic pulmonary thromboembolic disease. However, it is unclear whether neovascularization is predominantly appropriate and preserves lung tissue or whether it contributes further to lung pathology through edema formation and inflammation. In the present study we examined airway and lung parenchymal function 14 days after left pulmonary artery ligation. In rats as well as higher mammals, severe pulmonary ischemia results in bronchial vascular proliferation. Using labeled microspheres, we demonstrated an 18-fold increase in systemic blood flow to the ischemic left lung. Additionally, vascular remodeling extended to the tracheal venules, which showed an average 28% increase in venular diameter. Despite this increase in vascularity, airways resistance was not altered nor was methacholine responsiveness. Since these measurements include the entire lung, we suggest that the normal right lung, which represented 78% of the total lung, obscured the ability to detect a change. When functional indexes such as diffusing capacity, in situ lung volume, and vascular permeability of the left lung could be separated from right lung, significant changes were observed. Thus when comparing average left lung values of rats 14 days after left pulmonary artery ligation to left lungs of rats undergoing sham surgery, diffusing capacity of the left lung decreased by 72%, left lung volume decreased by 38%, and the vascular permeability to protein increased by 58%. No significant differences in inflammatory cell recruitment were observed, suggesting that acute ischemic inflammation had resolved. We conclude that despite the preservation of lung tissue, the proliferating bronchial neovasculature may contribute to a sustained decrement in pulmonary function.


Subject(s)
Ischemia/physiopathology , Lung/blood supply , Lung/physiopathology , Neovascularization, Pathologic/physiopathology , Pulmonary Artery/physiopathology , Animals , Blood Flow Velocity , Chronic Disease , Ischemia/complications , Male , Neovascularization, Pathologic/etiology , Pulmonary Circulation , Rats , Rats, Sprague-Dawley
17.
J Appl Physiol (1985) ; 109(5): 1292-300, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20724568

ABSTRACT

The airways contain a dense subepithelial microvascular plexus that is involved in the supply and clearance of substances to and from the airway wall. We set out to test the hypothesis that airway smooth muscle reactivity to bronchoconstricting agents may be dependent on airway mucosal blood flow. Immunohistochemical staining identified vasoconstrictor and vasodilator nerve fibers associated with subepithelial blood vessels in the guinea pig airways. Intravital microscopy of the tracheal mucosal microvasculature in anesthetized guinea pigs revealed that blockade of α-adrenergic receptors increased baseline arteriole diameter by ~40%, whereas the α-adrenergic receptor agonist phenylephrine produced a modest (5%) vasoconstriction in excess of the baseline tone. In subsequent in vivo experiments, tracheal contractions evoked by topically applied histamine were significantly reduced (P < 0.05) and enhanced by α-adrenergic receptor blockade and activation, respectively. α-Adrenergic ligands produced similar significant (P < 0.05) effects on airway smooth muscle contractions evoked by topically administered capsaicin, intravenously administered neurokinin A, inhaled histamine, and topically administered antigen in sensitized animals. These responses were independent of any direct effect of α-adrenergic ligands on the airway smooth muscle tone. The data suggest that changes in blood flow in the vessels supplying the airways regulate the reactivity of the underlying airway smooth muscle to locally released and exogenously administered agents by regulating their clearance. We speculate that changes in mucosal vascular function or changes in neuronal regulation of the airway vasculature may contribute to airways responsiveness in disease.


Subject(s)
Bronchoconstriction , Microcirculation , Microvessels/innervation , Muscle, Smooth/physiology , Respiratory Mucosa/blood supply , Sympathetic Nervous System/physiology , Trachea/blood supply , Administration, Inhalation , Administration, Topical , Adrenergic alpha-Agonists/administration & dosage , Adrenergic alpha-Antagonists/administration & dosage , Animals , Bronchoconstriction/drug effects , Bronchoconstrictor Agents/administration & dosage , Dose-Response Relationship, Drug , Guinea Pigs , Injections, Intravenous , Male , Muscle, Smooth/drug effects , Neuropeptides/administration & dosage , Regional Blood Flow , Time Factors , Vasoconstriction , Vasodilation , Vasomotor System/physiology
18.
Am J Physiol Lung Cell Mol Physiol ; 299(4): L535-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20693319

