Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
Cell Immunol ; 387: 104718, 2023 05.
Article in English | MEDLINE | ID: mdl-37068442

ABSTRACT

Natural killer (NK) cell phenotype and function are altered in patients with prostate cancer, and increased NK cell activity is associated with a better prognosis in patients with disease. For patients with advanced stage prostate cancer, immunotherapies are a promising approach when standard treatment options have been exhausted. With the rapid emergence of NK cell-based therapies, it is important to understand the mechanisms by which NK cells can be triggered to kill cancer cells that have developed immune-evasive strategies. Altering the cytokine profiles of advanced prostate cancer cells may be an area to explore when considering ways in which NK cell activation can be modulated. We have previously demonstrated that combining the cytokine, IL-27, with TLR3 agonist, poly(I:C), changes cytokine secretion in the advanced prostate cancer models, PC3 and DU145 cells. Herein, we extend our previous work to study the effect of primary human NK cells on prostate cancer cell death in an in vitro co-culture model. Stimulating PC3 and DU145 cells with IL-27 and poly(I:C) induced IFN-ß secretion, which was required for activation of primary human NK cells to kill these stimulated prostate cancer cells. PC3 cells were more sensitized to NK cell-mediated killing when compared to DU145 cells, which was attributed to differential levels of IFN-ß produced in response to stimulation with IL-27 and poly(I:C). IFN-ß increased granzyme B secretion and membrane-bound TRAIL expression by co-cultured NK cells. We further demonstrated that these NK cells killed PC3 cells in a partially TRAIL-dependent manner. This work provides mechanistic insight into how the cytotoxic function of NK cells can be improved to target cancer cells.


Subject(s)
Antineoplastic Agents , Interleukin-27 , Prostatic Neoplasms , Male , Humans , Interleukin-27/metabolism , PC-3 Cells , Killer Cells, Natural/metabolism , Antineoplastic Agents/pharmacology , Cytokines/metabolism , Cell Line, Tumor , Prostatic Neoplasms/metabolism
2.
J Immunol ; 204(12): 3171-3181, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32332109

ABSTRACT

Circulating NK cells are known to convert to a type 1 innate lymphoid cell (ILC1)-like phenotype in response to TGF-ß exposure. However, the precise cellular changes defining this process as well as the downstream signaling pathways guiding it remain poorly defined, particularly in humans. We used mass cytometry by time-of-flight (CyTOF) to model this phenotypic shift in vitro and identify a synergistic activity of TGF-ß and IL-15 in this cellular conversion. CyTOF profiling identified substantial heterogeneity in the propensity of NK cells to adopt an ILC1-like phenotype in culture, characterized by the step-wise acquisition of various markers, including CD69, CD9, CD103, and CD49a. Activating and inhibitory receptors, including NKG2A, NKG2D, KIR2DL1, KIR3DL1, NKp30, NKp44, and NKp46, were all found to be upregulated exclusively on the cellular subsets that converted most readily in response to TGF-ß. An assessment of downstream TGF-ß signaling identified TAK1-mediated activation of p38 MAPK as the critical pathway driving conversion. IL-15 enhanced TGF-ß-mediated conversion through Ras:RAC1 signaling as well as via the activation of MEK/ERK. Interestingly, the adoption of an ILC1-like phenotype was independent of the effect of IL-15 or TGF-ß on mTOR, as the culture of NK cells in the presence of mTOR inhibitors, such as rapamycin or torin1, had minimal impact on the degree of conversion. In conclusion, we have used in vitro human culture systems and CyTOF to define the conversion of circulating NK cells to an ILC1-like phenotype and have clarified the pathways responsible for this process.


Subject(s)
Immunity, Innate/immunology , Interleukin-15/metabolism , Killer Cells, Natural/metabolism , Lymphocytes/metabolism , Mitogen-Activated Protein Kinases/metabolism , Signal Transduction/immunology , Transforming Growth Factor beta/metabolism , Biomarkers/metabolism , Cells, Cultured , Humans , Interleukin-15/immunology , Killer Cells, Natural/immunology , Lymphocytes/immunology , Mitogen-Activated Protein Kinases/immunology , Phenotype , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
3.
Arterioscler Thromb Vasc Biol ; 40(11): 2605-2618, 2020 11.
Article in English | MEDLINE | ID: mdl-32998516

