Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Nat Immunol ; 20(1): 29-39, 2019 01.
Article in English | MEDLINE | ID: mdl-30538339

ABSTRACT

Macrophages promote both injury and repair after myocardial infarction, but discriminating functions within mixed populations remains challenging. Here we used fate mapping, parabiosis and single-cell transcriptomics to demonstrate that at steady state, TIMD4+LYVE1+MHC-IIloCCR2- resident cardiac macrophages self-renew with negligible blood monocyte input. Monocytes partially replaced resident TIMD4-LYVE1-MHC-IIhiCCR2- macrophages and fully replaced TIMD4-LYVE1-MHC-IIhiCCR2+ macrophages, revealing a hierarchy of monocyte contribution to functionally distinct macrophage subsets. Ischemic injury reduced TIMD4+ and TIMD4- resident macrophage abundance, whereas CCR2+ monocyte-derived macrophages adopted multiple cell fates within infarcted tissue, including those nearly indistinguishable from resident macrophages. Recruited macrophages did not express TIMD4, highlighting the ability of TIMD4 to track a subset of resident macrophages in the absence of fate mapping. Despite this similarity, inducible depletion of resident macrophages using a Cx3cr1-based system led to impaired cardiac function and promoted adverse remodeling primarily within the peri-infarct zone, revealing a nonredundant, cardioprotective role of resident cardiac macrophages.


Subject(s)
Macrophages/physiology , Myocardial Infarction/immunology , Myocardium/pathology , Animals , CX3C Chemokine Receptor 1/metabolism , Cell Differentiation , Cell Lineage , Cell Self Renewal , Gene Expression Profiling , Histocompatibility Antigens Class II/metabolism , Membrane Proteins/metabolism , Mice , Mice, Transgenic , Parabiosis , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Single-Cell Analysis , Ventricular Remodeling , Vesicular Transport Proteins/metabolism
2.
Nat Immunol ; 20(5): 664, 2019 May.
Article in English | MEDLINE | ID: mdl-30862954

ABSTRACT

In the version of this article initially published, the equal contribution of the third author was omitted. The footnote links for that author should be "Sara Nejat1,11" and the correct statement is as follows: "11These authors contributed equally: Sarah A. Dick, Jillian A. Macklin, Sara Nejat." The error has been corrected in the HTML and PDF versions of the article.

3.
Immunity ; 54(9): 2057-2071.e6, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34363749

ABSTRACT

Hypertension affects one-third of the world's population, leading to cardiac dysfunction that is modulated by resident and recruited immune cells. Cardiomyocyte growth and increased cardiac mass are essential to withstand hypertensive stress; however, whether immune cells are involved in this compensatory cardioprotective process is unclear. In normotensive animals, single-cell transcriptomics of fate-mapped self-renewing cardiac resident macrophages (RMs) revealed transcriptionally diverse cell states with a core repertoire of reparative gene programs, including high expression of insulin-like growth factor-1 (Igf1). Hypertension drove selective in situ proliferation and transcriptional activation of some cardiac RM states, directly correlating with increased cardiomyocyte growth. During hypertension, inducible ablation of RMs or selective deletion of RM-derived Igf1 prevented adaptive cardiomyocyte growth, and cardiac mass failed to increase, which led to cardiac dysfunction. Single-cell transcriptomics identified a conserved IGF1-expressing macrophage subpopulation in human cardiomyopathy. Here we defined the absolute requirement of RM-produced IGF-1 in cardiac adaptation to hypertension.


Subject(s)
Adaptation, Physiological/physiology , Hypertension/metabolism , Insulin-Like Growth Factor I/metabolism , Macrophages/metabolism , Ventricular Remodeling/physiology , Animals , Heart Failure/etiology , Heart Failure/metabolism , Heart Failure/pathology , Humans , Hypertension/complications , Hypertension/immunology , Infant , Male , Mice , Middle Aged , Myocardium/immunology , Myocardium/metabolism , Myocardium/pathology
4.
Immunity ; 47(5): 974-989.e8, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29166591

ABSTRACT

Innate and adaptive immune cells modulate heart failure pathogenesis during viral myocarditis, yet their identities and functions remain poorly defined. We utilized a combination of genetic fate mapping, parabiotic, transcriptional, and functional analyses and demonstrated that the heart contained two major conventional dendritic cell (cDC) subsets, CD103+ and CD11b+, which differentially relied on local proliferation and precursor recruitment to maintain their tissue residency. Following viral infection of the myocardium, cDCs accumulated in the heart coincident with monocyte infiltration and loss of resident reparative embryonic-derived cardiac macrophages. cDC depletion abrogated antigen-specific CD8+ T cell proliferative expansion, transforming subclinical cardiac injury to overt heart failure. These effects were mediated by CD103+ cDCs, which are dependent on the transcription factor BATF3 for their development. Collectively, our findings identified resident cardiac cDC subsets, defined their origins, and revealed an essential role for CD103+ cDCs in antigen-specific T cell responses during subclinical viral myocarditis.


