Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Physiol Lung Cell Mol Physiol ; 321(2): L416-L428, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34189964

ABSTRACT

Pulmonary hypertension (PH) is a devastating disease characterized by progressive elevation of pulmonary vascular resistance, right ventricular failure, and ultimately death. We have shown previously that insulin receptor substrate 2 (IRS2), a molecule highly critical to insulin resistance and metabolism, has an anti-inflammatory role in Th2-skewed lung inflammation and pulmonary vascular remodeling. Here, we investigated the hypothesis that IRS2 has an immunomodulatory role in human and experimental PH. Expression analysis showed that IRS2 was significantly decreased in the pulmonary vasculature of patients with pulmonary arterial hypertension and in rat models of PH. In mice, genetic ablation of IRS2 enhanced the hypoxia-induced signaling pathway of Akt and Forkhead box O1 (FOXO1) in the lung tissue and increased pulmonary vascular muscularization, proliferation, and perivascular macrophage recruitment. Furthermore, mice with homozygous IRS2 gene deletion showed a significant gene dosage-dependent increase in pulmonary vascular remodeling and right ventricular hypertrophy in response to hypoxia. Functional studies with bone marrow-derived macrophages isolated from homozygous IRS2 gene-deleted mice showed that hypoxia exposure led to enhancement of the Akt and ERK signaling pathway followed by increases in the pro-PH macrophage activation markers, vascular endothelial growth factor-A and arginase 1. Our data suggest that IRS2 contributes to anti-inflammatory effects by regulating macrophage activation and recruitment, which may limit the vascular inflammation, remodeling, and right ventricular hypertrophy that are seen in PH pathology. Restoring the IRS2 pathway may be an effective therapeutic approach for the treatment of PH and right heart failure.


Subject(s)
Hypertension, Pulmonary/metabolism , Hypoxia/metabolism , Insulin Receptor Substrate Proteins/metabolism , Vascular Remodeling , Animals , Disease Models, Animal , Female , Forkhead Box Protein O1/genetics , Forkhead Box Protein O1/metabolism , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Hypoxia/genetics , Hypoxia/pathology , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Insulin Receptor Substrate Proteins/genetics , Male , Mice , Mice, Knockout , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rats , Rats, Nude
2.
Circ Res ; 122(12): 1689-1702, 2018 06 08.
Article in English | MEDLINE | ID: mdl-29545367

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PH) is a life-threatening condition associated with immune dysregulation and abnormal regulatory T cell (Treg) activity, but it is currently unknown whether and how abnormal Treg function differentially affects males and females. OBJECTIVE: To evaluate whether and how Treg deficiency differentially affects male and female rats in experimental PH. METHODS AND RESULTS: Male and female athymic rnu/rnu rats, lacking Tregs, were treated with the VEGFR2 (vascular endothelial growth factor receptor 2) inhibitor SU5416 or chronic hypoxia and evaluated for PH; some animals underwent Treg immune reconstitution before SU5416 administration. Plasma PGI2 (prostacyclin) levels were measured. Lung and right ventricles were assessed for the expression of the vasoprotective proteins COX-2 (cyclooxygenase 2), PTGIS (prostacyclin synthase), PDL-1 (programmed death ligand 1), and HO-1 (heme oxygenase 1). Inhibitors of these pathways were administered to athymic rats undergoing Treg immune reconstitution. Finally, human cardiac microvascular endothelial cells cocultured with Tregs were evaluated for COX-2, PDL-1, HO-1, and ER (estrogen receptor) expression, and culture supernatants were assayed for PGI2 and IL (interleukin)-10. SU5416-treatment and chronic hypoxia produced more severe PH in female than male athymic rats. Females were distinguished by greater pulmonary inflammation, augmented right ventricular fibrosis, lower plasma PGI2 levels, decreased lung COX-2, PTGIS, HO-1, and PDL-1 expression and reduced right ventricular PDL-1 levels. In both sexes, Treg immune reconstitution protected against PH development and raised levels of plasma PGI2 and cardiopulmonary COX-2, PTGIS, PDL-1, and HO-1. Inhibiting COX-2, HO-1, and PD-1 (programmed death 1)/PDL-1 pathways abrogated Treg protection. In vitro, human Tregs directly upregulated endothelial COX-2, PDL-1, HO-1, ERs and increased supernatant levels of PGI2 and IL-10. CONCLUSIONS: In 2 animal models of PH based on Treg deficiency, females developed more severe PH than males. The data suggest that females are especially reliant on the normal Treg function to counteract the effects of pulmonary vascular injury leading to PH.


