Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Sci Rep ; 14(1): 12127, 2024 05 27.
Article in English | MEDLINE | ID: mdl-38802469

ABSTRACT

Antibiotic resistance is a paramount global health issue, with numerous bacterial strains continually fortifying their resistance against diverse antibiotics. This surge in resistance levels primarily stems from the overuse and misuse of antibiotics in human, animal, and environmental contexts. In this study, we advocate for exploring alternative molecules exhibiting antibacterial properties to counteract the escalating antibiotic resistance. We identified a synthetic antimicrobial peptide (AMP) by using computational search in AMP public databases and further engineering through molecular docking and dynamics. Microbiological evaluation, cytotoxicity, genotoycity, and hemolysis experiments were then performed. The designed AMP underwent rigorous testing for antibacterial and antibiofilm activities against Methicillin-Resistant Staphylococcus aureus (MRSA) and Escherichia coli (E. coli), representing gram-positive and gram-negative bacteria, respectively. Subsequently, the safety profile of the AMP was assessed in vitro using human fibroblast cells and a human blood sample. The selected AMP demonstrated robust antibacterial and antibiofilm efficacy against MRSA and E. coli, with an added assurance of non-cytotoxicity and non-genotoxicity towards human fibroblasts. Also, the AMP did not demonstrate any hemolytic activity. Our findings emphasize the considerable promise of the AMP as a viable alternative antibacterial agent, showcasing its potential to combat antibiotic resistance effectively.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Peptides , Biofilms , Escherichia coli , Methicillin-Resistant Staphylococcus aureus , Microbial Sensitivity Tests , Biofilms/drug effects , Methicillin-Resistant Staphylococcus aureus/drug effects , Escherichia coli/drug effects , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemistry , Molecular Docking Simulation , Hemolysis/drug effects , Computer Simulation
2.
Int J Mol Sci ; 23(19)2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36232443

ABSTRACT

Impaired adipogenesis is associated with the development of insulin resistance and an increased risk of type 2 diabetes (T2D). GATA Binding Protein 3 (GATA3) is implicated in impaired adipogenesis and the onset of insulin resistance. Therefore, we hypothesize that inhibition of GATA3 could promote adipogenesis, restore healthy fat distribution, and enhance insulin signaling. Primary human preadipocytes were treated with GATA3 inhibitor (DNAzyme hgd40). Cell proliferation, adipogenic capacity, gene expression, and insulin signaling were measured following well-established protocols. BALB/c mice were treated with DNAzyme hgd40 over a period of 2 weeks. Liposomes loaded with DNAzyme hgd40, pioglitazone (positive), or vehicle (negative) controls were administered subcutaneously every 2 days at the right thigh. At the end of the study, adipose tissues were collected and weighed from the site of injection, the opposite side, and the omental depot. Antioxidant enzyme (superoxide dismutase and catalase) activities were assessed in animals' sera, and gene expression was measured using well-established protocols. In vitro GATA3 inhibition induced the adipogenesis of primary human preadipocytes and enhanced insulin signaling through the reduced expression of p70S6K. In vivo GATA3 inhibition promoted adipogenesis at the site of injection and reduced MCP-1 expression. GATA3 inhibition also reduced omental tissue size and PPARγ expression. These findings suggest that modulating GATA3 expression offers a potential therapeutic benefit by correcting impaired adipogenesis, promoting healthy fat distribution, improving insulin sensitivity, and potentially lowering the risk of T2D.


Subject(s)
DNA, Catalytic , Diabetes Mellitus, Type 2 , Insulin Resistance , Adipogenesis/genetics , Animals , Antioxidants/therapeutic use , Catalase , Diabetes Mellitus, Type 2/metabolism , Humans , Insulin/therapeutic use , Insulin Resistance/genetics , Liposomes/therapeutic use , Mice , Obesity/metabolism , PPAR gamma/metabolism , Pioglitazone/therapeutic use , Ribosomal Protein S6 Kinases, 70-kDa , Superoxide Dismutase
3.
Viruses ; 14(7)2022 07 15.
Article in English | MEDLINE | ID: mdl-35891532