Subject(s)
Antigens, CD/analysis , Cardiovirus Infections/complications , Dendritic Cells/immunology , Encephalomyocarditis virus , Heart Failure/prevention & control , Integrin alpha Chains/analysis , Myocarditis/complications , Animals , CD11b Antigen/analysis , CD8-Positive T-Lymphocytes/immunology , Cardiovirus Infections/immunology , Cell Movement , Female , Hematopoiesis , Immunologic Memory , Male , Mice , Mice, Inbred C57BL , Myocarditis/immunology , Receptors, CCR2/physiology
5.
Proc Natl Acad Sci U S A ; 118(1)2021 01 05.
Article in English | MEDLINE | ID: mdl-33372158

ABSTRACT

Macrophages are the principal immune cells of the epididymis and testis, but their origins, heterogeneity, development, and maintenance are not well understood. Here, we describe distinct populations of epididymal and testicular macrophages that display an organ-specific cellular identity. Combining in vivo fate-mapping, chimeric and parabiotic mouse models with in-depth cellular analyses, we found that CD64hiMHCIIlo and CD64loMHCIIhi macrophage populations of epididymis and testis arise sequentially from yolk sac erythro-myeloid progenitors, embryonic hematopoiesis, and nascent neonatal monocytes. While monocytes were the major developmental source of both epididymal and testicular macrophages, both populations self-maintain in the steady-state independent of bone marrow hematopoietic precursors. However, after radiation-induced macrophage ablation or during infection, bone marrow-derived circulating monocytes are recruited to the epididymis and testis, giving rise to inflammatory macrophages that promote tissue damage. These results define the layered ontogeny, maintenance and inflammatory response of macrophage populations in the male reproductive organs.


Subject(s)
Infertility, Male/immunology , Macrophages/immunology , Macrophages/metabolism , Animals , Cell Differentiation , Cell Lineage , Epididymis/immunology , Epididymis/metabolism , Infertility, Male/metabolism , Infertility, Male/physiopathology , Male , Mice , Mice, Inbred C57BL , Monocytes/immunology , Testis/immunology , Testis/metabolism
6.
Am J Physiol Lung Cell Mol Physiol ; 315(5): L742-L751, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30091380

ABSTRACT

Deficient nitric oxide (NO) signaling plays a critical role in the pathogenesis of chronic neonatal pulmonary hypertension (PHT). Physiological NO signaling is regulated by S-nitrosothiols (SNOs), which act both as a reservoir for NO and as a reversible modulator of protein function. We have previously reported that therapy with inhaled NO (iNO) increased peroxynitrite-mediated nitration in the juvenile rat lung, although having minimal reversing effects on vascular remodeling. We hypothesized that sodium nitrite (NaNO2) would be superior to iNO in enhancing lung SNOs, thereby contributing to reversal of chronic hypoxic PHT. Rat pups were exposed to air or hypoxia (13% O2) from postnatal days 1 to 21. Dose-response prevention studies were conducted from days 1-21 to determine the optimal dose of NaNO2. Animals then received rescue therapy with daily subcutaneous NaNO2 (20 mg/kg), vehicle, or were continuously exposed to iNO (20 ppm) from days 14-21. Chronic PHT secondary to hypoxia was both prevented and reversed by treatment with NaNO2. Rescue NaNO2 increased lung NO and SNO contents to a greater extent than iNO, without causing nitration. Seven lung SNO proteins upregulated by treatment with NaNO2 were identified by multiplex tandem mass tag spectrometry, one of which was leukotriene A4 hydrolase (LTA4H). Rescue therapy with a LTA4H inhibitor, SC57461A (10 mg·kg-1·day-1 sc), partially reversed chronic hypoxic PHT. We conclude that NaNO2 was superior to iNO in increasing tissue NO and SNO generation and reversing chronic PHT, in part via upregulated SNO-LTA4H.


Subject(s)
Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/complications , Indicators and Reagents/administration & dosage , Sodium Nitrite/administration & dosage , Vascular Remodeling/drug effects , Administration, Inhalation , Animals , Animals, Newborn , Chronic Disease , Female , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/pathology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/pathology , Male , Nitric Oxide/metabolism , Peroxynitrous Acid/metabolism , Rats , Rats, Sprague-Dawley , Signal Transduction
7.
Am J Physiol Lung Cell Mol Physiol ; 312(2): L208-L216, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27913427

ABSTRACT

Bronchopulmonary dysplasia (BPD) is a chronic lung injury characterized by impaired alveologenesis that may persist into adulthood. Rat models of BPD using varying degrees of hyperoxia to produce injury either cause early mortality or spontaneously recover following removal of the inciting stimulus, thus limiting clinical relevance. We sought to refine an established rat model induced by exposure to 60% O2 from birth by following hyperoxia with intermittent hypoxia (IH). Rats exposed from birth to air or 60% O2 until day 14 were recovered in air with or without IH (FIO2 = 0.10 for 10 min every 6 h) until day 28 Animals exposed to 60% O2 and recovered in air had no evidence of abnormal lung morphology on day 28 or at 10-12 wk. In contrast, 60% O2-exposed animals recovered in IH had persistently increased mean chord length, more dysmorphic septal crests, and fewer peripheral arteries. Recovery in IH also increased pulmonary vascular resistance, Fulton index, and arterial wall thickness. IH-mediated abnormalities in lung structure (but not pulmonary hypertension) persisted when reexamined at 10-12 wk, accompanied by increased pulmonary vascular reactivity and decreased exercise tolerance. Increased mean chord length secondary to IH was prevented by treatment with a peroxynitrite decomposition catalyst [5,10,15,20-Tetrakis(4-sulfonatophenyl)-21H,23H-porphyrin iron (III) chloride, 30 mg/kg/day, days 14-28], an effect accompanied by fewer inflammatory cells. We conclude that IH during recovery from hyperoxia-induced injury prevents recovery of alveologenesis and leads to changes in lung and pulmonary vascular function lasting into adulthood, thus more closely mimicking contemporary BPD.