Subject(s)
Hypertension, Pulmonary/prevention & control , Sex Factors , T-Lymphocytes, Regulatory/physiology , Angiogenesis Inhibitors/pharmacology , Animals , B7-H1 Antigen/analysis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Chronic Disease , Cyclooxygenase 2/analysis , Cyclooxygenase 2/metabolism , Cytochrome P-450 Enzyme System/analysis , Cytochrome P-450 Enzyme System/metabolism , Epoprostenol/antagonists & inhibitors , Epoprostenol/blood , Epoprostenol/metabolism , Female , Heme Oxygenase (Decyclizing)/analysis , Heme Oxygenase (Decyclizing)/antagonists & inhibitors , Heme Oxygenase (Decyclizing)/metabolism , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/chemically induced , Hypertension, Pulmonary/etiology , Hypoxia/complications , Indoles/pharmacology , Intramolecular Oxidoreductases/analysis , Intramolecular Oxidoreductases/antagonists & inhibitors , Intramolecular Oxidoreductases/metabolism , Lung/metabolism , Male , Prostaglandins I/biosynthesis , Pyrroles/pharmacology , Rats , Rats, Nude , Receptors, Estrogen/analysis , Receptors, Estrogen/antagonists & inhibitors , Receptors, Estrogen/metabolism , T-Lymphocytes, Regulatory/immunology , Vascular Endothelial Growth Factor Receptor-2/antagonists & inhibitors
3.
Circulation ; 136(20): 1920-1935, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-28935667

ABSTRACT

BACKGROUND: Immune dysregulation has been linked to occlusive vascular remodeling in pulmonary arterial hypertension (PAH) that is hereditary, idiopathic, or associated with other conditions. Circulating autoantibodies, lung perivascular lymphoid tissue, and elevated cytokines have been related to PAH pathogenesis but without a clear understanding of how these abnormalities are initiated, perpetuated, and connected in the progression of disease. We therefore set out to identify specific target antigens in PAH lung immune complexes as a starting point toward resolving these issues to better inform future application of immunomodulatory therapies. METHODS: Lung immune complexes were isolated and PAH target antigens were identified by liquid chromatography tandem mass spectrometry, confirmed by enzyme-linked immunosorbent assay, and localized by confocal microscopy. One PAH antigen linked to immunity and inflammation was pursued and a link to PAH pathophysiology was investigated by next-generation sequencing, functional studies in cultured monocytes and endothelial cells, and hemodynamic and lung studies in a rat. RESULTS: SAM domain and HD domain-containing protein 1 (SAMHD1), an innate immune factor that suppresses HIV replication, was identified and confirmed as highly expressed in immune complexes from 16 hereditary and idiopathic PAH versus 12 control lungs. Elevated SAMHD1 was localized to endothelial cells, perivascular dendritic cells, and macrophages, and SAMHD1 antibodies were prevalent in tertiary lymphoid tissue. An unbiased screen using metagenomic sequencing related SAMHD1 to increased expression of human endogenous retrovirus K (HERV-K) in PAH versus control lungs (n=4). HERV-K envelope and deoxyuridine triphosphate nucleotidohydrolase mRNAs were elevated in PAH versus control lungs (n=10), and proteins were localized to macrophages. HERV-K deoxyuridine triphosphate nucleotidohydrolase induced SAMHD1 and proinflammatory cytokines (eg, interleukin 6, interleukin 1ß, and tumor necrosis factor α) in circulating monocytes, pulmonary arterial endothelial cells, and also activated B cells. Vulnerability of pulmonary arterial endothelial cells (PAEC) to apoptosis was increased by HERV-K deoxyuridine triphosphate nucleotidohydrolase in an interleukin 6-independent manner. Furthermore, 3 weekly injections of HERV-K deoxyuridine triphosphate nucleotidohydrolase induced hemodynamic and vascular changes of pulmonary hypertension in rats (n=8) and elevated interleukin 6. CONCLUSIONS: Our study reveals that upregulation of the endogenous retrovirus HERV-K could both initiate and sustain activation of the immune system and cause vascular changes associated with PAH.