ABSTRACT

There is no doubt that infectious diseases present global impact on the economy, society, health, mental state, and even political aspects, causing a long-lasting dent, and the situation will surely worsen if and when the viral spread becomes out of control, as seen during the still ongoing coronavirus disease 2019 (COVID-19) pandemic. Despite the considerable achievements made in viral prevention and treatment, there are still significant challenges that can be overcome through careful understanding of the viral mechanism of action to establish common ground for innovating new preventative and treatment strategies. Viruses can be regarded as devil nanomachines, and one innovative approach to face and stop the spread of viral infections is the development of nanoparticles that can act similar to them as drug/vaccine carriers. Moreover, we can use the properties that different viruses have in designing nanoparticles that reassemble the virus conformational structures but that do not present the detrimental threats to human health that native viruses possess. This review discusses the current preventative strategies (i.e., vaccination) used in facing viral infections and the associated limitations, highlighting the importance of innovating new approaches to face viral infectious diseases and discussing the current nanoapplications in vaccine development and the challenges that still face the nanovaccine field.


Subject(s)
COVID-19 , Viral Vaccines , COVID-19/prevention & control , COVID-19 Vaccines , Humans , Pandemics/prevention & control , SARS-CoV-2
4.
Int J Mol Sci ; 23(3)2022 Jan 26.
Article in English | MEDLINE | ID: mdl-35163328

ABSTRACT

Cardiovascular diseases (CVDs) are the leading causes of morbidity and mortality worldwide. However, despite the recent developments in the management of CVDs, the early and long outcomes vary considerably in patients, especially with the current challenges facing the detection and treatment of CVDs. This disparity is due to a lack of advanced diagnostic tools and targeted therapies, requiring innovative and alternative methods. Nanotechnology offers the opportunity to use nanomaterials in improving health and controlling diseases. Notably, nanotechnologies have recognized potential applicability in managing chronic diseases in the past few years, especially cancer and CVDs. Of particular interest is the use of nanoparticles as drug carriers to increase the pharmaco-efficacy and safety of conventional therapies. Different strategies have been proposed to use nanoparticles as drug carriers in CVDs; however, controversies regarding the selection of nanomaterials and nanoformulation are slowing their clinical translation. Therefore, this review focuses on nanotechnology for drug delivery and the application of nanomedicine in CVDs.


Subject(s)
Cardiovascular Diseases , Nanoparticles , Nanostructures , Cardiovascular Diseases/drug therapy , Drug Carriers/therapeutic use , Drug Delivery Systems , Humans , Nanomedicine/methods , Nanoparticles/therapeutic use , Nanostructures/therapeutic use , Nanotechnology
5.
Front Cell Dev Biol ; 9: 681347, 2021.
Article in English | MEDLINE | ID: mdl-34497803

ABSTRACT

Blood outgrowth smooth muscle cells (BO-SMCs) offer the means to study vascular cells without the requirement for surgery providing opportunities for drug discovery, tissue engineering, and personalized medicine. However, little is known about these cells which meant that their therapeutic potential remains unexplored. Our objective was to investigate for the first time the ability of BO-SMCs and vessel-derived smooth muscle cells to sense the thromboxane mimetic U46619 by measuring intracellular calcium elevation and contraction. U46619 (10-6 M) increased cytosolic calcium in BO-SMCs and vascular smooth muscle cells (VSMCs) but not in fibroblasts. Increased calcium signal peaked between 10 and 20 s after U46619 in both smooth muscle cell types. Importantly, U46619 (10-9 to 10-6 M) induced concentration-dependent contractions of both BO-SMCs and VSMCs but not in fibroblasts. In summary, we show that functional responses of BO-SMCs are in line with VSMCs providing critical evidence of their application in biomedical research.

6.
Sci Rep ; 11(1): 4336, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619326

ABSTRACT

Pulmonary arterial hypertension (PAH) is an incurable disease, although symptoms are treated with a range of dilator drugs. Despite their clinical benefits, these drugs are limited by systemic side-effects. It is, therefore, increasingly recognised that using controlled drug-release nanoformulation, with future modifications for targeted drug delivery, may overcome these limitations. This study presents the first evaluation of a promising nanoformulation (highly porous iron-based metal-organic framework (MOF); nanoMIL-89) as a carrier for the PAH-drug sildenafil, which we have previously shown to be relatively non-toxic in vitro and well-tolerated in vivo. In this study, nanoMIL-89 was prepared and charged with a payload of sildenafil (generating Sil@nanoMIL-89). Sildenafil release was measured by Enzyme-Linked Immunosorbent Assay (ELISA), and its effect on cell viability and dilator function in mouse aorta were assessed. Results showed that Sil@nanoMIL-89 released sildenafil over 6 h, followed by a more sustained release over 72 h. Sil@nanoMIL-89 showed no significant toxicity in human blood outgrowth endothelial cells for concentrations up to100µg/ml; however, it reduced the viability of the human pulmonary artery smooth muscle cells (HPASMCs) at concentrations > 3 µg/ml without inducing cellular cytotoxicity. Finally, Sil@nanoMIL-89 induced vasodilation of mouse aorta after a lag phase of 2-4 h. To our knowledge, this study represents the first demonstration of a novel nanoformulation displaying delayed drug release corresponding to vasodilator activity. Further pharmacological assessment of our nanoformulation, including in PAH models, is required and constitutes the subject of ongoing investigations.