Subject(s)
Bronchopulmonary Dysplasia/complications , Bronchopulmonary Dysplasia/pathology , Hyperoxia/complications , Hypoxia/complications , Lung Injury/complications , Pulmonary Alveoli/growth & development , Pulmonary Alveoli/pathology , Animals , Animals, Newborn , Biomarkers/metabolism , Catalysis , Disease Models, Animal , Female , Hyperoxia/pathology , Hypertension, Pulmonary/complications , Hypoxia/pathology , Lung Injury/pathology , Male , Metalloporphyrins/pharmacology , Peroxynitrous Acid/metabolism , Physical Conditioning, Animal , Pneumonia/complications , Rats, Sprague-Dawley
8.
Mol Cell Biochem ; 435(1-2): 163-173, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28526936

ABSTRACT

Emerging evidence suggests that long non-coding RNAs (lncRNAs) represent a cellular hub coordinating various cellular processes that are critical in health and disease. Mechanical stress triggers changes in vascular smooth muscle cells (VSMCs) that in turn contribute to pathophysiological changes within the vasculature. We sought to evaluate the role that lncRNAs play in mechanical stretch-induced alterations of human aortic smooth muscle cells (HASMCs). RNA (lncRNA and mRNA) samples isolated from HASMCs that had been subjected to 10 or 20% elongation (1 Hz) for 24 h were profiled with the Arraystar Human LncRNA Microarray V3.0. LncRNA expression was quantified in parallel via qRT-PCR. Of the 30,586 human lncRNAs screened, 580 were differentially expressed (DE, P < 0.05) in stretched HASMCs. Amongst the 26,109 protein-coding transcripts evaluated, 25 of those DE were associated with 25 of the aforementioned DE lncRNAs (P < 0.05). Subsequent Kyoto Encyclopedia of Genes and Genomes analysis revealed that the DE mRNAs were largely associated with the tumor necrosis factor signaling pathway and inflammation. Gene Ontology analysis indicated that the DE mRNAs were associated with cell differentiation, stress response, and response to external stimuli. We describe the first transcriptome profile of stretch-induced changes in HASMCs and provide novel insights into the regulatory switches that may be fundamental in governing aberrant VSMC remodeling.


Subject(s)
Aorta/metabolism , Gene Expression Profiling , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Oligonucleotide Array Sequence Analysis , RNA, Long Noncoding/biosynthesis , Stress, Mechanical , Aorta/cytology , Humans , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology
9.
Am J Physiol Lung Cell Mol Physiol ; 311(5): L985-L999, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27694473

ABSTRACT

Chronic neonatal pulmonary hypertension (PHT) frequently results in early death. Systemically administered Rho-kinase (ROCK) inhibitors prevent and reverse chronic PHT in neonatal rats, but at the cost of severe adverse effects, including systemic hypotension and growth restriction. Simvastatin has pleiotropic inhibitory effects on isoprenoid intermediates that may limit activity of RhoA, which signals upstream of ROCK. We therefore hypothesized that statin treatment would safely limit pulmonary vascular RhoA activity and prevent and reverse experimental chronic neonatal PHT via downstream inhibitory effects on pathological ROCK activity. Sprague-Dawley rats in normoxia (room air) or moderate normobaric hypoxia (13% O2) received simvastatin (2 mg·kg-1·day-1 ip) or vehicle from postnatal days 1-14 (prevention protocol) or from days 14-21 (rescue protocol). Chronic hypoxia increased RhoA and ROCK activity in lung tissue. Simvastatin reduced lung content of the isoprenoid intermediate farnesyl pyrophosphate and decreased RhoA/ROCK signaling in the hypoxia-exposed lung. Preventive or rescue treatment of chronic hypoxia-exposed animals with simvastatin decreased pulmonary vascular resistance, right ventricular hypertrophy, and pulmonary arterial remodeling. Preventive simvastatin treatment improved weight gain, did not lower systemic blood pressure, and did not cause apparent toxic effects on skeletal muscle, liver or brain. Rescue therapy with simvastatin improved exercise capacity. We conclude that simvastatin limits RhoA/ROCK activity in the chronic hypoxia-exposed lung, thus preventing or ameliorating hemodynamic and structural markers of chronic PHT and improving long-term outcome, without causing adverse effects.


Subject(s)
Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/prevention & control , Signal Transduction/drug effects , Simvastatin/therapeutic use , rhoA GTP-Binding Protein/metabolism , Animals , Animals, Newborn , Biosynthetic Pathways/drug effects , Blood Pressure/drug effects , Cholesterol/blood , Chronic Disease , Female , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/complications , Hypoxia/blood , Hypoxia/complications , Hypoxia/drug therapy , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lung/drug effects , Lung/enzymology , Lung/pathology , Male , Myelin Sheath/metabolism , Nitric Oxide Synthase Type III/metabolism , Organ Size/drug effects , Physical Conditioning, Animal , Polyisoprenyl Phosphates/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/pathology , Pulmonary Artery/physiopathology , Rats, Sprague-Dawley , Sesquiterpenes/metabolism , Simvastatin/pharmacology , Vascular Remodeling/drug effects , rho-Associated Kinases/metabolism
10.
Am J Physiol Lung Cell Mol Physiol ; 311(2): L292-302, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27317685

