Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
FASEB J ; 34(1): 1745-1754, 2020 01.
Article in English | MEDLINE | ID: mdl-31914612

ABSTRACT

Blood vessels are comprised of endothelial and smooth muscle cells. Obtaining both types of cells from vessels of living donors is not possible without invasive surgery. To address this, we have devised a strategy whereby human endothelial and smooth muscle cells derived from blood progenitors from the same donor could be cultured with autologous leukocytes to generate a same donor "vessel in a dish" bioassay. Autologous sets of blood outgrowth endothelial cells (BOECs), smooth muscle cells (BO-SMCs), and leukocytes were obtained from four donors. Cells were treated in monoculture and cumulative coculture conditions. The endothelial specific mediator endothelin-1 along with interleukin (IL)-6, IL-8, tumor necrosis factor α, and interferon gamma-induced protein 10 were measured under control culture conditions and after stimulation with cytokines. Cocultures remained viable throughout. The profile of individual mediators released from cells was consistent with what we know of endothelial and smooth muscle cells cultured from blood vessels. For the first time, we report a proof of concept study where autologous blood outgrowth "vascular" cells and leukocytes were studied alone and in coculture. This novel bioassay has usefulness in vascular biology research, patient phenotyping, drug testing, and tissue engineering.


Subject(s)
Endothelial Cells/physiology , Leukocytes/physiology , Muscle, Smooth, Vascular/physiology , Myocytes, Smooth Muscle/physiology , Biological Assay/methods , Cells, Cultured , Coculture Techniques/methods , Cytokines/metabolism , Drug Discovery/methods , Endothelial Cells/metabolism , Humans , Interleukin-6/metabolism , Leukocytes/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype , Tissue Engineering/methods , Tumor Necrosis Factor-alpha/metabolism
3.
Circ Res ; 122(4): 555-559, 2018 02 16.
Article in English | MEDLINE | ID: mdl-29298774

ABSTRACT

RATIONALE: The balance between vascular prostacyclin, which is antithrombotic, and platelet thromboxane A2, which is prothrombotic, is fundamental to cardiovascular health. Prostacyclin and thromboxane A2 are formed after the concerted actions of cPLA2α (cytosolic phospholipase A2) and COX (cyclooxygenase). Urinary 2,3-dinor-6-keto-PGF1α (PGI-M) and 11-dehydro-TXB2 (TX-M) have been taken as biomarkers of prostacyclin and thromboxane A2 formation within the circulation and used to explain COX biology and patient phenotypes, despite concerns that urinary PGI-M and TX-M originate in the kidney. OBJECTIVE: We report data from a remarkable patient carrying an extremely rare genetic mutation in cPLA2α, causing almost complete loss of prostacyclin and thromboxane A2, who was transplanted with a normal kidney resulting in an experimental scenario of whole-body cPLA2α knockout, kidney-specific knockin. By studying this patient, we can determine definitively the contribution of the kidney to the productions of PGI-M and TX-M and test their validity as markers of prostacyclin and thromboxane A2 in the circulation. METHODS AND RESULTS: Metabolites were measured using liquid chromatography-tandem mass spectrometry. Endothelial cells were grown from blood progenitors. Before kidney transplantation, the patient's endothelial cells and platelets released negligible levels of prostacyclin (measured as 6-keto-prostaglandin F1α) and thromboxane A2 (measured as TXB2), respectively. Likewise, the urinary levels of PGI-M and TX-M were very low. After transplantation and the establishment of normal renal function, the levels of PGI-M and TX-M in the patient's urine rose to within normal ranges, whereas endothelial production of prostacyclin and platelet production of thromboxane A2 remained negligible. CONCLUSIONS: These data show that PGI-M and TX-M can be derived exclusively from the kidney without contribution from prostacyclin made by endothelial cells or thromboxane A2 by platelets in the general circulation. Previous work relying on urinary metabolites of prostacyclin and thromboxane A2 as markers of whole-body endothelial and platelet function now requires reevaluation.


Subject(s)
6-Ketoprostaglandin F1 alpha/analogs & derivatives , Allografts/metabolism , Kidney Transplantation , Kidney/metabolism , Loss of Function Mutation , Phospholipases A2, Cytosolic/genetics , Thromboxane B2/analogs & derivatives , 6-Ketoprostaglandin F1 alpha/metabolism , 6-Ketoprostaglandin F1 alpha/urine , Biomarkers/urine , Cells, Cultured , Female , Humans , Middle Aged , Phenotype , Phospholipases A2, Cytosolic/metabolism , Prostaglandin-Endoperoxide Synthases/metabolism , Thromboxane B2/metabolism , Thromboxane B2/urine
4.
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.