Subject(s)
Metal-Organic Frameworks , Phosphodiesterase 5 Inhibitors/administration & dosage , Pulmonary Arterial Hypertension/drug therapy , Sildenafil Citrate/administration & dosage , Theranostic Nanomedicine , Animals , Aorta/drug effects , Cell Survival/drug effects , Drug Liberation , Humans , Kinetics , Metal-Organic Frameworks/chemistry , Metal-Organic Frameworks/ultrastructure , Mice , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Phosphodiesterase 5 Inhibitors/chemistry , Phosphodiesterase 5 Inhibitors/pharmacokinetics , Pulmonary Arterial Hypertension/etiology , Sildenafil Citrate/chemistry , Sildenafil Citrate/pharmacokinetics , Spectrum Analysis , Vasodilator Agents/administration & dosage , Vasodilator Agents/chemistry , Vasodilator Agents/pharmacokinetics
7.
Nanomaterials (Basel) ; 10(6)2020 May 27.
Article in English | MEDLINE | ID: mdl-32471187

ABSTRACT

Abstract: Cardiovascular diseases (CVDs) are the leading cause of morbidity and mortality worldwide. Alteration of endothelial cells and the underlying vasculature plays a central role in the pathogenesis of various CVDs. The application of nanoscale materials such as nanoparticles in biomedicine has opened new horizons in the treatment of CVDs. We have previously shown that the iron metal-organic framework nanoparticle, Materials Institut Lavoisier-89 (nanoMIL-89) represents a viable vehicle for future drug delivery of pulmonary arterial hypertension. In this study, we have assessed the cellular uptake of nanoMIL-89 in pulmonary artery endothelial and smooth muscle cells using microscopy imaging techniques. We also tested the cellular responses to nanoMIL-89 using molecular and cellular assays. Microscopic images showed cellular internalization of nanoMIL-89, packaging into endocytic vesicles, and passing to daughter cells during mitosis. Moreover, nanoMIL-89 showed anti-inflammatory activity without any significant cytotoxicity. Our results indicate that nanoMIL-89 formulation may offer promising therapeutic opportunities and set forth a new prototype for drug delivery not only in CVDs, but also for other diseases yet incurable, such as diabetes and cancer.

8.
Hum Mutat ; 40(2): 230-240, 2019 02.
Article in English | MEDLINE | ID: mdl-30408270

ABSTRACT

Homocystinuria is a rare inborn error of methionine metabolism caused by cystathionine ß-synthase (CBS) deficiency. The prevalence of homocystinuria in Qatar is 1:1,800 births, mainly due to a founder Qatari missense mutation, c.1006C>T; p.R336C (p.Arg336Cys). We characterized the structure-function relationship of the p.R336C-mutant protein and investigated the effect of different chemical chaperones to restore p.R336C-CBS activity using three models: in silico, ΔCBS yeast, and CRISPR/Cas9 p.R336C knock-in HEK293T and HepG2 cell lines. Protein modeling suggested that the p.R336C induces severe conformational and structural changes, perhaps influencing CBS activity. Wild-type CBS, but not the p.R336C mutant, was able to restore the yeast growth in ΔCBS-deficient yeast in a complementation assay. The p.R336C knock-in HEK293T and HepG2 cells decreased the level of CBS expression and reduced its structural stability; however, treatment of the p.R336C knock-in HEK293T cells with betaine, a chemical chaperone, restored the stability and tetrameric conformation of CBS, but not its activity. Collectively, these results indicate that the p.R336C mutation has a deleterious effect on CBS structure, stability, and activity, and using the chemical chaperones approach for treatment could be ineffective in restoring p.R336C CBS activity.