ABSTRACT

Systemically-administered bleomycin causes inflammation, arrested lung growth, and pulmonary hypertension (PHT) in the neonatal rat, similar to human infants with severe bronchopulmonary dysplasia (BPD). Leukotrienes (LTs) are inflammatory lipid mediators produced by multiple cell types in the lung. The major LTs, LTB4 and cysteinyl LTs, are suggested to contribute to BPD, but their specific roles remain largely unexplored in experimental models. We hypothesized that LTs are increased in bleomycin-induced BPD-like injury, and that inhibition of LT production would prevent inflammatory cell influx and thereby ameliorate lung injury. Rat pups were exposed to bleomycin (1 mg·kg(-1)·day(-1) ip) or vehicle (control) from postnatal days 1-14 and were treated with either zileuton (5-lipoxygenase inhibitor), montelukast (cysteinyl LT1 receptor antagonist), or SC57461A (LTA4 hydrolase inhibitor) 10 mg·kg(-1)·day(-1) ip. Bleomycin led to increased lung content of LTB4, but not cysteinyl LTs. Bleomycin-induced increases in tissue neutrophils and macrophages and lung contents of LTB4 and tumor necrosis factor-α were all prevented by treatment with zileuton. Treatment with zileuton or SC57461A also prevented the hemodynamic and structural markers of chronic PHT, including raised pulmonary vascular resistance, increased Fulton index, and arterial wall remodeling. However, neither treatment prevented impaired alveolarization or vascular hypoplasia secondary to bleomycin. Treatment with montelukast had no effect on macrophage influx, PHT, or on abnormal lung structure. We conclude that LTB4 plays a crucial role in lung inflammation and PHT in experimental BPD. Agents targeting LTB4 or LTB4-mediated signaling may have utility in infants at risk of developing BPD-associated PHT.


Subject(s)
Bronchopulmonary Dysplasia/immunology , Hypertension, Pulmonary/immunology , Leukotriene B4/physiology , Macrophages/immunology , Animals , Animals, Newborn , Bleomycin , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/metabolism , Cell Movement/immunology , Female , Gene Expression , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Rats, Sprague-Dawley
11.
Am J Respir Cell Mol Biol ; 52(6): 717-27, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25337652

ABSTRACT

Chronic neonatal pulmonary hypertension frequently culminates in right ventricular (RV) failure and death. In juvenile rats, RV systolic dysfunction secondary to chronic hypoxia is rescued by systemic treatment with a Rho kinase (ROCK) inhibitor. To explore the relationship between ROCK inhibitor-mediated decreases in pulmonary vascular resistance and pressure, RV hypertrophy, and systolic dysfunction, we compared the effects of systemically administered to inhaled (pulmonary-selective) ROCK inhibitor on RV systolic function. Rat pups were exposed to air or hypoxia (13% O2) from Postnatal Days 1 to 21 and received rescue treatment with aerosolized fasudil (200 mM) for 15 minutes three times daily or intraperitoneal Y27632 (15 mg/kg twice daily) from Days 14 to 21. Chronic hypoxia differentially increased RhoA and ROCK activity in the right, but not left, cardiac ventricle. Inhaled ROCK inhibitor normalized pulmonary vascular resistance and caused regression of RV hypertrophy and pulmonary arterial wall remodeling but did not improve RV systolic dysfunction (decreased stroke volume and tricuspid annular plane systolic excursion). Systemic, but not inhaled, ROCK inhibitor normalized up-regulated ROCK and phosphodiesterase 5 activities in the right ventricle. Treatment with sildenafil (100 mg/kg/d intraperitoneally from Days 14 to 21) improved RV systolic function. Collectively, these data indicate that pressure unloading and regressed arterial and cardiac remodeling did not lead to recovery of systolic function while right ventricular ROCK activity remained increased. Right ventricle-specific up-regulation of RhoA/ROCK activity is critical to hypoxia-mediated systolic dysfunction, in part by regulating the activity of phosphodiesterase 5.


Subject(s)
Hypertension, Pulmonary/enzymology , Hypertrophy, Right Ventricular/enzymology , Ventricular Dysfunction, Right/enzymology , rho-Associated Kinases/physiology , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/administration & dosage , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , Animals , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Heart Ventricles/enzymology , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/drug therapy , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/etiology , Piperazines/pharmacology , Purines/pharmacology , Rats , Sildenafil Citrate , Sulfonamides/pharmacology , Vascular Resistance/drug effects , Ventricular Dysfunction, Right/drug therapy , Ventricular Dysfunction, Right/etiology , rho-Associated Kinases/antagonists & inhibitors , rhoA GTP-Binding Protein/metabolism
12.
Am J Physiol Lung Cell Mol Physiol ; 309(1): L53-62, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-26136527

ABSTRACT

Neutrophil (PMNL) influx precedes lung macrophage (LM) influx into the lung following exposure of newborn pups to 60% O2. We hypothesized that PMNL were responsible for the signals leading to LM influx. This was confirmed when inhibition of PMNL influx with a CXC chemokine receptor-2 antagonist, SB-265610, also prevented the 60% O2-dependent LM influx, LM-derived nitrotyrosine formation, and pruning of small arterioles. Exposure to 60% O2 was associated with increased lung contents of neutrophil elastase and α-elastin, a marker of denatured elastin, and a decrease in elastin fiber density. This led us to speculate that neutrophil elastase-induced elastin fragments were the chemokines that led to a LM influx into the 60% O2-exposed lung. Inhibition of neutrophil elastase with sivelestat or elafin attenuated the LM influx. Sivelestat also attenuated the 60% O2-induced decrease in elastin fiber density. Daily injections of pups with an antibody to α-elastin prevented the 60% O2-dependent LM influx, impaired alveologenesis, and impaired small vessel formation. This suggests that neutrophil elastase inhibitors may protect against neonatal lung injury not only by preventing structural elastin degradation, but also by blocking elastin fragment-induced LM influx, thus preventing tissue injury from LM-derived peroxynitrite formation.