Subject(s)
Hypertension, Pulmonary/immunology , Inflammation Mediators/immunology , Up-Regulation/physiology , Viral Proteins/biosynthesis , Viral Proteins/immunology , Adolescent , Adult , Animals , Antigen-Antibody Complex/biosynthesis , Antigen-Antibody Complex/immunology , Cells, Cultured , Child , Coculture Techniques , Female , Humans , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/pathology , Infant , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/metabolism , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Leukocytes, Mononuclear/pathology , Male , Middle Aged , Rats , Rats, Sprague-Dawley , SAM Domain and HD Domain-Containing Protein 1/biosynthesis , SAM Domain and HD Domain-Containing Protein 1/immunology , Young Adult
4.
Circ Res ; 109(8): 867-79, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21868697

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is an incurable disease associated with viral infections and connective tissue diseases. The relationship between inflammation and disease pathogenesis in these disorders remains poorly understood. OBJECTIVE: To determine whether immune dysregulation due to absent T-cell populations directly contributes to the development of PAH. METHODS AND RESULTS: Vascular endothelial growth factor receptor 2 (VEGFR2) blockade induced significant pulmonary endothelial apoptosis in T-cell-deficient rats but not in immune-reconstituted (IR) rats. T cell-lymphopenia in association with VEGFR2 blockade resulted in periarteriolar inflammation with macrophages, and B cells even prior to vascular remodeling and elevated pulmonary pressures. IR prevented early inflammation and attenuated PAH development. IR with either CD8 T cells alone or with CD4-depleted spleen cells was ineffective in preventing PAH, whereas CD4-depleting immunocompetent euthymic animals increased PAH susceptibility. IR with either CD4(+)CD25(hi) or CD4(+)CD25(-) T cell subsets prior to vascular injury attenuated the development of PAH. IR limited perivascular inflammation and endothelial apoptosis in rat lungs in association with increased FoxP3(+), IL-10- and TGF-ß-expressing CD4 cells, and upregulation of pulmonary bone morphogenetic protein receptor type 2 (BMPR2)-expressing cells, a receptor that activates endothelial cell survival pathways. CONCLUSIONS: PAH may arise when regulatory T-cell (Treg) activity fails to control endothelial injury. These studies suggest that regulatory T cells normally function to limit vascular injury and may protect against the development of PAH.


Subject(s)
Endothelium, Vascular/immunology , Hypertension, Pulmonary/immunology , Hypertension, Pulmonary/prevention & control , T-Lymphocytes, Regulatory/immunology , Vascular System Injuries/immunology , Vascular System Injuries/prevention & control , Animals , Endothelium, Vascular/pathology , Hypertension, Pulmonary/pathology , Rats , Rats, Nude , Vascular System Injuries/pathology
5.
Am J Physiol Lung Cell Mol Physiol ; 303(3): L215-27, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22683569