ABSTRACT

OBJECTIVE: Pulmonary arterial hypertension is a disease of proliferative vascular occlusion that is strongly linked to mutations in BMPR2-the gene encoding the BMPR-II (BMP [bone morphogenetic protein] type II receptor). The endothelial-selective BMPR-II ligand, BMP9, reverses disease in animal models of pulmonary arterial hypertension and suppresses the proliferation of healthy endothelial cells. However, the impact of BMPR2 loss on the antiproliferative actions of BMP9 has yet to be assessed. Approach and Results: BMP9 suppressed proliferation in blood outgrowth endothelial cells from healthy control subjects but increased proliferation in blood outgrowth endothelial cells from pulmonary arterial hypertension patients with BMPR2 mutations. This shift from growth suppression to enhanced proliferation was recapitulated in control human pulmonary artery endothelial cells following siRNA-mediated BMPR2 silencing, as well as in mouse pulmonary endothelial cells isolated from endothelial-conditional Bmpr2 knockout mice (Bmpr2EC-/-). BMP9-induced proliferation was not attributable to altered metabolic activity or elevated TGFß (transforming growth factor beta) signaling but was linked to the prolonged induction of the canonical BMP target ID1 in the context of BMPR2 loss. In vivo, daily BMP9 administration to neonatal mice impaired both retinal and lung vascular patterning in control mice (Bmpr2EC+/+) but had no measurable effect on mice bearing a heterozygous endothelial Bmpr2 deletion (Bmpr2EC+/-) and caused excessive angiogenesis in both vascular beds for Bmpr2EC-/- mice. CONCLUSIONS: BMPR2 loss reverses the endothelial response to BMP9, causing enhanced proliferation. This finding has potential implications for the proposed translation of BMP9 as a treatment for pulmonary arterial hypertension and suggests the need for focused patient selection in clinical trials.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II/deficiency , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Growth Differentiation Factor 2/pharmacology , Pulmonary Arterial Hypertension/drug therapy , Adult , Aged , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Case-Control Studies , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Growth Differentiation Factor 2/toxicity , Humans , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Pulmonary Arterial Hypertension/genetics , Pulmonary Arterial Hypertension/metabolism , Pulmonary Arterial Hypertension/pathology , Signal Transduction , Young Adult
4.
Circulation ; 138(3): 287-304, 2018 07 17.
Article in English | MEDLINE | ID: mdl-29431643

ABSTRACT

BACKGROUND: Mitotic fission is increased in pulmonary arterial hypertension (PAH), a hyperproliferative, apoptosis-resistant disease. The fission mediator dynamin-related protein 1 (Drp1) must complex with adaptor proteins to cause fission. Drp1-induced fission has been therapeutically targeted in experimental PAH. Here, we examine the role of 2 recently discovered, poorly understood Drp1 adapter proteins, mitochondrial dynamics protein of 49 and 51 kDa (MiD49 and MiD51), in normal vascular cells and explore their dysregulation in PAH. METHODS: Immunoblots of pulmonary artery smooth muscle cells (control, n=6; PAH, n=8) and immunohistochemistry of lung sections (control, n=6; PAH, n=6) were used to assess the expression of MiD49 and MiD51. The effects of manipulating MiDs on cell proliferation, cell cycle, and apoptosis were assessed in human and rodent PAH pulmonary artery smooth muscle cells with flow cytometry. Mitochondrial fission was studied by confocal imaging. A microRNA (miR) involved in the regulation of MiD expression was identified using microarray techniques and in silico analyses. The expression of circulatory miR was assessed with quantitative reverse transcription-polymerase chain reaction in healthy volunteers (HVs) versus patients with PAH from Sheffield, UK (plasma: HV, n=29, PAH, n=27; whole blood: HV, n=11, PAH, n=14) and then confirmed in a cohort from Beijing, China (plasma: HV, n=19, PAH, n=36; whole blood: HV, n=20, PAH, n=39). This work was replicated in monocrotaline and Sugen 5416-hypoxia, preclinical PAH models. Small interfering RNAs targeting MiDs or an miR mimic were nebulized to rats with monocrotaline-induced PAH (n=4-10). RESULTS: MiD expression is increased in PAH pulmonary artery smooth muscle cells, which accelerates Drp1-mediated mitotic fission, increases cell proliferation, and decreases apoptosis. Silencing MiDs (but not other Drp1 binding partners, fission 1 or mitochondrial fission factor) promotes mitochondrial fusion and causes G1-phase cell cycle arrest through extracellular signal-regulated kinases 1/2- and cyclin-dependent kinase 4-dependent mechanisms. Augmenting MiDs in normal cells causes fission and recapitulates the PAH phenotype. MiD upregulation results from decreased miR-34a-3p expression. Circulatory miR-34a-3p expression is decreased in both patients with PAH and preclinical models of PAH. Silencing MiDs or augmenting miR-34a-3p regresses experimental PAH. CONCLUSIONS: In health, MiDs regulate Drp1-mediated fission, whereas in disease, epigenetic upregulation of MiDs increases mitotic fission, which drives pathological proliferation and apoptosis resistance. The miR-34a-3p-MiD pathway offers new therapeutic targets for PAH.