Subject(s)
Elastin/metabolism , Leukocyte Elastase/metabolism , Macrophages/immunology , Neutrophils/immunology , Oxygen/toxicity , Animals , Animals, Newborn , Cell Movement/immunology , Elafin/pharmacology , Elastin/immunology , Female , Glycine/analogs & derivatives , Glycine/pharmacology , Leukocyte Elastase/antagonists & inhibitors , Lung/pathology , Lung Injury/immunology , Maternal Exposure , Oxygen/pharmacology , Peroxynitrous Acid/biosynthesis , Phenylurea Compounds/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-8B/antagonists & inhibitors , Sulfonamides/pharmacology , Triazoles/pharmacology , Vascular Remodeling
13.
Am J Physiol Lung Cell Mol Physiol ; 308(6): L503-10, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25595650

ABSTRACT

Arginase is an enzyme that limits substrate L-arginine bioavailability for the production of nitric oxide by the nitric oxide synthases and produces L-ornithine, which is a precursor for collagen formation and tissue remodeling. We studied the pulmonary vascular effects of arginase inhibition in an established model of repeated systemic bleomycin sulfate administration in neonatal rats that results in pulmonary hypertension and lung injury mimicking the characteristics typical of bronchopulmonary dysplasia. We report that arginase expression is increased in the lungs of bleomycin-exposed neonatal rats and that treatment with the arginase inhibitor amino-2-borono-6-hexanoic acid prevented the bleomycin-induced development of pulmonary hypertension and deposition of collagen. Arginase inhibition resulted in increased L-arginine and L-arginine bioavailability and increased pulmonary nitric oxide production. Arginase inhibition also normalized the expression of inducible nitric oxide synthase, and reduced bleomycin-induced nitrative stress while having no effect on bleomycin-induced inflammation. Our data suggest that arginase is a promising target for therapeutic interventions in neonates aimed at preventing lung vascular remodeling and pulmonary hypertension.


Subject(s)
Aminocaproates/pharmacology , Antibiotics, Antineoplastic/adverse effects , Arginase/antagonists & inhibitors , Bleomycin/adverse effects , Boron Compounds/pharmacology , Collagen/metabolism , Hypertension, Pulmonary , Lung/enzymology , Vascular Remodeling/drug effects , Animals , Antibiotics, Antineoplastic/pharmacology , Arginase/metabolism , Arginine/metabolism , Bleomycin/pharmacology , Bronchopulmonary Dysplasia/chemically induced , Bronchopulmonary Dysplasia/enzymology , Bronchopulmonary Dysplasia/pathology , Bronchopulmonary Dysplasia/prevention & control , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Lung/pathology , Lung Injury/chemically induced , Lung Injury/enzymology , Lung Injury/pathology , Lung Injury/prevention & control , Nitric Oxide/metabolism , Rats , Rats, Sprague-Dawley
14.
Am J Respir Cell Mol Biol ; 50(1): 61-73, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23947621

ABSTRACT

Bleomycin-induced lung injury is characterized in the neonatal rat by inflammation dominated by neutrophils and macrophages, inhibited distal airway and vascular development, and pulmonary hypertension, similar to human infants with severe bronchopulmonary dysplasia. Rho-kinase (ROCK) is known to mediate lung injury in adult animals via stimulatory effects on inflammation. We therefore hypothesized that inhibition of ROCK may ameliorate bleomycin-induced lung injury in the neonatal rat. Pups received daily intraperitoneal bleomycin or saline from Postnatal Days 1 through 14 with or without Y-27632, a ROCK inhibitor. Treatment with Y-27632 prevented bleomycin-induced pulmonary hypertension, as evidenced by normalized pulmonary vascular resistance, decreased right-ventricular hypertrophy, and attenuated remodeling of pulmonary resistance arteries. Bleomycin-induced changes in distal lung architecture, including septal thinning, inhibited alveolarization, and decreased numbers of peripheral arteries and capillaries, were partially or completely normalized by Y-27632. Treatment with Y-27632 or a CXCR2 antagonist, SB265610, also abrogated tissue neutrophil influx, while having no effect on macrophages. However, treatment with SB265610 did not prevent bleomycin-induced lung injury. Lung content of angiostatic thrombospondin-1 (TSP1) was increased significantly in the lungs of bleomycin-exposed animals, and was completely attenuated by treatment with Y-27632. Thrombin-stimulated TSP1 production by primary cultured rat pulmonary artery endothelial cells was also attenuated by Y-27632. Taken together, our findings suggest a preventive effect of Y-27632 on bleomycin-mediated injury by a mechanism unrelated to inflammatory cells. Our data suggest that improvements in lung morphology may have been related to indirect stimulatory effects on angiogenesis via down-regulation of TSP1.