5.
Cytokine ; 85: 101-8, 2016 09.
Article in English | MEDLINE | ID: mdl-27309676

ABSTRACT

In October 2013, the International Life Sciences Institute - Health and Environmental Sciences Institute Immunotoxicology Technical Committee (ILSI-HESI ITC) held a one-day workshop entitled, "Workshop on Cytokine Release: State-of-the-Science, Current Challenges and Future Directions". The workshop brought together scientists from pharmaceutical, academic, health authority, and contract research organizations to discuss novel approaches and current challenges for the use of in vitro cytokine release assays (CRAs) for the identification of cytokine release syndrome (CRS) potential of novel monoclonal antibody (mAb) therapeutics. Topics presented encompassed a regulatory perspective on cytokine release and assessment, case studies regarding the translatability of preclinical cytokine data to the clinic, and the latest state of the science of CRAs, including comparisons between mAb therapeutics within one platform and across several assay platforms, a novel physiological assay platform, and assay optimization approaches such as determination of FcR expression profiles and use of statistical tests. The data and approaches presented confirmed that multiple CRA platforms are in use for identification of CRS potential and that the choice of a particular CRA platform is highly dependent on the availability of resources for individual laboratories (e.g. positive and negative controls, number of human blood donors), the assay through-put required, and the mechanism-of-action of the therapeutic candidate to be tested. Workshop participants agreed that more data on the predictive performance of CRA platforms is needed, and current efforts to compare in vitro assay results with clinical cytokine assessments were discussed. In summary, many laboratories continue to focus research efforts on the improvement of the translatability of current CRA platforms as well explore novel approaches which may lead to more accurate, and potentially patient-specific, CRS prediction in the future.


Subject(s)
Cytokines/blood , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Biological Assay/methods , Humans , Immune System Diseases/blood , Immune System Diseases/drug therapy
6.
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
7.
FASEB J ; 29(11): 4568-78, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26183771

ABSTRACT

Eicosanoids are important vascular regulators, but the phospholipase A2 (PLA2) isoforms supporting their production within the cardiovascular system are not fully understood. To address this, we have studied platelets, endothelial cells, and leukocytes from 2 siblings with a homozygous loss-of-function mutation in group IVA cytosolic phospholipase A2 (cPLA2α). Chromatography/mass spectrometry was used to determine levels of a broad range of eicosanoids produced by isolated vascular cells, and in plasma and urine. Eicosanoid release data were paired with studies of cellular function. Absence of cPLA2α almost abolished eicosanoid synthesis in platelets (e.g., thromboxane A2, control 20.5 ± 1.4 ng/ml vs. patient 0.1 ng/ml) and leukocytes [e.g., prostaglandin E2 (PGE2), control 21.9 ± 7.4 ng/ml vs. patient 1.9 ng/ml], and this was associated with impaired platelet activation and enhanced inflammatory responses. cPLA2α-deficient endothelial cells showed reduced, but not absent, formation of prostaglandin I2 (prostacyclin; control 956 ± 422 pg/ml vs. patient 196 pg/ml) and were primed for inflammation. In the urine, prostaglandin metabolites were selectively influenced by cPLA2α deficiency. For example, prostacyclin metabolites were strongly reduced (18.4% of control) in patients lacking cPLA2α, whereas PGE2 metabolites (77.8% of control) were similar to healthy volunteer levels. These studies constitute a definitive account, demonstrating the fundamental role of cPLA2α to eicosanoid formation and cellular responses within the human circulation.


Subject(s)
Antigens, Human Platelet/genetics , Blood Platelets/enzymology , Dinoprostone/genetics , Endothelial Cells/enzymology , Epoprostenol/genetics , Leukocytes/enzymology , Mutation , Adult , Blood Platelets/pathology , Dinoprostone/biosynthesis , Endothelial Cells/pathology , Epoprostenol/biosynthesis , Female , Humans , Leukocytes/pathology , Male , Platelet Activation/genetics
8.
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
10.
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
11.
PLoS One ; 9(4): e91119, 2014.
Article in English | MEDLINE | ID: mdl-24690886