Subject(s)
GTP Phosphohydrolases/genetics , Hypertension, Pulmonary/genetics , Microtubule-Associated Proteins/genetics , Mitochondria/genetics , Mitochondrial Proteins/genetics , Myocytes, Smooth Muscle/physiology , Peptide Elongation Factors/genetics , Pulmonary Artery/pathology , Telangiectasis/congenital , Animals , Apoptosis , Cell Proliferation , Disease Models, Animal , Dynamins , Epigenesis, Genetic , Humans , MicroRNAs/genetics , Mitochondrial Dynamics , Protein Binding , Pulmonary Arterial Hypertension , RNA, Small Interfering/genetics , Rats , Telangiectasis/genetics
5.
Thorax ; 74(2): 164-176, 2019 02.
Article in English | MEDLINE | ID: mdl-30554141

ABSTRACT

INTRODUCTION: Skeletal muscle dysfunction is a clinically important complication of pulmonary arterial hypertension (PAH). Growth/differentiation factor 15 (GDF-15), a prognostic marker in PAH, has been associated with muscle loss in other conditions. We aimed to define the associations of GDF-15 and muscle wasting in PAH, to assess its utility as a biomarker of muscle loss and to investigate its downstream signalling pathway as a therapeutic target. METHODS: GDF-15 levels and measures of muscle size and strength were analysed in the monocrotaline (MCT) rat, Sugen/hypoxia mouse and in 30 patients with PAH. In C2C12 myotubes the downstream targets of GDF-15 were identified. The pathway elucidated was then antagonised in vivo. RESULTS: Circulating GDF-15 levels correlated with tibialis anterior (TA) muscle fibre diameter in the MCT rat (Pearson r=-0.61, p=0.003). In patients with PAH, plasma GDF-15 levels of <564 pg/L predicted those with preserved muscle strength with a sensitivity and specificity of ≥80%. In vitro GDF-15 stimulated an increase in phosphorylation of TGFß-activated kinase 1 (TAK1). Antagonising TAK1, with 5(Z)-7-oxozeaenol, in vitro and in vivo led to an increase in fibre diameter and a reduction in mRNA expression of atrogin-1 in both C2C12 cells and in the TA of animals who continued to grow. Circulating GDF-15 levels were also reduced in those animals which responded to treatment. CONCLUSIONS: Circulating GDF-15 is a biomarker of muscle loss in PAH that is responsive to treatment. TAK1 inhibition shows promise as a method by which muscle atrophy may be directly prevented in PAH. TRIAL REGISTRATION NUMBER: NCT01847716; Results.


Subject(s)
Growth Differentiation Factor 15/metabolism , Hypertension, Pulmonary/complications , MAP Kinase Kinase Kinases/metabolism , Muscular Atrophy/etiology , Transforming Growth Factor beta/metabolism , Adult , Animals , Biomarkers/metabolism , Blotting, Western , Enzyme-Linked Immunosorbent Assay , Female , Humans , Hypertension, Pulmonary/metabolism , Immunohistochemistry , Male , Mice , Middle Aged , Muscle, Skeletal/metabolism , Muscular Atrophy/metabolism , Rats , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Signal Transduction
6.
Am J Respir Cell Mol Biol ; 59(4): 467-478, 2018 10.
Article in English | MEDLINE | ID: mdl-29676587

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by increased proliferation and resistance to apoptosis of pulmonary vascular cells. Increased expression of translationally controlled tumor protein (TCTP), a prosurvival and antiapoptotic mediator, has recently been demonstrated in patients with heritable PAH; however, its role in the pathobiology of PAH remains unclear. Silencing of TCTP in blood outgrowth endothelial cells (BOECs) isolated from control subjects led to significant changes in morphology, cytoskeletal organization, increased apoptosis, and decreased directionality during migration. Because TCTP is also localized in extracellular vesicles, we isolated BOEC-derived extracellular vesicles (exosomes and microparticles) by sequential ultracentrifugation. BOECs isolated from patients harboring BMPR2 mutations released more exosomes than those derived from control subjects in proapoptotic conditions. Furthermore, TCTP expression was significantly higher in exosomes than in microparticles, indicating that TCTP is mainly exported via exosomes. Coculture assays demonstrated that exosomes transferred TCTP from ECs to pulmonary artery smooth muscle cells, suggesting a role for endothelial-derived TCTP in conferring proliferation and apoptotic resistance. In an experimental model of PAH, rats treated with monocrotaline demonstrated increased concentrations of TCTP in the lung and plasma. Consistent with this finding, we observed increased circulating TCTP levels in patients with idiopathic PAH compared with control subjects. Therefore, our data suggest an important role for TCTP in regulating the critical vascular cell phenotypes that have been implicated in the pathobiology of PAH. In addition, this research implicates TCTP as a potential biomarker for the onset and development of PAH.