ABSTRACT

Mechanical ventilation (MV) with O(2)-rich gas (MV-O(2)) offers life-saving treatment for newborn infants with respiratory failure, but it also can promote lung injury, which in neonates translates to defective alveolar formation and disordered lung elastin, a key determinant of lung growth and repair. Prior studies in preterm sheep and neonatal mice showed that MV-O(2) stimulated lung elastase activity, causing degradation and remodeling of matrix elastin. These changes yielded an inflammatory response, with TGF-ß activation, scattered elastic fibers, and increased apoptosis, culminating in defective alveolar septation and arrested lung growth. To see whether sustained inhibition of elastase activity would prevent these adverse pulmonary effects of MV-O(2), we did studies comparing wild-type (WT) and mutant neonatal mice genetically modified to express in their vascular endothelium the human serine elastase inhibitor elafin (Eexp). Five-day-old WT and Eexp mice received MV with 40% O(2) (MV-O(2)) for 24-36 h. WT and Eexp controls breathed 40% O(2) without MV. MV-O(2) increased lung elastase and MMP-9 activity, resulting in elastin degradation (urine desmosine doubled), TGF-ß activation (pSmad-2 increased 6-fold), apoptosis (cleaved-caspase-3 increased 10-fold), and inflammation (NF-κB activation, influx of neutrophils and monocytes) in lungs of WT vs. unventilated controls. These changes were blocked or blunted during MV-O(2) of Eexp mice. Scattered lung elastin and emphysematous alveoli observed in WT mice after 36 h of MV-O(2) were attenuated in Eexp mice. Both WT and Eexp mice showed defective VEGF signaling (decreased lung VEGF-R2 protein) and loss of pulmonary microvessels after lengthy MV-O(2), suggesting that elafin's beneficial effects during MV-O(2) derived primarily from preserving matrix elastin and suppressing lung inflammation, thereby enabling alveolar formation during MV-O(2). These results suggest that degradation and remodeling of lung elastin can contribute to defective lung growth in response to MV-O(2) and might be targeted therapeutically to prevent ventilator-induced neonatal lung injury.


Subject(s)
Elafin/physiology , Pancreatic Elastase/antagonists & inhibitors , Pneumonia/genetics , Pneumonia/prevention & control , Ventilator-Induced Lung Injury/genetics , Ventilator-Induced Lung Injury/prevention & control , Animals , Animals, Newborn , Apoptosis , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , Humans , Immunoblotting , Immunoenzyme Techniques , Macrophages/cytology , Macrophages/metabolism , Male , Mice , Mice, Transgenic , Monocytes/cytology , Monocytes/metabolism , Neutrophils/cytology , Neutrophils/metabolism , Oxygen/metabolism , Pancreatic Elastase/metabolism , Pulmonary Alveoli/pathology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Respiration, Artificial , Respiratory Insufficiency/prevention & control , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta/metabolism , Vascular Endothelial Growth Factor Receptor-2/genetics , Vascular Endothelial Growth Factor Receptor-2/metabolism
6.
Am J Respir Crit Care Med ; 184(5): 537-46, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21562133

ABSTRACT

RATIONALE: Mechanical ventilation with O2-rich gas (MV-O2) offers life-saving treatment for respiratory failure, but also promotes lung injury. We previously reported that MV-O2 of newborn mice increased lung elastase activity, causing elastin degradation and redistribution of elastic fibers from septal tips to alveolar walls. These changes were associated with transforming growth factor (TGF)-ß activation and increased apoptosis leading to defective alveolarization and lung growth arrest, as seen in neonatal chronic lung disease. OBJECTIVES: To determine if intratracheal treatment of newborn mice with the serine elastase inhibitor elafin would prevent MV-O2-induced lung elastin degradation and the ensuing cascade of events causing lung growth arrest. METHODS: Five-day-old mice were treated via tracheotomy with recombinant human elafin or vehicle (lactated-Ringer solution), followed by MV with 40% O2 for 8-24 hours; control animals breathed 40% O2 without MV. At study's end, lungs were harvested to assess key variables noted below. MEASUREMENTS AND MAIN RESULTS: MV-O2 of vehicle-treated pups increased lung elastase and matrix metalloproteinase-9 activity when compared with unventilated control animals, causing elastin degradation (urine desmosine doubled), TGF-ß activation (pSmad-2 tripled), and apoptosis (cleaved-caspase-3 increased 10-fold). Quantitative lung histology showed larger and fewer alveoli, greater inflammation, and scattered elastic fibers. Elafin blocked these MV-O2-induced changes. CONCLUSIONS: Intratracheal elafin, by blocking lung protease activity, prevented MV-O2-induced elastin degradation, TGF-ß activation, apoptosis, and dispersion of matrix elastin, and attenuated lung structural abnormalities noted in vehicle-treated mice after 24 hours of MV-O2. These findings suggest that elastin breakdown contributes to defective lung growth in response to MV-O2 and might be targeted therapeutically to prevent MV-O2-induced lung injury.