ABSTRACT

Human embryonic stem cell-derived endothelial cells (hESC-EC), as well as other stem cell derived endothelial cells, have a range of applications in cardiovascular research and disease treatment. Endothelial cells sense Gram-negative bacteria via the pattern recognition receptors (PRR) Toll-like receptor (TLR)-4 and nucleotide-binding oligomerisation domain-containing protein (NOD)-1. These pathways are important in terms of sensing infection, but TLR4 is also associated with vascular inflammation and atherosclerosis. Here, we have compared TLR4 and NOD1 responses in hESC-EC with those of endothelial cells derived from other stem cells and with human umbilical vein endothelial cells (HUVEC). HUVEC, endothelial cells derived from blood progenitors (blood outgrowth endothelial cells; BOEC), and from induced pluripotent stem cells all displayed both a TLR4 and NOD1 response. However, hESC-EC had no TLR4 function, but did have functional NOD1 receptors. In vivo conditioning in nude rats did not confer TLR4 expression in hESC-EC. Despite having no TLR4 function, hESC-EC sensed Gram-negative bacteria, a response that was found to be mediated by NOD1 and the associated RIP2 signalling pathways. Thus, hESC-EC are TLR4 deficient but respond to bacteria via NOD1. This data suggests that hESC-EC may be protected from unwanted TLR4-mediated vascular inflammation, thus offering a potential therapeutic advantage.


Subject(s)
Endothelial Cells/metabolism , Endothelial Cells/microbiology , Haemophilus influenzae/physiology , Human Embryonic Stem Cells/cytology , Human Embryonic Stem Cells/microbiology , Nod1 Signaling Adaptor Protein/metabolism , Animals , Endothelial Cells/cytology , Gene Knockdown Techniques , Haemophilus Infections/microbiology , Human Umbilical Vein Endothelial Cells/metabolism , Human Umbilical Vein Endothelial Cells/microbiology , Humans , Induced Pluripotent Stem Cells/metabolism , RNA, Small Interfering/metabolism , Rats, Nude , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Stem Cell Transplantation , Toll-Like Receptor 4/metabolism
12.
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
13.
Glob Cardiol Sci Pract ; 2014(4): 382-93, 2014.
Article in English | MEDLINE | ID: mdl-25780793

ABSTRACT

Prostacyclin is a powerful cardioprotective hormone released by the endothelium of all blood vessels. Prostacyclin exists in equilibrium with other vasoactive hormones and a disturbance in the balance of these factors leads to cardiovascular disease including pulmonary arterial hypertension. Since it's discovery in the 1970s concerted efforts have been made to make the best therapeutic utility of prostacyclin, particularly in the treatment of pulmonary arterial hypertension. This has centred on working out the detailed pharmacology of prostacyclin and then synthesising new molecules based on its structure that are more stable or more easily tolerated. In addition, newer molecules have been developed that are not analogues of prostacyclin but that target the receptors that prostacyclin activates. Prostacyclin and related drugs have without doubt revolutionised the treatment and management of pulmonary arterial hypertension but are seriously limited by side effects within the systemic circulation. With the dawn of nanomedicine and targeted drug or stem cell delivery systems it will, in the very near future, be possible to make new formulations of prostacyclin that can evade the systemic circulation allowing for safe delivery to the pulmonary vessels. In this way, the full therapeutic potential of prostacyclin can be realised opening the possibility that pulmonary arterial hypertension will become, if not curable, a chronic manageable disease that is no longer fatal. This review discusses these and other issues relating to prostacyclin and its use in pulmonary arterial hypertension.

14.
Article in English | MEDLINE | ID: mdl-23742950

ABSTRACT

Cyclooxygenase (COX) is required for prostanoid (e.g. prostaglandin PGE2) production. Constitutive COX-1 and inducible COX-2 are implicated in lung diseases, such as idiopathic pulmonary fibrosis (IPF). Using lung fibroblasts from humans and wild type, COX-1(-/-) and COX-2(-/-) mice, we investigated how COX activity modulates cell growth and inflammatory responses induced by activators of Toll-like receptors (TLRs) 1-8. In mouse tissue, PGE2 release from fresh lung was COX-1 driven, in lung in culture (24h) COX-1 and COX-2 driven, and from proliferating lung fibroblasts exclusively COX-2 driven. COX-2 limited proliferation in lung fibroblasts and both isoforms limited KC release induced by a range of TLR agonists. Less effect of COX was seen on TLR-induced IP-10 release. In human lung fibroblasts inhibition of COX with diclofenac was associated with increased release of IL-8 and IP-10. Our results may have implications for the treatment of IPF.