Subject(s)
Enzyme Inhibitors/pharmacology , Lung Injury/prevention & control , Pneumonia/diagnostic imaging , Pneumonia/pathology , rho-Associated Kinases/antagonists & inhibitors , Amides/pharmacology , Animals , Animals, Newborn , Bleomycin/adverse effects , Chemokines/metabolism , Down-Regulation/drug effects , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Hypertrophy, Right Ventricular/drug therapy , Hypertrophy, Right Ventricular/metabolism , Lung/drug effects , Lung/metabolism , Lung/pathology , Lung Injury/chemically induced , Lung Injury/metabolism , Macrophages/diagnostic imaging , Macrophages/drug effects , Macrophages/metabolism , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neutrophils/drug effects , Neutrophils/metabolism , Neutrophils/pathology , Pneumonia/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Pulmonary Artery/pathology , Pyridines/pharmacology , Radiography , Rats , Rats, Sprague-Dawley , Thrombospondin 1/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vascular Resistance/drug effects , rho-Associated Kinases/metabolism
15.
Am J Physiol Lung Cell Mol Physiol ; 303(1): L75-87, 2012 Jul 01.
Article in English | MEDLINE | ID: mdl-22582114

ABSTRACT

Bleomycin-induced lung injury is characterized in the neonatal rat by inflammation, arrested lung growth, and pulmonary hypertension (PHT), as observed in human infants with severe bronchopulmonary dysplasia. Inhalation of CO(2) (therapeutic hypercapnia) has been described to limit cytokine production and to have anti-inflammatory effects on the injured lung; we therefore hypothesized that therapeutic hypercapnia would prevent bleomycin-induced lung injury. Spontaneously breathing rat pups were treated with bleomycin (1 mg/kg/d ip) or saline vehicle from postnatal days 1-14 while being continuously exposed to 5% CO(2) (Pa(CO(2)) elevated by 15-20 mmHg), 7% CO(2) (Pa(CO(2)) elevated by 35 mmHg), or normocapnia. Bleomycin-treated animals exposed to 7%, but not 5%, CO(2), had significantly attenuated lung tissue macrophage influx and PHT, as evidenced by normalized pulmonary vascular resistance and right ventricular systolic function, decreased right ventricular hypertrophy, and attenuated remodeling of pulmonary resistance arteries. The level of CO(2) neither prevented increased tissue neutrophil influx nor led to improvements in decreased lung weight, septal thinning, impaired alveolarization, or decreased numbers of peripheral arteries. Bleomycin led to increased expression and content of lung tumor necrosis factor (TNF)-α, which was found to colocalize with tissue macrophages and to be attenuated by exposure to 7% CO(2). Inhibition of TNF-α signaling with the soluble TNF-2 receptor etanercept (0.4 mg/kg ip from days 1-14 on alternate days) prevented bleomycin-induced PHT without decreasing tissue macrophages and, similar to CO(2), had no effect on arrested alveolar development. Our findings are consistent with a preventive effect of therapeutic hypercapnia with 7% CO(2) on bleomycin-induced PHT via attenuation of macrophage-derived TNF-α. Neither tissue macrophages nor TNF-α appeared to contribute to arrested lung development induced by bleomycin. That 7% CO(2) normalized pulmonary vascular resistance and right ventricular function without improving inhibited airway and vascular development suggests that vascular hypoplasia does not contribute significantly to functional changes of PHT in this model.


Subject(s)
Hypercapnia/physiopathology , Hypertension, Pulmonary/prevention & control , Macrophages/metabolism , Pulmonary Alveoli/physiopathology , Pulmonary Artery/physiopathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/metabolism , Animals , Animals, Newborn/metabolism , Animals, Newborn/physiology , Bleomycin/toxicity , Carbon Dioxide/administration & dosage , Carbon Dioxide/blood , Hypercapnia/blood , Hypercapnia/chemically induced , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Hypertrophy, Right Ventricular/metabolism , Hypertrophy, Right Ventricular/physiopathology , Inflammation/metabolism , Inflammation/physiopathology , Lung/metabolism , Lung/physiopathology , Lung Injury/metabolism , Lung Injury/physiopathology , Lung Injury/prevention & control , Neutrophils/metabolism , Pulmonary Alveoli/metabolism , Pulmonary Artery/drug effects , Pulmonary Artery/metabolism , Rats , Ventricular Function, Right/drug effects
16.
Am J Physiol Heart Circ Physiol ; 302(12): H2599-611, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22505643