Subject(s)
Biomarkers, Tumor/metabolism , Exosomes/metabolism , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology , Vascular Remodeling , Animals , Apoptosis , Biomarkers, Tumor/blood , Bone Morphogenetic Protein Receptors, Type II/genetics , Cell Movement , Cell Proliferation , Cell Shape , Disease Models, Animal , Endothelial Cells/metabolism , Exosomes/ultrastructure , Humans , Hypertension, Pulmonary/blood , Hypertension, Pulmonary/pathology , Lentivirus/metabolism , Lung/metabolism , Male , Monocrotaline , Mutation/genetics , Myocytes, Smooth Muscle/metabolism , Protein Transport , Pulmonary Artery/pathology , Rats, Sprague-Dawley , Tumor Protein, Translationally-Controlled 1
7.
Circulation ; 136(25): 2451-2467, 2017 Dec 19.
Article in English | MEDLINE | ID: mdl-28971999

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is characterized by abnormal growth and enhanced glycolysis of pulmonary artery endothelial cells. However, the mechanisms underlying alterations in energy production have not been identified. METHODS: Here, we examined the miRNA and proteomic profiles of blood outgrowth endothelial cells (BOECs) from patients with heritable PAH caused by mutations in the bone morphogenetic protein receptor type 2 (BMPR2) gene and patients with idiopathic PAH to determine mechanisms underlying abnormal endothelial glycolysis. We hypothesized that in BOECs from patients with PAH, the downregulation of microRNA-124 (miR-124), determined with a tiered systems biology approach, is responsible for increased expression of the splicing factor PTBP1 (polypyrimidine tract binding protein), resulting in alternative splicing of pyruvate kinase muscle isoforms 1 and 2 (PKM1 and 2) and consequently increased PKM2 expression. We questioned whether this alternative regulation plays a critical role in the hyperglycolytic phenotype of PAH endothelial cells. RESULTS: Heritable PAH and idiopathic PAH BOECs recapitulated the metabolic abnormalities observed in pulmonary artery endothelial cells from patients with idiopathic PAH, confirming a switch from oxidative phosphorylation to aerobic glycolysis. Overexpression of miR-124 or siRNA silencing of PTPB1 restored normal proliferation and glycolysis in heritable PAH BOECs, corrected the dysregulation of glycolytic genes and lactate production, and partially restored mitochondrial respiration. BMPR2 knockdown in control BOECs reduced the expression of miR-124, increased PTPB1, and enhanced glycolysis. Moreover, we observed reduced miR-124, increased PTPB1 and PKM2 expression, and significant dysregulation of glycolytic genes in the rat SUGEN-hypoxia model of severe PAH, characterized by reduced BMPR2 expression and endothelial hyperproliferation, supporting the relevance of this mechanism in vivo. CONCLUSIONS: Pulmonary vascular and circulating progenitor endothelial cells isolated from patients with PAH demonstrate downregulation of miR-124, leading to the metabolic and proliferative abnormalities in PAH ECs via PTPB1 and PKM1/PKM2. Therefore, the manipulation of this miRNA or its targets could represent a novel therapeutic approach for the treatment of PAH.


Subject(s)
Familial Primary Pulmonary Hypertension/pathology , Heterogeneous-Nuclear Ribonucleoproteins/metabolism , MicroRNAs/metabolism , Polypyrimidine Tract-Binding Protein/metabolism , Pyruvate Kinase/metabolism , Animals , Antagomirs/metabolism , Bone Morphogenetic Protein Receptors, Type II/antagonists & inhibitors , Bone Morphogenetic Protein Receptors, Type II/genetics , Bone Morphogenetic Protein Receptors, Type II/metabolism , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/metabolism , Familial Primary Pulmonary Hypertension/genetics , Familial Primary Pulmonary Hypertension/metabolism , Glycolysis , Heterogeneous-Nuclear Ribonucleoproteins/antagonists & inhibitors , Heterogeneous-Nuclear Ribonucleoproteins/genetics , Humans , Lim Kinases/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Monocarboxylic Acid Transporters/metabolism , Polypyrimidine Tract-Binding Protein/antagonists & inhibitors , Polypyrimidine Tract-Binding Protein/genetics , Pyruvate Kinase/genetics , RNA Interference , RNA, Small Interfering/metabolism , Rats , Smad1 Protein/metabolism , Smad5 Protein/metabolism , Symporters/metabolism
8.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L977-L990, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30234375

ABSTRACT

Natural killer (NK) cells are cytotoxic innate lymphoid cells with an established role in the regulation of vascular structure in pregnancy and cancer. Impaired NK cell function has been identified in patients with pulmonary arterial hypertension (PAH), a disease of obstructive vascular remodeling in the lungs, as well as in multiple rodent models of disease. However, the precise contribution of NK cell impairment to the initiation and progression of PAH remains unknown. Here, we report the development of spontaneous pulmonary hypertension in two independent genetic models of NK cell dysfunction, including Nfil3-/- mice, which are deficient in NK cells due to the absence of the NFIL3 transcription factor, and Ncr1-Gfp mice, which lack the NK activating receptor NKp46. Mouse models of NK insufficiency exhibited increased right ventricular systolic pressure and muscularization of the pulmonary arteries in the absence of elevated left ventricular end-diastolic pressure, indicating that the development of pulmonary hypertension was not secondary to left heart dysfunction. In cases of severe NK cell impairment or loss, a subset of mice failed to develop pulmonary hypertension and instead exhibited reduced systemic blood pressure, demonstrating an extension of vascular abnormalities beyond the pulmonary circulation into the systemic vasculature. In both mouse models, the development of PAH was linked to elevated interleukin-23 production, whereas systemic hypotension in Ncr1-Gfp mice was accompanied by a loss of angiopoietin-2. Together, these results support an important role for NK cells in the regulation of pulmonary and systemic vascular function and the pathogenesis of PAH.