Subject(s)
Cyclooxygenase 1/genetics , Cyclooxygenase 2/genetics , Cytokines/metabolism , Fibroblasts/enzymology , Membrane Proteins/genetics , Toll-Like Receptors/agonists , Animals , Cell Proliferation , Cells, Cultured , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/pharmacology , Diclofenac/pharmacology , Dinoprostone/metabolism , Fibroblasts/immunology , Fibroblasts/metabolism , Host-Pathogen Interactions , Humans , Idiopathic Pulmonary Fibrosis/enzymology , Immunity, Innate , Lipopolysaccharides/pharmacology , Lung/immunology , Lung/pathology , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Poly I-C/pharmacology , Toll-Like Receptors/metabolism
15.
Br J Clin Pharmacol ; 75(4): 897-906, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22703602

ABSTRACT

Stem cell therapy and organ regeneration are therapeutic approaches that will, we suggest, become mainstream for the treatment of human disease. Endothelial cells, which line the luminal surface of every vessel in the body, are essential components in any organ regeneration programme. There are a number of potentially therapeutic endothelial cell types, including embryonic, adult progenitor and induced pluripotent stem cell-derived endothelial cells, as well as host vascular cells. The features (benefits as well as disadvantages) of each cell type that make them potentially useful in therapy are important to consider. The field of stem cell biology is well developed in terms of protocols for generating endothelium. However, where there is a distinct and urgent unmet need for knowledge concerning how the endothelial cells from these different sources function as endothelium and how susceptible they may be to inflammation and atherosclerosis. Furthermore, where stem cells have been used in clinical trials there is little commonality in protocols for deriving the cells (and thereby the specific phenotype of cells used), administering the cells, dosing the cells and/or in assessing efficacy attributed to the cells themselves. This review discusses these and other issues relating to stem cell-derived endothelial cells in cell therapy for cardiovascular disease.


Subject(s)
Cardiovascular Diseases/pathology , Cardiovascular Diseases/surgery , Cell Transplantation/methods , Endothelial Cells/cytology , Endothelial Cells/transplantation , Stem Cells/cytology , Bone Marrow Transplantation/methods , Cell Differentiation , Embryonic Stem Cells/cytology , Humans , Induced Pluripotent Stem Cells/cytology
16.
PLoS One ; 7(8): e42386, 2012.
Article in English | MEDLINE | ID: mdl-22870324

ABSTRACT

Understanding the mechanisms by which pathogens induce vascular inflammation and dysfunction may reveal novel therapeutic targets in sepsis and related conditions. The intracellular receptor NOD1 recognises peptidoglycan which features in the cell wall of gram negative and some gram positive bacteria. NOD1 engagement generates an inflammatory response via activation of NFκB and MAPK pathways. We have previously shown that stimulation of NOD1 directly activates blood vessels and causes experimental shock in vivo. In this study we have used an ex vivo vessel-organ culture model to characterise the relative contribution of the endothelium in the response of blood vessels to NOD1 agonists. In addition we present the novel finding that NOD1 directly activates human blood vessels. Using human cultured cells we confirm that endothelial cells respond more avidly to NOD1 agonists than vascular smooth muscle cells. Accordingly we have sought to pharmacologically differentiate NOD1 and TLR4 mediated signalling pathways in human endothelial cells, focussing on TAK1, NFκB and p38 MAPK. In addition we profile novel inhibitors of RIP2 and NOD1 itself, which specifically inhibit NOD1 ligand induced inflammatory signalling in the vasculature. This paper is the first to demonstrate activation of whole human artery by NOD1 stimulation and the relative importance of the endothelium in the sensing of NOD1 ligands by vessels. This data supports the potential utility of NOD1 and RIP2 as therapeutic targets in human disease where vascular inflammation is a clinical feature, such as in sepsis and septic shock.


Subject(s)
Endothelial Cells/immunology , MAP Kinase Signaling System/immunology , Nod1 Signaling Adaptor Protein/immunology , Toll-Like Receptor 4/immunology , Vasculitis/immunology , Animals , Cells, Cultured , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelium, Vascular/immunology , Endothelium, Vascular/pathology , Gram-Negative Bacteria/immunology , Gram-Negative Bacteria/metabolism , Humans , MAP Kinase Kinase Kinases/immunology , MAP Kinase Kinase Kinases/metabolism , Male , Muscle, Smooth, Vascular/immunology , Muscle, Smooth, Vascular/pathology , NF-kappa B/immunology , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/agonists , Nod1 Signaling Adaptor Protein/metabolism , Peptidoglycan/immunology , Peptidoglycan/metabolism , Rats , Rats, Sprague-Dawley , Receptor-Interacting Protein Serine-Threonine Kinase 2/immunology , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Toll-Like Receptor 4/metabolism , Vasculitis/metabolism , Vasculitis/pathology , Vasculitis/therapy , p38 Mitogen-Activated Protein Kinases/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...