Subject(s)
Elafin/pharmacology , Lung/growth & development , Organogenesis/drug effects , Pancreatic Elastase/antagonists & inhibitors , Protease Inhibitors/pharmacology , Respiration, Artificial , Respiratory Insufficiency/therapy , Animals , Animals, Newborn , Apoptosis , Disease Models, Animal , Lung/drug effects , Lung/enzymology , Mice , Pancreatic Elastase/metabolism , Respiratory Insufficiency/enzymology , Respiratory Insufficiency/physiopathology
7.
Front Immunol ; 12: 684657, 2021.
Article in English | MEDLINE | ID: mdl-34489935

ABSTRACT

Pulmonary arterial hypertension (PAH) is a chronic, incurable condition characterized by pulmonary vascular remodeling, perivascular inflammation, and right heart failure. Regulatory T cells (Tregs) stave off autoimmunity, and there is increasing evidence for their compromised activity in the inflammatory milieu of PAH. Abnormal Treg function is strongly correlated with a predisposition to PAH in animals and patients. Athymic Treg-depleted rats treated with SU5416, an agent causing pulmonary vascular injury, develop PAH, which is prevented by infusing missing CD4+CD25highFOXP3+ Tregs. Abnormal Treg activity may also explain why PAH disproportionately affects women more than men. This mini review focuses on the role of Tregs in PAH with a special view to sexual dimorphism and the future promise of Treg therapy.


Subject(s)
Pulmonary Arterial Hypertension/immunology , Pulmonary Arterial Hypertension/prevention & control , T-Lymphocytes, Regulatory/immunology , Vascular System Injuries/immunology , Vascular System Injuries/prevention & control , Animals , Autoimmunity , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Humans , Indoles/adverse effects , Pulmonary Arterial Hypertension/pathology , Pyrroles/adverse effects , Rats , Sex Characteristics , Vascular System Injuries/pathology
8.
J Pharmacol Exp Ther ; 326(1): 51-8, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18375790

ABSTRACT

Exogenous prostacyclin is effective in reducing pulmonary vascular resistance in some forms of human pulmonary hypertension (PH). To explore whether endogenous prostaglandins played a similar role in pulmonary hypertension, we examined the effect of deleting cyclooxygenase (COX)-gene isoforms in a chronic hypoxia model of PH. Pulmonary hypertension, examined by direct measurement of right ventricular end systolic pressure (RVESP), right ventricular hypertrophy (n = 8), and hematocrit (n = 3), was induced by 3 weeks of hypobaric-hypoxia in wild-type and COX-knockout (KO) mice. RVESP was increased in wild-type hypoxic mice compared with normoxic controls (24.4 +/- 1.4 versus 13.8 +/- 1.9 mm Hg; n = 8; p < 0.05). COX-2 KO mice showed a greater increase in RVESP following hypoxia (36.8 +/- 2.7 mm Hg; p < 0.05). Urinary thromboxane (TX)B(2) excretion increased following hypoxia (44.6 +/- 11.1 versus 14.7 +/- 1.8 ng/ml; n = 6; p < 0.05), an effect that was exacerbated by COX-2 gene disruption (54.5 +/- 10.8 ng/ml; n = 6). In contrast, the increase in 6-keto-prostacyclin(1alpha) excretion following hypoxia was reduced by COX-2 gene disruption (29 +/- 3 versus 52 +/- 4.6 ng/ml; p < 0.01). Tail cut bleed times were lower following hypoxia, and there was evidence of intravascular thrombosis in lung vessels that was exacerbated by disruption of COX-2 and reduced by deletion of COX-1. The TXA(2)/endoperoxide receptor antagonist ifetroban (50 mg/kg/day) offset the effect of deleting the COX-2 gene, attenuating the hypoxia-induced rise in RVESP and intravascular thrombosis. COX-2 gene deletion exacerbates pulmonary hypertension, enhances sensitivity to TXA(2), and induces intravascular thrombosis in response to hypoxia. The data provide evidence that endogenous prostaglandins modulate the pulmonary response to hypoxia.