Subject(s)
Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Killer Cells, Natural/pathology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Disease Models, Animal , Endothelial Cells/pathology , Humans , Lung/pathology , Mice , Natural Cytotoxicity Triggering Receptor 1/genetics , Pulmonary Artery/pathology , Vascular Remodeling/genetics
9.
Am J Respir Crit Care Med ; 195(4): 515-529, 2017 Feb 15.
Article in English | MEDLINE | ID: mdl-27648837

ABSTRACT

RATIONALE: Pulmonary arterial hypertension (PAH) is an obstructive vasculopathy characterized by excessive pulmonary artery smooth muscle cell (PASMC) proliferation, migration, and apoptosis resistance. This cancer-like phenotype is promoted by increased cytosolic calcium ([Ca2+]cyto), aerobic glycolysis, and mitochondrial fission. OBJECTIVES: To determine how changes in mitochondrial calcium uniporter (MCU) complex (MCUC) function influence mitochondrial dynamics and contribute to PAH's cancer-like phenotype. METHODS: PASMCs were isolated from patients with PAH and healthy control subjects and assessed for expression of MCUC subunits. Manipulation of the pore-forming subunit, MCU, in PASMCs was achieved through small interfering RNA knockdown or MCU plasmid-mediated up-regulation, as well as through modulation of the upstream microRNAs (miRs) miR-138 and miR-25. In vivo, nebulized anti-miRs were administered to rats with monocrotaline-induced PAH. MEASUREMENTS AND MAIN RESULTS: Impaired MCUC function, resulting from down-regulation of MCU and up-regulation of an inhibitory subunit, mitochondrial calcium uptake protein 1, is central to PAH's pathogenesis. MCUC dysfunction decreases intramitochondrial calcium ([Ca2+]mito), inhibiting pyruvate dehydrogenase activity and glucose oxidation, while increasing [Ca2+]cyto, promoting proliferation, migration, and fission. In PAH PASMCs, increasing MCU decreases cell migration, proliferation, and apoptosis resistance by lowering [Ca2+]cyto, raising [Ca2+]mito, and inhibiting fission. In normal PASMCs, MCUC inhibition recapitulates the PAH phenotype. In PAH, elevated miRs (notably miR-138) down-regulate MCU directly and also by decreasing MCU's transcriptional regulator cAMP response element-binding protein 1. Nebulized anti-miRs against miR-25 and miR-138 restore MCU expression, reduce cell proliferation, and regress established PAH in the monocrotaline model. CONCLUSIONS: These results highlight miR-mediated MCUC dysfunction as a unifying mechanism in PAH that can be therapeutically targeted.


Subject(s)
Calcium Channels/genetics , Calcium-Binding Proteins/genetics , Cation Transport Proteins/genetics , Genetic Therapy/methods , Hypertension, Pulmonary/genetics , MicroRNAs/genetics , Mitochondrial Membrane Transport Proteins/genetics , Muscle, Smooth, Vascular/pathology , Pulmonary Artery/pathology , Animals , Apoptosis/genetics , Calcium/metabolism , Calcium Channels/metabolism , Case-Control Studies , Cell Culture Techniques/methods , Cell Proliferation/drug effects , Cyclic AMP Response Element-Binding Protein/metabolism , Cytosol/metabolism , Disease Models, Animal , Down-Regulation/genetics , Glycolysis , Humans , Hypertension, Pulmonary/pathology , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Phenotype , Pulmonary Artery/drug effects , Pyruvate Dehydrogenase Complex/metabolism , Rats , Up-Regulation/genetics
11.
Am J Physiol Regul Integr Comp Physiol ; 313(6): R693-R705, 2017 Dec 01.
Article in English | MEDLINE | ID: mdl-28978513

ABSTRACT

Systemic hypertension, preeclampsia, and pulmonary arterial hypertension (PAH) are diseases of high blood pressure in the systemic or pulmonary circulation. Beyond the well-defined contribution of more traditional pathophysiological mechanisms, such as changes in the renin-angiotensin-aldosterone system, to the development of these hypertensive disorders, there is substantial clinical evidence supporting an important role for inflammation and immunity in the pathogenesis of each of these three conditions. Over the last decade, work in small animal models, bearing targeted deficiencies in specific cytokines or immune cell subsets, has begun to clarify the immune-mediated mechanisms that drive changes in vascular structure and tone in hypertensive disease. By summarizing the clinical and experimental evidence supporting a contribution of the immune system to systemic hypertension, preeclampsia, and PAH, the current review highlights the cellular and molecular pathways that are common to all three hypertensive disorders. These mechanisms are centered on an imbalance in CD4+ helper T cell populations, defined by excessive Th17 responses and impaired Treg activity, as well as the excessive activation or impairment of additional immune cell types, including macrophages, dendritic cells, CD8+ T cells, B cells, and natural killer cells. The identification of common immune mechanisms in systemic hypertension, preeclampsia, and PAH raises the possibility of new therapeutic strategies that target the immune component of hypertension across multiple disorders.


Subject(s)
Hypertension, Pulmonary/immunology , Hypertension/immunology , Pre-Eclampsia/immunology , Adult , Female , Humans , Hypertension/physiopathology , Hypertension/therapy , Hypertension, Pulmonary/physiopathology , Hypertension, Pulmonary/therapy , Immunotherapy/methods , Pre-Eclampsia/physiopathology , Pre-Eclampsia/therapy , Pregnancy , Translational Research, Biomedical
13.
Am J Respir Crit Care Med ; 192(11): 1355-65, 2015 Dec 01.
Article in English | MEDLINE | ID: mdl-26308618

ABSTRACT

RATIONALE: Schistosomiasis is a major cause of pulmonary arterial hypertension (PAH). Mutations in the bone morphogenetic protein type-II receptor (BMPR-II) are the commonest genetic cause of PAH. OBJECTIVES: To determine whether Bmpr2(+/-) mice are more susceptible to schistosomiasis-induced pulmonary vascular remodeling. METHODS: Wild-type (WT) and Bmpr2(+/-) mice were infected percutaneously with Schistosoma mansoni. At 17 weeks postinfection, right ventricular systolic pressure and liver and lung egg counts were measured. Serum, lung and liver cytokine, pulmonary vascular remodeling, and liver histology were assessed. MEASUREMENTS AND MAIN RESULTS: By 17 weeks postinfection, there was a significant increase in pulmonary vascular remodeling in infected mice. This was greater in Bmpr2(+/-) mice and was associated with an increase in egg deposition and cytokine expression, which induced pulmonary arterial smooth muscle cell proliferation, in the lungs of these mice. Interestingly, Bmpr2(+/-) mice demonstrated dilatation of the hepatic central vein at baseline and postinfection, compared with WT. Bmpr2(+/-) mice also showed significant dilatation of the liver sinusoids and an increase in inflammatory cells surrounding the central hepatic vein, compared with WT. This is consistent with an increase in the transhepatic passage of eggs. CONCLUSIONS: This study has shown that levels of BMPR-II expression modify the pulmonary vascular response to chronic schistosomiasis. The likely mechanism involves the increased passage of eggs to the lungs, caused by altered diameter of the hepatic veins and sinusoids in Bmpr2(+/-) mice. Genetically determined differences in the remodeling of hepatic vessels may represent a new risk factor for PAH associated with schistosomiasis.


Subject(s)
Bone Morphogenetic Protein Receptors, Type II , Hypertension, Pulmonary/physiopathology , Liver/parasitology , Pulmonary Artery/physiopathology , Schistosomiasis/physiopathology , Vascular Remodeling/genetics , Animals , Cell Proliferation , Disease Models, Animal , Female , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/parasitology , Mice , Pulmonary Artery/parasitology , Schistosoma mansoni , Schistosomiasis/genetics , Signal Transduction , Vascular Remodeling/physiology
14.
Circulation ; 129(21): 2125-35, 2014 May 27.
Article in English | MEDLINE | ID: mdl-24657995

ABSTRACT

BACKGROUND: Pulmonary arterial hypertension (PAH) is a lethal disease characterized by excessive proliferation of pulmonary vascular endothelial cells (ECs). Hereditary PAH (HPAH) is often caused by mutations in the bone morphogenetic protein receptor type 2 gene (BMPR2). However, the mechanisms by which these mutations cause PAH remain unclear. Therefore, we screened for dysregulated proteins in blood-outgrowth ECs of HPAH patients with BMPR2 mutations compared with healthy control subjects. METHODS AND RESULTS: A total of 416 proteins were detected with 2-dimensional PAGE in combination with liquid chromatography/tandem mass spectrometry analysis, of which 22 exhibited significantly altered abundance in blood-outgrowth ECs from patients with HPAH. One of these proteins, translationally controlled tumor protein (TCTP), was selected for further study because of its well-established role in promoting tumor cell growth and survival. Immunostaining showed marked upregulation of TCTP in lungs from patients with HPAH and idiopathic PAH, associated with remodeled vessels of complex lesions. Increased TCTP expression was also evident in the SU5416 rat model of severe and irreversible PAH, associated with intimal lesions, colocalizing with proliferating ECs and the adventitia of remodeled vessels but not in the vascular media. Furthermore, silencing of TCTP expression increased apoptosis and abrogated the hyperproliferative phenotype of blood-outgrowth ECs from patients with HPAH, raising the possibility that TCTP may be a link in the emergence of apoptosis-resistant, hyperproliferative vascular cells after EC apoptosis. CONCLUSION: Proteomic screening identified TCTP as a novel mediator of endothelial prosurvival and growth signaling in PAH, possibly contributing to occlusive pulmonary vascular remodeling triggered by EC apoptosis.


Subject(s)
Biomarkers, Tumor/physiology , Endothelial Cells/pathology , Endothelial Cells/physiology , Hypertension, Pulmonary/diagnosis , Hypertension, Pulmonary/genetics , Proteomics/methods , Adult , Aged , Animals , Bone Morphogenetic Protein Receptors, Type II/genetics , Familial Primary Pulmonary Hypertension , Female , Humans , Male , Middle Aged , Mutation/genetics , Rats , Rats, Sprague-Dawley , Survival/physiology , Tumor Protein, Translationally-Controlled 1 , Young Adult
17.
Am J Pathol ; 182(5): 1541-51, 2013 May.
Article in English | MEDLINE | ID: mdl-23518411

ABSTRACT

The capacity of imatinib mesylate to reverse established pulmonary arterial hypertension (PAH) has been attributed to a reduction in pulmonary arterial muscularization via inhibition of platelet-derived growth factor receptor-ß on vascular smooth muscle cells. However, there is also a significant immunomodulatory component to the action of imatinib that may account for its efficacy in PAH. We found that monocrotaline-induced pulmonary hypertension was associated with a significant decrease in pulmonary natural killer (NK) cells and T lymphocytes and the accumulation of macrophages in the lungs of F344 rats. The prevention of pulmonary hypertension by imatinib blocked these changes in pulmonary leukocyte content and induced elevations in pulmonary interferon-γ, tumor necrosis factor α, and IL-10, corresponding to the enhanced activity of splenic NK cells ex vivo. Treatment with anti-asialo GM1 antiserum (ASGM1), which ablated circulating NK cells and depleted T cells, eliminated the therapeutic benefit of imatinib. ASGM1-treated animals also exhibited significant pulmonary arteriolar muscularization in response to monocrotaline challenge compared with immunocompetent controls despite daily imatinib administration to both groups. In the athymic rat, imatinib decreased right ventricular hypertrophy and pulmonary arteriolar muscularization in monocrotaline-challenged animals versus saline-treated controls but did not prevent pulmonary macrophage accumulation or the development of pulmonary hypertension. These data demonstrate that the immunomodulatory effects of imatinib are critical to its therapeutic action in experimental PAH.


Subject(s)
Benzamides/therapeutic use , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/pathology , Lymphocytes/metabolism , Piperazines/therapeutic use , Pyrimidines/therapeutic use , Animals , Apoptosis/drug effects , Benzamides/pharmacology , Cytokines/metabolism , Disease Models, Animal , Hypertension, Pulmonary/chemically induced , Hypertrophy, Right Ventricular/pathology , Imatinib Mesylate , Immunomodulation/drug effects , Leukocyte Count , Lymphocyte Depletion , Lymphocytes/drug effects , Male , Monocrotaline , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Piperazines/pharmacology , Pyrimidines/pharmacology , Rats , Rats, Inbred F344 , Rats, Nude
18.
Hypertension ; 81(10): 2189-2201, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39162036

ABSTRACT

BACKGROUND: Drp1 (dynamin-related protein 1), a large GTPase, mediates the increased mitochondrial fission, which contributes to hyperproliferation of pulmonary artery smooth muscle cells in pulmonary arterial hypertension (PAH). We developed a potent Drp1 GTPase inhibitor, Drpitor1a, but its specificity, pharmacokinetics, and efficacy in PAH are unknown. METHODS: Drpitor1a's ability to inhibit recombinant and endogenous Drp1 GTPase was assessed. Drpitor1a's effects on fission were studied in control and PAH human pulmonary artery smooth muscle cells (hPASMC) and blood outgrowth endothelial cells (BOEC). Cell proliferation and apoptosis were studied in hPASMC. Pharmacokinetics and tissue concentrations were measured following intravenous and oral drug administration. Drpitor1a's efficacy in regressing monocrotaline-PAH was assessed in rats. In a pilot study, Drpitor1a reduced PA remodeling only in females. Subsequently, we compared Drpitor1a to vehicles in control and monocrotaline-PAH females. RESULTS: Drp1 GTPase activity was increased in PAH hPASMC. Drpitor1a inhibited the GTPase activity of recombinant and endogenous Drp1 and reversed the increased fission, seen in PAH hPASMC and PAH BOEC. Drpitor1a inhibited proliferation and induced apoptosis in PAH hPASMC without affecting electron transport chain activity, respiration, fission/fusion mediator expression, or mitochondrial Drp1 translocation. Drpitor1a did not inhibit proliferation or alter mitochondrial dynamics in normal hPASMC. Drpitor1a regressed monocrotaline-PAH without systemic vascular effects or toxicity. CONCLUSIONS: Drpitor1a is a specific Drp1 GTPase inhibitor that reduces mitochondrial fission in PAH hPASMC and PAH BOEC. Drpitor1a reduces proliferation and induces apoptosis in PAH hPASMC and regresses monocrotaline-PAH. Drp1 is a therapeutic target in PAH, and Drpitor1a is a potential therapy with an interesting therapeutic sexual dimorphism.


Subject(s)
Apoptosis , Cell Proliferation , Dynamins , Hypertension, Pulmonary , Mitochondrial Dynamics , Myocytes, Smooth Muscle , Pulmonary Artery , Animals , Female , Humans , Male , Rats , Apoptosis/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Disease Models, Animal , Dynamins/antagonists & inhibitors , Dynamins/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , GTP Phosphohydrolases/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Mitochondrial Dynamics/drug effects , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Pulmonary Artery/drug effects , Rats, Sprague-Dawley , Middle Aged
19.
Circulation ; 126(9): 1099-109, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22832786

ABSTRACT

BACKGROUND: Beyond their role as innate immune effectors, natural killer (NK) cells are emerging as important regulators of angiogenesis and vascular remodeling. Pulmonary arterial hypertension (PAH) is characterized by severe pulmonary vascular remodeling and has long been associated with immune dysfunction. Despite this association, a role for NK cells in disease pathology has not yet been described. METHODS AND RESULTS: Analysis of whole blood lymphocytes and isolated NK cells from PAH patients revealed an expansion of the functionally defective CD56(-)/CD16(+) NK subset that was not observed in patients with chronic thromboembolic pulmonary hypertension. NK cells from PAH patients also displayed decreased levels of the activating receptor NKp46 and the killer immunoglobulin-like receptors 2DL1/S1 and 3DL1, reduced secretion of the cytokine macrophage inflammatory protein-1ß, and a significant impairment in cytolytic function associated with decreased killer immunoglobulin-like receptor 3DL1 expression. Genotyping patients (n=222) and controls (n=191) for killer immunoglobulin-like receptor gene polymorphisms did not explain these observations. Rather, we show that NK cells from PAH patients exhibit increased responsiveness to transforming growth factor-ß, which specifically downregulates disease-associated killer immunoglobulin-like receptors. NK cell number and cytotoxicity were similarly decreased in the monocrotaline rat and chronic hypoxia mouse models of PAH, accompanied by reduced production of interferon-γ in NK cells from hypoxic mice. NK cells from PAH patients also produced elevated quantities of matrix metalloproteinase 9, consistent with a capacity to influence vascular remodeling. CONCLUSIONS: Our work is the first to identify an impairment of NK cells in PAH and suggests a novel and substantive role for innate immunity in the pathobiology of this disease.


Subject(s)
Hypertension, Pulmonary/immunology , Killer Cells, Natural/immunology , Adult , Aged , Animals , CD56 Antigen/analysis , Chemokine CCL4/metabolism , Cytotoxicity, Immunologic/drug effects , Extracellular Matrix/metabolism , Female , GPI-Linked Proteins/analysis , Genotype , Humans , Hypertension, Pulmonary/etiology , Hypertension, Pulmonary/genetics , Immunophenotyping , Killer Cells, Natural/chemistry , Killer Cells, Natural/drug effects , Killer Cells, Natural/pathology , Male , Matrix Metalloproteinase 9/analysis , Mice , Mice, Inbred C57BL , Middle Aged , Natural Cytotoxicity Triggering Receptor 1 , Pulmonary Embolism/complications , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, IgG/analysis , Receptors, KIR2DL1/biosynthesis , Receptors, KIR2DL1/genetics , Receptors, KIR3DL1/biosynthesis , Receptors, KIR3DL1/genetics , Receptors, KIR3DS1/biosynthesis , Receptors, KIR3DS1/genetics , Transforming Growth Factor beta/pharmacology
20.
Cancers (Basel) ; 15(6)2023 Mar 13.
Article in English | MEDLINE | ID: mdl-36980629

ABSTRACT

Natural killer (NK) cells are cytotoxic group 1 innate lymphoid cells (ILC), known for their role as killers of stressed, cancerous, and virally infected cells. Beyond this cytotoxic function, NK cell subsets can influence broader immune responses through cytokine production and have been linked to central roles in non-immune processes, such as the regulation of vascular remodeling in pregnancy and cancer. Attempts to exploit the anti-tumor functions of NK cells have driven the development of various NK cell-based therapies, which have shown promise in both pre-clinical disease models and early clinical trials. However, certain elements of the tumor microenvironment, such as elevated transforming growth factor (TGF)-ß, hypoxia, and indoalemine-2,3-dioxygenase (IDO), are known to suppress NK cell function, potentially limiting the longevity and activity of these approaches. Recent studies have also identified these factors as contributors to NK cell plasticity, defined by the conversion of classical cytotoxic NK cells into poorly cytotoxic, tissue-resident, or ILC1-like phenotypes. This review summarizes the current approaches for NK cell-based cancer therapies and examines the challenges presented by tumor-linked NK cell suppression and plasticity. Ongoing efforts to overcome these challenges are discussed, along with the potential utility of NK cell therapies to applications outside cancer.

SELECTION OF CITATIONS
SEARCH DETAIL