Subject(s)
Cystathionine beta-Synthase/genetics , Homocystinuria/genetics , Molecular Chaperones/genetics , Mutant Proteins/genetics , Computer Simulation , Cystathionine beta-Synthase/chemistry , Enzyme Stability , Gene Expression Regulation, Enzymologic/genetics , HEK293 Cells , Hep G2 Cells , Homocystinuria/metabolism , Homocystinuria/pathology , Humans , Methionine/metabolism , Molecular Chaperones/chemistry , Mutant Proteins/chemistry , Mutation, Missense/genetics , Protein Folding , Protein Structure, Tertiary , Qatar , Structure-Activity Relationship
9.
Pulm Circ ; 7(3): 643-653, 2017.
Article in English | MEDLINE | ID: mdl-28447910

ABSTRACT

Pulmonary arterial hypertension (PAH) is a progressive and debilitating condition. Despite promoting vasodilation, current drugs have a therapeutic window within which they are limited by systemic side effects. Nanomedicine uses nanoparticles to improve drug delivery and/or reduce side effects. We hypothesize that this approach could be used to deliver PAH drugs avoiding the systemic circulation. Here we report the use of iron metal organic framework (MOF) MIL-89 and PEGylated MIL-89 (MIL-89 PEG) as suitable carriers for PAH drugs. We assessed their effects on viability and inflammatory responses in a wide range of lung cells including endothelial cells grown from blood of donors with/without PAH. Both MOFs conformed to the predicted structures with MIL-89 PEG being more stable at room temperature. At concentrations up to 10 or 30 µg/mL, toxicity was only seen in pulmonary artery smooth muscle cells where both MOFs reduced cell viability and CXCL8 release. In endothelial cells from both control donors and PAH patients, both preparations inhibited the release of CXCL8 and endothelin-1 and in macrophages inhibited inducible nitric oxide synthase activity. Finally, MIL-89 was well-tolerated and accumulated in the rat lungs when given in vivo. Thus, the prototypes MIL-89 and MIL-89 PEG with core capacity suitable to accommodate PAH drugs are relatively non-toxic and may have the added advantage of being anti-inflammatory and reducing the release of endothelin-1. These data are consistent with the idea that these materials may not only be useful as drug carriers in PAH but also offer some therapeutic benefit in their own right.

10.
J Cardiovasc Transl Res ; 9(2): 162-4, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26960567

ABSTRACT

Pulmonary arterial hypertension (PAH) is a chronic and progressive disease which continues to carry an unacceptably high mortality and morbidity. The nitric oxide (NO) pathway has been implicated in the pathophysiology and progression of the disease. Its extremely short half-life and systemic effects have hampered the clinical use of NO in PAH. In an attempt to circumvent these major limitations, we have developed a new NO-nanomedicine formulation. The formulation was based on hydrogel-like polymeric composite NO-releasing nanoparticles (NO-RP). The kinetics of NO release from the NO-RP showed a peak at about 120 min followed by a sustained release for over 8 h. The NO-RP did not affect the viability or inflammation responses of endothelial cells. The NO-RP produced concentration-dependent relaxations of pulmonary arteries in mice with PAH induced by hypoxia. In conclusion, NO-RP drugs could considerably enhance the therapeutic potential of NO therapy for PAH.


Subject(s)
Antihypertensive Agents/pharmacology , Arterial Pressure/drug effects , Hypertension, Pulmonary/drug therapy , Nanoparticles , Nitric Oxide Donors/pharmacology , Nitric Oxide/metabolism , Pulmonary Artery/drug effects , Animals , Antihypertensive Agents/chemistry , Antihypertensive Agents/metabolism , Dose-Response Relationship, Drug , Drug Compounding , Hypertension, Pulmonary/metabolism , Hypertension, Pulmonary/physiopathology , Kinetics , Mice , Nanomedicine , Nitric Oxide Donors/chemistry , Nitric Oxide Donors/metabolism , Pulmonary Artery/metabolism , Pulmonary Artery/physiopathology
11.
Br J Clin Pharmacol ; 81(4): 621-33, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26561399