Subject(s)
Cyclooxygenase 2/physiology , Hypertension, Pulmonary/enzymology , Hypoxia/enzymology , Venous Thrombosis/enzymology , Animals , Cyclooxygenase 2/genetics , Female , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Hypoxia/complications , Hypoxia/genetics , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Venous Thrombosis/etiology , Venous Thrombosis/genetics
9.
Blood Adv ; 2(19): 2568-2580, 2018 10 09.
Article in English | MEDLINE | ID: mdl-30301812

ABSTRACT

Conventional local tumor irradiation (LTI), delivered in small daily doses over several weeks, is used clinically as a palliative, rather than curative, treatment for chemotherapy-resistant diffuse large B-cell lymphoma (DLBCL) for patients who are ineligible for hematopoietic cell transplantation. Our goal was to test the hypothesis that accelerated, but not conventional, LTI would be more curative by inducing T cell-mediated durable remissions. We irradiated subcutaneous A20 and BL3750 lymphoma tumors in mice with a clinically relevant total radiation dose of 30 Gy LTI, delivered in 10 doses of 3 Gy over 4 days (accelerated irradiation) or as 10 doses of 3 Gy over 12 days (conventional irradiation). Compared with conventional LTI, accelerated LTI resulted in more complete and durable tumor remissions. The majority of these mice were resistant to rechallenge with lymphoma cells, demonstrating the induction of memory antitumor immunity. The increased efficacy of accelerated LTI correlated with higher levels of tumor cell necrosis vs apoptosis and expression of "immunogenic cell death" markers, including calreticulin, heat shock protein 70 (Hsp70), and Hsp90. Accelerated LTI-induced remissions were not seen in immunodeficient Rag-2 -/- mice, CD8+ T-cell-depleted mice, or Batf-3 -/- mice lacking CD8α+ and CD103+ dendritic cells. Accelerated, but not conventional, LTI in immunocompetent hosts induced marked increases in tumor-infiltrating CD4+ and CD8+ T cells and MHCII+CD103+CD11c+ dendritic cells and corresponding reductions in exhausted PD-1+Eomes+CD8+ T cells and CD4+CD25+FOXP3+ regulatory T cells. These findings raise the possibility that accelerated LTI can provide effective immune control of human DLBCL.


Subject(s)
Lymphoma, B-Cell/immunology , T-Lymphocytes/immunology , Animals , Biomarkers , Cross-Priming/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Humans , Immunity , Immunophenotyping , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Lymphoma, B-Cell/mortality , Lymphoma, B-Cell/pathology , Lymphoma, B-Cell/radiotherapy , Male , Mice , Mice, Knockout , Radiotherapy/methods , Remission Induction , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
10.
Expert Rev Cardiovasc Ther ; 14(8): 939-51, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27096622

ABSTRACT

INTRODUCTION: Inflammatory cells are present in the lungs from patients with many, if not all, forms of severe pulmonary hypertension. AREAS COVERED: Historically the first inflammatory cell identified in the pulmonary vascular lesions was the mast cell. T and B lymphocytes, as well as macrophages, are present in and around the pulmonary arterioles and many patients have elevated blood levels of interleukin 1 and 6; some patients show elevated levels of leukotriene B4. An overlap between collagen-vascular disease-associated pulmonary arterial hypertension (PAH) and idiopathic PAH exists, yet only a few studies have been designed that evaluate the effect of anti-inflammatory treatments. Here we review the pertinent data that connect PAH and inflammation/autoimmune dysregulation and evaluate experimental models of severe PAH with an emphasis on the Sugen/athymic rat model of severe PAH. Expert commentary: We postulate that there are several inflammatory phenotypes and predict that there will be several anti-inflammatory treatment strategies for severe PAH.


Subject(s)
Familial Primary Pulmonary Hypertension/therapy , Hypertension, Pulmonary/therapy , Inflammation/therapy , Animals , Anti-Inflammatory Agents/therapeutic use , Humans , Hypertension, Pulmonary/physiopathology , Lung/pathology
11.
Circulation ; 110(17): 2701-7, 2004 Oct 26.
Article in English | MEDLINE | ID: mdl-15492320

ABSTRACT

BACKGROUND: Pulmonary hypertension induced by chronic hypoxia is characterized by thickening of pulmonary artery walls, elevated pulmonary vascular resistance, and right-heart failure. Prostacyclin analogues reduce pulmonary pressures in this condition; raising the possibility that cycloxygenase-2 (COX-2) modulates the response of the pulmonary vasculature to hypoxia. METHODS AND RESULTS: Sprague-Dawley rats in which pulmonary hypertension was induced by hypobaric hypoxia for 14 days were treated concurrently with the selective COX-2 inhibitor SC236 or vehicle. Mean pulmonary arterial pressure (mPAP) was elevated after hypoxia (28.1+/-3.2 versus 17.2+/-3.1 mm Hg; n=8, P<0.01), with thickening of small pulmonary arteries and increased COX-2 expression and prostacyclin formation. Selective inhibition of COX-2 aggravated the increase in mPAP (42.8+/-5.9 mm Hg; n=8, P<0.05), an effect that was attenuated by the thromboxane (TX) A2/prostaglandin endoperoxide receptor antagonist ifetroban. Urinary TXB2 increased during hypoxia (5.9+/-0.9 versus 1.2+/-0.2 ng/mg creatinine; n=6, P<0.01) and was further increased by COX-2 inhibition (8.5+/-0.7 ng/mg creatinine; n=6, P< 0.05). In contrast, urinary excretion of the prostacyclin metabolite 6-ketoprostaglandin F1alpha decreased with COX-2 inhibition (8.6+/-3.0 versus 27.0+/-4.8 ng/mg creatinine; n=6, P< 0.05). Platelet activation was enhanced after chronic hypoxia. COX-2 inhibition further reduced the PFA-100 closure time and enhanced platelet deposition in the smaller pulmonary arteries, effects that were attenuated by ifetroban. Mice with targeted disruption of the COX-2 gene exposed to chronic hypoxia had exacerbated right ventricular end-systolic pressure, whereas targeted disruption of COX-1 had no effect. CONCLUSIONS: COX-2 expression is increased and regulates platelet activity and intravascular thrombosis in hypoxia-induced pulmonary hypertension.


Subject(s)
Hypertension, Pulmonary/enzymology , Hypertension, Pulmonary/etiology , Hypoxia/complications , Platelet Activation , Prostaglandin-Endoperoxide Synthases/physiology , Animals , Cyclooxygenase 1 , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Hypertension, Pulmonary/physiopathology , Male , Membrane Proteins , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/urine , Pyrazoles/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Thrombosis/etiology , Thromboxane-A Synthase/metabolism
12.
Hypertension ; 66(6): 1227-1239, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26558820

ABSTRACT

A recent study demonstrated a significant role for leukotriene B4 (LTB4) causing pulmonary vascular remodeling in pulmonary arterial hypertension. LTB4 was found to directly injure luminal endothelial cells and promote growth of the smooth muscle cell layer of pulmonary arterioles. The purpose of this study was to determine the effects of LTB4 on the pulmonary adventitial layer, largely composed of fibroblasts. Here, we demonstrate that LTB4 enhanced human pulmonary artery adventitial fibroblast proliferation, migration, and differentiation in a dose-dependent manner through its cognate G-protein-coupled receptor, BLT1. LTB4 activated human pulmonary artery adventitial fibroblast by upregulating p38 mitogen-activated protein kinase as well as Nox4-signaling pathways. In an autoimmune model of pulmonary hypertension, inhibition of these pathways blocked perivascular inflammation, decreased Nox4 expression, reduced reactive oxygen species production, reversed arteriolar adventitial fibroblast activation, and attenuated pulmonary hypertension development. This study uncovers a novel mechanism by which LTB4 further promotes pulmonary arterial hypertension pathogenesis, beyond its established effects on endothelial and smooth muscle cells, by activating adventitial fibroblasts.


Subject(s)
Fibroblasts/drug effects , Hypertension, Pulmonary/metabolism , Leukotriene B4/pharmacology , Pulmonary Artery/metabolism , Animals , Arachidonate 5-Lipoxygenase/metabolism , Cell Differentiation/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Chemokines/genetics , Chemokines/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Gene Expression/drug effects , Humans , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Imidazoles/pharmacology , Leukotriene B4/metabolism , Microscopy, Confocal , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Pulmonary Artery/pathology , Pyridines/pharmacology , Rats, Nude , Reverse Transcriptase Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
13.
Sci Transl Med ; 5(200): 200ra117, 2013 Aug 28.
Article in English | MEDLINE | ID: mdl-23986401

ABSTRACT

Pulmonary hypertension (PH) is a serious condition that affects mainly young and middle-aged women, and its etiology is poorly understood. A prominent pathological feature of PH is accumulation of macrophages near the arterioles of the lung. In both clinical tissue and the SU5416 (SU)/athymic rat model of severe PH, we found that the accumulated macrophages expressed high levels of leukotriene A4 hydrolase (LTA4H), the biosynthetic enzyme for leukotriene B4 (LTB4). Moreover, macrophage-derived LTB4 directly induced apoptosis in pulmonary artery endothelial cells (PAECs). Further, LTB4 induced proliferation and hypertrophy of human pulmonary artery smooth muscle cells. We found that LTB4 acted through its receptor, BLT1, to induce PAEC apoptosis by inhibiting the protective endothelial sphingosine kinase 1 (Sphk1)-endothelial nitric oxide synthase (eNOS) pathway. Blocking LTA4H decreased in vivo LTB4 levels, prevented PAEC apoptosis, restored Sphk1-eNOS signaling, and reversed fulminant PH in the SU/athymic rat model of PH. Antagonizing BLT1 similarly reversed established PH. Inhibition of LTB4 biosynthesis or signal transduction in SU-treated athymic rats with established disease also improved cardiac function and reopened obstructed arterioles; this approach was also effective in the monocrotaline model of severe PH. Human plexiform lesions, one hallmark of PH, showed increased numbers of macrophages, which expressed LTA4H, and patients with connective tissue disease-associated pulmonary arterial hypertension exhibited significantly higher LTB4 concentrations in the systemic circulation than did healthy subjects. These results uncover a possible role for macrophage-derived LTB4 in PH pathogenesis and identify a pathway that may be amenable to therapeutic targeting.


Subject(s)
Endothelial Cells/pathology , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/prevention & control , Leukotriene B4/antagonists & inhibitors , Leukotriene B4/biosynthesis , Macrophages/metabolism , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Eicosanoids/metabolism , Endothelial Cells/drug effects , Endothelial Cells/enzymology , Familial Primary Pulmonary Hypertension , Female , Humans , Hypertrophy , Leukotriene B4/pharmacology , Macrophage Activation/drug effects , Macrophages/drug effects , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Pulmonary Artery/pathology , Rats , Signal Transduction/drug effects
14.
Trends Cardiovasc Med ; 21(6): 166-71, 2011 Aug.
Article in English | MEDLINE | ID: mdl-22814424

ABSTRACT

Pulmonary hypertension (PH) is a disease of high lethality arising from numerous causes. For a significant subset of PH patients, autoimmune biomarkers or frank autoimmune disease are simultaneously present, but the extent to which lung inflammation contributes to PH is unknown. However, emerging experimental and clinical evidence suggests that immune dysregulation may lead to the propagation of vascular injury and PH. A recent preclinical study demonstrated that regulatory T cells are important mediators normally enlisted to control inflammation and that, if absent or dysfunctional, may predispose to the development of PH.


Subject(s)
Hypertension, Pulmonary/immunology , T-Lymphocytes, Regulatory/physiology , Animals , Bone Morphogenetic Protein Receptors, Type II/metabolism , Disease Models, Animal , Humans , Hypertension, Pulmonary/prevention & control , Inflammation/immunology , Inflammation/prevention & control , Rats , Rats, Nude , Vascular System Injuries/immunology , Vascular System Injuries/prevention & control
SELECTION OF CITATIONS
SEARCH DETAIL