ABSTRACT

Pulmonary artery obstruction and subsequent lung ischemia have been shown to induce systemic angiogenesis despite preservation of normoxia. The underlying mechanisms, however, remain poorly understood. In a mouse model of lung ischemia induced by left pulmonary artery ligation (LPAL), we showed previously, the formation of a new systemic vasculature to the ischemic lung. We hypothesize that LPAL in the mouse increases reactive oxygen species (ROS) production, and these molecules play an initiating role in subsequent lung neovascularization. We used oxidant-sensitive dyes (DHE and H(2)DCF-DA) to quantify ROS and measured the antioxidant-reduced glutathione (GSH) and its oxidized form (GSSG) as indicators of ROS levels after LPAL. The magnitude of systemic neovascularization was determined by measuring systemic blood flow to the left lung with radiolabeled microspheres 14 days after LPAL. An increase in ROS was observed early (30 min: 55% increase in H(2)DCF-DA) after LPAL, with a return to baseline by 24 h. GSH/GSSG was decreased (∼50%) 4 h after LPAL, suggesting earlier ROS upregulation. Mice treated with the antioxidant N-acetylcysteine showed attenuated angiogenesis (62% of wild-type LPAL), and mice lacking Nrf2, a transcription factor important for antioxidant synthesis, resulted in increased neovascularization (207% of wild-type LPAL). Overall, GSH/GSSG was inversely associated with the magnitude of neovascularization. These results demonstrate that LPAL induces an early and transient ROS upregulation, and ROS appear to play a role in promoting ischemia-induced angiogenesis.


Subject(s)
Disease Models, Animal , Ischemia/metabolism , Lung/blood supply , Neovascularization, Pathologic/metabolism , Reactive Oxygen Species/metabolism , Acetylcysteine/therapeutic use , Acetylcysteine/toxicity , Animals , Antioxidants/therapeutic use , Glutathione/metabolism , Glutathione Disulfide/metabolism , Ischemia/chemically induced , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism , Neovascularization, Pathologic/drug therapy , Pulmonary Artery/cytology , Pulmonary Artery/metabolism
19.
Microvasc Res ; 80(1): 18-22, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20144627

ABSTRACT

We showed previously in a mouse model of lung ischemia-induced angiogenesis, enhanced expression of the three ELR+ CXC chemokines (KC, LIX, and MIP-2) and that blockade of the ligand receptor CXCR(2) limited neovascularization. The present study was undertaken to determine the relative abundance and angiogenic potential of the three CXC chemokines and whether RhoA activation explained the measured differences in potencies. We found that LIX showed the greatest absolute amount in the in vivo model 4 h after left pulmonary artery obstruction (LIX>KC>MIP-2; p<0.05). In vitro, LIX induced the greatest degree of arterial endothelial cell chemotaxis and KC was without effect. A significant increase (approximately 40%) in active RhoA was observed with both LIX and MIP-2 compared with vehicle control (p<0.05). On average, LIX induced the greatest amount of tube formation within pleural tissue in culture. Thus, the results of the present study suggest that among the three ELR+ CXC chemokines, LIX predominates in eliciting a pro-angiogenic phenotype.


Subject(s)
Angiogenic Proteins/pharmacology , Chemokines, CXC/pharmacology , Neovascularization, Physiologic/drug effects , Angiogenic Proteins/metabolism , Animals , Antibodies/immunology , Antibodies/pharmacology , Aorta/cytology , Chemokine CXCL1/metabolism , Chemokine CXCL1/pharmacology , Chemokine CXCL2/metabolism , Chemokine CXCL2/pharmacology , Chemokine CXCL5/metabolism , Chemokine CXCL5/pharmacology , Chemokines, CXC/metabolism , Chemotaxis/drug effects , Endothelial Cells/cytology , Endothelial Cells/drug effects , Enzyme Inhibitors/pharmacology , Ischemia/metabolism , Ligation , Lung/drug effects , Lung/physiology , Male , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/physiology , Neuropeptides/antagonists & inhibitors , Neuropeptides/metabolism , Pulmonary Artery/surgery , Receptors, Interleukin-8A/antagonists & inhibitors , Receptors, Interleukin-8A/immunology , Receptors, Interleukin-8B/antagonists & inhibitors , Receptors, Interleukin-8B/immunology , Tissue Culture Techniques , rac GTP-Binding Proteins/antagonists & inhibitors , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein , rho GTP-Binding Proteins/antagonists & inhibitors , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
20.
J Breath Res ; 3(1): 016003, 2009 Mar.
Article in English | MEDLINE | ID: mdl-21383451

ABSTRACT

Reactive oxygen species are associated with tissue inflammation and injury. Our laboratory has demonstrated that ethane, a stable product of lipid peroxidation, in exhaled breath can be used to measure total body oxidative stress. An ischemia-reperfusion model of lung injury in sheep has been studied in which pulmonary and bronchial lung perfusion could be interrupted and restored. The goal of this study was to investigate whether two commercial formulations of propofol and the individual components of the commercial formulations attenuated the oxidative stress produced in this model. Breath ethane and breath carbon monoxide were measured as biomarkers of oxidative stress that occur at reperfusion of ischemic tissue. Data were analyzed by a standard least-squares-fit model. One of the formulations for propofol, which contained the preservative ethylenediaminetetraacetic acid (EDTA), was found to decrease the overall level of oxidative stress in sheep. Furthermore, while several models of severe lung injury demonstrate additional production of reactive oxygen species, our model of ischemia/reperfusion of lung tissue did not.

SELECTION OF CITATIONS
SEARCH DETAIL
...