ABSTRACT

AIMS: In vivo platelet function is a product of intrinsic platelet reactivity, modifiable by dual antiplatelet therapy (DAPT), and the extrinsic inhibitory endothelial mediators, nitric oxide (NO) and prostacyclin (PGI2 ), that are powerfully potentiated by P2Y12 receptor blockade. This implies that for individual patients endothelial mediator production is an important determinant of DAPT effectiveness. Here, we have investigated this idea using platelets taken from healthy volunteers treated with anti-platelet drugs. METHODS: Three groups of male volunteers (n = 8) received either prasugrel (10 mg), aspirin (75 mg) or DAPT (prasugrel + aspirin) once daily for 7 days. Platelet reactivity in the presence of diethylammonium (Z)-1-(N,N-diethylamino)diazen-1-ium-1,2-diolate (DEA/NONOate) and PGI2 was studied before and following treatment. RESULTS: Ex vivo, PGI2 and/or DEA/NONOate had little inhibitory effect on TRAP-6-induced platelet reactivity in control conditions. However, in the presence of DAPT, combination of DEA/NONOate + PGI2 reduced platelet aggregation (74 ± 3% to 19 ± 6%, P < 0.05). In vitro studies showed even partial (25%) P2Y12 receptor blockade produced a significant (67 ± 2% to 39 ± 10%, P < 0.05) inhibition when DEA/NONOate + PGI2 was present. CONCLUSIONS: We have demonstrated that PGI2 and NO synergize with P2Y12 receptor antagonists to produce powerful platelet inhibition. Furthermore, even with submaximal P2Y12 blockade the presence of PGI2 and NO greatly enhances platelet inhibition. Our findings highlight the importance of endothelial mediator in vivo modulation of P2Y12 inhibition and introduces the concept of refining ex vivo platelet function testing by incorporating an assessment of endothelial function to predict thrombotic outcomes better and adjust therapy to prevent adverse outcomes in individual patients.


Subject(s)
Aspirin/pharmacology , Epoprostenol/pharmacology , Nitric Oxide/pharmacology , Platelet Activation/drug effects , Prasugrel Hydrochloride/pharmacology , Purinergic P2Y Receptor Antagonists/pharmacology , Adolescent , Adult , Aspirin/administration & dosage , Blood Platelets/drug effects , Drug Synergism , Epoprostenol/administration & dosage , Epoprostenol/metabolism , Healthy Volunteers , Humans , In Vitro Techniques , Male , Nitric Oxide/administration & dosage , Nitric Oxide/metabolism , Nitric Oxide Donors/pharmacology , Platelet Aggregation/drug effects , Prasugrel Hydrochloride/administration & dosage , Purinergic P2Y Receptor Antagonists/administration & dosage , Young Adult
12.
FASEB J ; 29(6): 2595-602, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25746794

ABSTRACT

There is an urgent unmet need for human tissue bioassays to predict cytokine storm responses to biologics. Current bioassays that detect cytokine storm responses in vitro rely on endothelial cells, usually from umbilical veins or cell lines, cocultured with freshly isolated peripheral blood mononuclear cells (PBMCs) from healthy adult volunteers. These assays therefore comprise cells from 2 separate donors and carry the disadvantage of mismatched tissues and lack the advantage of personalized medicine. Current assays also do not fully delineate mild (such as Campath) and severe (such as TGN1412) cytokine storm-inducing drugs. Here, we report a novel bioassay where endothelial cells grown from stem cells in the peripheral blood (blood outgrowth endothelial cells) and PBMCs from the same donor can be used to create an autologous coculture bioassay that responds by releasing a plethora of cytokines to authentic TGN1412 but only modestly to Campath and not to control antibodies such as Herceptin, Avastin, and Arzerra. This assay performed better than the traditional mixed donor assay in terms of cytokine release to TGN1412 and, thus, we suggest provides significant advancement and a definitive system by which biologics can be tested and paves the way for personalized medicine.


Subject(s)
Biological Products/pharmacology , Cytokines/metabolism , Endothelial Cells/drug effects , Leukocytes, Mononuclear/drug effects , Alemtuzumab , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Bevacizumab , Biological Assay/methods , Cell Proliferation/drug effects , Coculture Techniques , Culture Media/pharmacology , Endothelial Cells/cytology , Endothelial Cells/metabolism , Enzyme-Linked Immunosorbent Assay , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-2/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Reproducibility of Results , Serum/chemistry , Trastuzumab , Tumor Necrosis Factor-alpha/metabolism
13.
Biochem Biophys Res Commun ; 455(3-4): 172-7, 2014 Dec 12.
Article in English | MEDLINE | ID: mdl-25449267

ABSTRACT

Endothelial cells form a highly specialised lining of all blood vessels where they provide an anti-thrombotic surface on the luminal side and protect the underlying vascular smooth muscle on the abluminal side. Specialised functions of endothelial cells include their unique ability to release vasoactive hormones and to morphologically adapt to complex shear stress. Stem cell derived-endothelial cells have a growing number of applications and will be critical in any organ regeneration programme. Generally endothelial cells are identified in stem cell studies by well-recognised markers such as CD31. However, the ability of stem cell-derived endothelial cells to release vasoactive hormones and align with shear stress has not been studied extensively. With this in mind, we have compared directly the ability of endothelial cells derived from a range of stem cell sources, including embryonic stem cells (hESC-EC) and adult progenitors in blood (blood out growth endothelial cells, BOEC) with those cultured from mature vessels, to release the vasoconstrictor peptide endothelin (ET)-1, the cardioprotective hormone prostacyclin, and to respond morphologically to conditions of complex shear stress. All endothelial cell types, except hESC-EC, released high and comparable levels of ET-1 and prostacyclin. Under static culture conditions all endothelial cell types, except for hESC-EC, had the typical cobblestone morphology whilst hESC-EC had an elongated phenotype. When cells were grown under shear stress endothelial cells from vessels (human aorta) or BOEC elongated and aligned in the direction of shear. By contrast hESC-EC did not align in the direction of shear stress. These observations show key differences in endothelial cells derived from embryonic stem cells versus those from blood progenitor cells, and that BOEC are more similar than hESC-EC to endothelial cells from vessels. This may be advantageous in some settings particularly where an in vitro test bed is required. However, for other applications, because of low ET-1 release hESC-EC may prove to be protected from vascular inflammation.


Subject(s)
Endothelial Cells/cytology , Hormones/metabolism , Stem Cells/cytology , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/cytology , Endothelin-1/metabolism , Epoprostenol/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Inflammation , Interleukin-8/metabolism , Leukocytes, Mononuclear/cytology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Shear Strength , Stress, Mechanical , Vasoconstrictor Agents/metabolism
14.
Circ Res ; 114(4): 677-88, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24334027

ABSTRACT

RATIONALE: Evidence is increasing of a link between interferon (IFN) and pulmonary arterial hypertension (PAH). Conditions with chronically elevated endogenous IFNs such as systemic sclerosis are strongly associated with PAH. Furthermore, therapeutic use of type I IFN is associated with PAH. This was recognized at the 2013 World Symposium on Pulmonary Hypertension where the urgent need for research into this was highlighted. OBJECTIVE: To explore the role of type I IFN in PAH. METHODS AND RESULTS: Cells were cultured using standard approaches. Cytokines were measured by ELISA. Gene and protein expression were measured using reverse transcriptase polymerase chain reaction, Western blotting, and immunohistochemistry. The role of type I IFN in PAH in vivo was determined using type I IFN receptor knockout (IFNAR1(-/-)) mice. Human lung cells responded to types I and II but not III IFN correlating with relevant receptor expression. Type I, II, and III IFN levels were elevated in serum of patients with systemic sclerosis associated PAH. Serum interferon γ inducible protein 10 (IP10; CXCL10) and endothelin 1 were raised and strongly correlated together. IP10 correlated positively with pulmonary hemodynamics and serum brain natriuretic peptide and negatively with 6-minute walk test and cardiac index. Endothelial cells grown out of the blood of PAH patients were more sensitive to the effects of type I IFN than cells from healthy donors. PAH lung demonstrated increased IFNAR1 protein levels. IFNAR1(-/-) mice were protected from the effects of hypoxia on the right heart, vascular remodeling, and raised serum endothelin 1 levels. CONCLUSIONS: These data indicate that type I IFN, via an action of IFNAR1, mediates PAH.


Subject(s)
Hypertension, Pulmonary/immunology , Interferon-alpha/immunology , Interferon-beta/immunology , Receptor, Interferon alpha-beta/immunology , Scleroderma, Systemic/immunology , Animals , Cells, Cultured , Chemokine CXCL10/immunology , Chemokine CXCL10/metabolism , Disease Models, Animal , Endothelial Cells/cytology , Endothelial Cells/immunology , Endothelin-1/immunology , Endothelin-1/metabolism , Familial Primary Pulmonary Hypertension , Humans , Hypertension, Pulmonary/metabolism , Interferon-alpha/metabolism , Interferon-alpha/pharmacology , Interferon-beta/metabolism , Interferon-beta/pharmacology , Interferon-gamma/immunology , Interferon-gamma/pharmacology , Lung/cytology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organ Culture Techniques , Receptor, Interferon alpha-beta/genetics , Receptor, Interferon alpha-beta/metabolism , Scleroderma, Systemic/metabolism , Signal Transduction/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...