ABSTRACT

Sustained therapeutic hypercapnia prevents pulmonary hypertension in experimental animals, but its rescue effects on established disease have not been studied. Therapies that inhibit Rho-kinase (ROCK) and/or augment nitric oxide (NO)-cyclic guanosine monophosphate (cGMP) signaling can reverse or prevent progression of chronic pulmonary hypertension. Our objective in the present study was to determine whether sustained rescue treatment with inhaled CO(2) (therapeutic hypercapnia) would improve structural and functional changes of chronic hypoxic pulmonary hypertension. Spontaneously breathing pups were exposed to normoxia (21% O(2)) or hypoxia (13% O(2)) from postnatal days 1-21 with or without 7% CO(2) (Pa(CO(2)) elevated by ∼25 mmHg) or 10% CO(2) (Pa(CO(2)) elevated by ∼40 mmHg) from days 14 to 21. Compared with hypoxia alone, animals exposed to hypoxia and 10% CO(2) had significantly (P < 0.05) decreased pulmonary vascular resistance, right-ventricular systolic pressure, right-ventricular hypertrophy, and medial wall thickness of pulmonary resistance arteries as well as decreased lung phosphodiesterase (PDE) V, RhoA, and ROCK activity. Rescue treatment with 10% CO(2), or treatment with a ROCK inhibitor (15 mg/kg ip Y-27632 twice daily from days 14 to 21), also increased pulmonary arterial endothelial nitric oxide synthase and lung NO content. In contrast, cGMP content and cGMP-dependent protein kinase (PKG) activity were increased by exposure to 10% CO(2), but not by ROCK inhibition with Y-27632. In vitro exposure of pulmonary artery smooth muscle cells to hypercapnia suppressed serum-induced ROCK activity, which was prevented by inhibition of PKG with Rp-8-Br-PET-cGMPS. We conclude that sustained hypercapnia dose-dependently inhibited ROCK activity, augmented NO-cGMP-PKG signaling, and led to partial improvements in the hemodynamic and structural abnormalities of chronic hypoxic PHT in juvenile rats. Increased PKG content and activity appears to play a major upstream role in CO(2)-induced suppression of ROCK activity in pulmonary arterial smooth muscle.


Subject(s)
Carbon Dioxide/therapeutic use , Hypertension, Pulmonary/drug therapy , Hypoxia/drug therapy , Pulmonary Artery/drug effects , rho-Associated Kinases/metabolism , Animals , Carbon Dioxide/administration & dosage , Cyclic GMP-Dependent Protein Kinases/metabolism , Cyclic Nucleotide Phosphodiesterases, Type 5/metabolism , Hypertension, Pulmonary/enzymology , Hypoxia/enzymology , Lung/blood supply , Lung/drug effects , Lung/enzymology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Pulmonary Artery/enzymology , Rats , Signal Transduction/drug effects , Vasodilation/drug effects , rhoA GTP-Binding Protein/metabolism
17.
Elife ; 112022 12 14.
Article in English | MEDLINE | ID: mdl-36515584

ABSTRACT

The epididymis functions as transition zone for post-testicular sperm maturation and storage and faces contrasting immunological challenges, i.e. tolerance towards spermatozoa vs. reactivity against pathogens. Thus, normal organ function and integrity relies heavily on a tightly controlled immune balance. Previous studies described inflammation-associated tissue damage solely in the distal regions (corpus, cauda), but not in the proximal regions (initial segment, caput). To understand the observed region-specific immunity along the epididymal duct, we have used an acute bacterial epididymitis mouse model and analyzed the disease progression. Whole transcriptome analysis using RNAseq 10 days post infection showed a pro-inflammatory environment within the cauda, while the caput exhibited only minor transcriptional changes. High-dimensional flow cytometry analyses revealed drastic changes in the immune cell composition upon infection with uropathogenic Escherichia coli. A massive influx of neutrophils and monocytes was observed exclusively in distal regions and was associated with bacterial appearance and tissue alterations. In order to clarify the reasons for the region-specific differences in the intensity of immune responses, we investigated the heterogeneity of resident immune cell populations under physiological conditions by scRNASeq analysis of extravascular CD45+ cells. Twelve distinct immune cell subsets were identified, displaying substantial differences in distribution along the epididymis as further assessed by flow cytometry and immunofluorescence staining. Macrophages constituted the majority of resident immune cells and were further separated in distinct subgroups based on their transcriptional profile, tissue location and monocyte-dependence. Crucially, the proximal and distal regions showed striking differences in their immunological landscapes. These findings indicate that resident immune cells are strategically positioned along the epididymal duct, potentially providing different immunological environments required for addressing the contrasting immunological challenges and thus, preserving tissue integrity and organ function.


Subject(s)
Epididymis , Semen , Mice , Male , Animals , Sperm Maturation , Spermatozoa , Testis
18.
Sci Immunol ; 7(67): eabf7777, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34995099

ABSTRACT

Resident macrophages orchestrate homeostatic, inflammatory, and reparative activities. It is appreciated that different tissues instruct specialized macrophage functions. However, individual tissues contain heterogeneous subpopulations, and how these subpopulations are related is unclear. We asked whether common transcriptional and functional elements could reveal an underlying framework across tissues. Using single-cell RNA sequencing and random forest modeling, we observed that four genes could predict three macrophage subsets that were present in murine heart, liver, lung, kidney, and brain. Parabiotic and genetic fate mapping studies revealed that these core markers predicted three unique life cycles across 17 tissues. TLF+ (expressing TIMD4 and/or LYVE1 and/or FOLR2) macrophages were maintained through self-renewal with minimal monocyte input; CCR2+ (TIMD4−LYVE1−FOLR2−) macrophages were almost entirely replaced by monocytes, and MHC-IIhi macrophages (TIMD4−LYVE1−FOLR2−CCR2−), while receiving modest monocyte contribution, were not continually replaced. Rather, monocyte-derived macrophages contributed to the resident macrophage population until they reached a defined upper limit after which they did not outcompete pre-existing resident macrophages. Developmentally, TLF+ macrophages were first to emerge in the yolk sac and early fetal organs. Fate mapping studies in the mouse and human single-cell RNA sequencing indicated that TLF+ macrophages originated from both yolk sac and fetal monocyte precursors. Furthermore, TLF+ macrophages were the most transcriptionally conserved subset across mouse tissues and between mice and humans, despite organ- and species-specific transcriptional differences. Here, we define the existence of three murine macrophage subpopulations based on common life cycle properties and core gene signatures and provide a common starting point to understand tissue macrophage heterogeneity.


Subject(s)
Folate Receptor 2/immunology , Histocompatibility Antigens Class II/immunology , Macrophages/immunology , Membrane Proteins/immunology , Receptors, CCR2/immunology , Vesicular Transport Proteins/immunology , Animals , Life Cycle Stages/immunology , Macrophage Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Receptors, CCR2/deficiency
19.
Am J Physiol Lung Cell Mol Physiol ; 300(3): L319-29, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21148793

ABSTRACT

During early postnatal alveolar formation, the lung tissue of rat pups undergoes a physiological remodeling involving apoptosis of distal lung cells. Exposure of neonatal rats to severe hyperoxia (≥95% O(2)) both arrests lung growth and results in increased lung cell apoptosis. In contrast, exposure to moderate hyperoxia (60% O(2)) for 14 days does not completely arrest lung cell proliferation and is associated with parenchymal thickening. On the basis of similarities in lung architecture observed following either exposure to 60% O(2), or pharmacological inhibition of physiological apoptosis, we hypothesized that exposure to 60% O(2) would result in an inhibition of physiological lung cell apoptosis. Consistent with this hypothesis, we observed that the parenchymal thickening induced by exposure to 60% O(2) was associated with decreased numbers of apoptotic cells, increased expressions of the antiapoptotic regulator Bcl-xL, and the putative antiapoptotic protein survivin, and decreased expressions of the proapoptotic cleaved caspases-3 and -7. In summary, exposure of the neonatal rat lung to moderate hyperoxia results in an inhibition of physiological apoptosis, which contributes to the parenchymal thickening observed in the resultant lung injury.


Subject(s)
Apoptosis/drug effects , Lung Injury/chemically induced , Lung Injury/pathology , Oxygen/pharmacology , Signal Transduction/drug effects , Air , Animals , Animals, Newborn , Blotting, Western , Caspase 3/metabolism , Cell Count , Cell Death/drug effects , Female , Immunohistochemistry , Lung/drug effects , Lung/enzymology , Lung/pathology , Rats , Rats, Sprague-Dawley , Staurosporine/pharmacology
20.
Am J Physiol Heart Circ Physiol ; 299(6): H1854-64, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20889845

ABSTRACT

Chronic pulmonary hypertension in infancy and childhood is characterized by a fixed and progressive increase in pulmonary arterial pressure and resistance, pulmonary arterial remodeling, and right ventricular hypertrophy and systolic dysfunction. These abnormalities are replicated in neonatal rats chronically exposed to hypoxia from birth in which increased activity of Rho-kinase (ROCK) is critical to injury, as evidenced by preventive effects of ROCK inhibitors. Our objective in the present study was to examine the reversing effects of a late or rescue approach to treatment with a ROCK inhibitor on the pulmonary and cardiac manifestations of established chronic hypoxic pulmonary hypertension. Rat pups were exposed to air or hypoxia (13% O(2)) from postnatal day 1 and were treated with Y-27632 (15 mg/kg) or saline vehicle by twice daily subcutaneous injection commencing on day 14, for up to 7 days. Treatment with Y-27632 significantly attenuated right ventricular hypertrophy, reversed arterial wall remodeling, and completely normalized right ventricular systolic function in hypoxia-exposed animals. Reversal of arterial wall remodeling was accompanied by increased apoptosis and attenuated content of endothelin (ET)-1 and ET(A) receptors. Treatment of primary cultured juvenile rat pulmonary artery smooth muscle cells with Y-27632 attenuated serum-stimulated ROCK activity and proliferation and increased apoptosis. Smooth muscle apoptosis was also induced by short interfering RNA-mediated knockdown of ROCK-II, but not of ROCK-I. We conclude that sustained rescue treatment with a ROCK inhibitor reversed both the hemodynamic and structural abnormalities of chronic hypoxic pulmonary hypertension in juvenile rats and normalized right ventricular systolic function. Attenuated expression and activity of ET-1 and its A-type receptor on pulmonary arterial smooth muscle was a likely contributor to the stimulatory effects of ROCK inhibition on apoptosis. In addition, our data suggest that ROCK-II may be dominant in enhancing survival of pulmonary arterial smooth muscle.


Subject(s)
Amides/pharmacology , Hypertension, Pulmonary/drug therapy , Hypoxia/complications , Muscle, Smooth, Vascular/drug effects , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Ventricular Dysfunction, Right/drug therapy , Ventricular Function, Right , Ventricular Remodeling/drug effects , rho-Associated Kinases/antagonists & inhibitors , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Age Factors , Aging , Amides/administration & dosage , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chronic Disease , Disease Models, Animal , Endothelin-1/metabolism , Hemodynamics , Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/physiopathology , Hypertrophy, Right Ventricular/enzymology , Hypertrophy, Right Ventricular/etiology , Hypertrophy, Right Ventricular/physiopathology , Hypertrophy, Right Ventricular/prevention & control , Hypoxia/enzymology , Hypoxia/physiopathology , Injections, Subcutaneous , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocardium/enzymology , Myocardium/pathology , Protein Kinase Inhibitors/administration & dosage , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Pyridines/administration & dosage , RNA Interference , Rats , Receptor, Endothelin A/metabolism , Ventricular Dysfunction, Right/enzymology , Ventricular Dysfunction, Right/etiology , Ventricular Dysfunction, Right/physiopathology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL