Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
2.
Semin Immunol ; 56: 101536, 2021 08.
Article in English | MEDLINE | ID: mdl-34862118

ABSTRACT

Theranostics, literally derived from the combination of the words diagnostics and therapy, is an emerging field of clinical and preclinical research, where contrast agents, drugs and diagnostic techniques are combined to simultaneously diagnose and treat pathologies. Nanoparticles are extensively employed in theranostics due to their potential to target specific organs and their multifunctional capacity. In this review, we will discuss the current state of theranostic nanomedicine, providing key examples of its application in the imaging and treatment of cardiovascular inflammation.


Subject(s)
Nanomedicine , Nanoparticles , Humans , Inflammation , Nanomedicine/methods , Nanoparticles/therapeutic use , Precision Medicine , Theranostic Nanomedicine/methods
3.
Cardiovasc Res ; 117(13): 2589-2609, 2021 11 22.
Article in English | MEDLINE | ID: mdl-34698811

ABSTRACT

Both animal models and human observational and genetic studies have shown that immune and inflammatory mechanisms play a key role in hypertension and its complications. We review the effects of immunomodulatory interventions on blood pressure, target organ damage, and cardiovascular risk in humans. In experimental and small clinical studies, both non-specific immunomodulatory approaches, such as mycophenolate mofetil and methotrexate, and medications targeting T and B lymphocytes, such as tacrolimus, cyclosporine, everolimus, and rituximab, lower blood pressure and reduce organ damage. Mechanistically targeted immune interventions include isolevuglandin scavengers to prevent neo-antigen formation, co-stimulation blockade (abatacept, belatacept), and anti-cytokine therapies (e.g. secukinumab, tocilizumab, canakinumab, TNF-α inhibitors). In many studies, trial designs have been complicated by a lack of blood pressure-related endpoints, inclusion of largely normotensive study populations, polypharmacy, and established comorbidities. Among a wide range of interventions reviewed, TNF-α inhibitors have provided the most robust evidence of blood pressure lowering. Treatment of periodontitis also appears to deliver non-pharmacological anti-hypertensive effects. Evidence of immunomodulatory drugs influencing hypertension-mediated organ damage are also discussed. The reviewed animal models, observational studies, and trial data in humans, support the therapeutic potential of immune-targeted therapies in blood pressure lowering and in hypertension-mediated organ damage. Targeted studies are now needed to address their effects on blood pressure in hypertensive individuals.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Antihypertensive Agents/therapeutic use , Blood Pressure/drug effects , Hypertension/drug therapy , Immunomodulating Agents/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Inflammation/drug therapy , Translational Research, Biomedical , Animals , Humans , Hypertension/genetics , Hypertension/immunology , Hypertension/physiopathology , Immunosuppressive Agents/therapeutic use , Inflammation/genetics , Inflammation/immunology , Inflammation/physiopathology , Inflammation Mediators/immunology , Molecular Targeted Therapy , Signal Transduction , Tumor Necrosis Factor Inhibitors/therapeutic use
4.
Br J Pharmacol ; 178(21): 4216-4245, 2021 11.
Article in English | MEDLINE | ID: mdl-34378206

ABSTRACT

Cardiovascular diseases (CVDs), including atherosclerosis, are chronic inflammatory diseases characterised by a complex and evolving tissue micro-environment. Molecular heterogeneity of inflammatory responses translates into clinical outcomes. However, current medical imaging modalities are unable to reveal the cellular and molecular events at a level of detail that would allow more accurate and timely diagnosis and treatment. This is an inherent limitation of the current imaging tools, which are restricted to anatomical or functional data. Molecular imaging-the visualisation and quantification of molecules in the body-is already established in the clinic in the form of PET, yet the use of PET in CVD is limited. In this visual review, we will guide you through the current state of molecular imaging research, assessing the respective strengths and weaknesses of molecular imaging modalities, including those already being used in the clinic such as PET and MRI and emerging technologies at preclinical stage, such as photoacoustic imaging. We discuss the basic principles of each technology and provide key examples of their application in imaging inflammation in CVD and the added value into the diagnostic decision-making process. Finally, we discuss the barriers to the rapid successful clinical translation of these novel diagnostic modalities. LINKED ARTICLES: This article is part of a themed issue on Molecular imaging - visual themed issue. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v178.21/issuetoc.


Subject(s)
Cardiovascular Diseases , Inflammation , Cardiovascular Diseases/diagnostic imaging , Humans , Inflammation/diagnostic imaging , Molecular Imaging
6.
Pharmacol Ther ; 211: 107550, 2020 07.
Article in English | MEDLINE | ID: mdl-32325067

ABSTRACT

Inflammation is a key factor in multiple diseases including primary immune-mediated inflammatory diseases e.g. rheumatoid arthritis but also, less obviously, in many other common conditions, e.g. cardiovascular disease and diabetes. Together, chronic inflammatory diseases contribute to the majority of global morbidity and mortality. However, our understanding of the underlying processes by which the immune response is activated and sustained is limited by a lack of cellular and molecular information obtained in situ. Molecular imaging is the visualization, detection and quantification of molecules in the body. The ability to reveal information on inflammatory biomarkers, pathways and cells can improve disease diagnosis, guide and monitor therapeutic intervention and identify new targets for research. The optimum molecular imaging modality will possess high sensitivity and high resolution and be capable of non-invasive quantitative imaging of multiple disease biomarkers while maintaining an acceptable safety profile. The mainstays of current clinical imaging are computed tomography (CT), magnetic resonance imaging (MRI), ultrasound (US) and nuclear imaging such as positron emission tomography (PET). However, none of these have yet progressed to routine clinical use in the molecular imaging of inflammation, therefore new approaches are required to meet this goal. This review sets out the respective merits and limitations of both established and emerging imaging modalities as clinically useful molecular imaging tools in addition to potential theranostic applications.


Subject(s)
Inflammation/diagnostic imaging , Molecular Imaging/methods , Animals , Cardiovascular Diseases/diagnostic imaging , Cardiovascular Diseases/physiopathology , Diabetes Mellitus/diagnosis , Diabetes Mellitus/physiopathology , Humans , Inflammation/physiopathology , Inflammation/therapy , Magnetic Resonance Imaging/methods , Tomography, X-Ray Computed/methods , Ultrasonography/methods
7.
Cardiovasc Res ; 116(2): 306-316, 2020 02 01.
Article in English | MEDLINE | ID: mdl-30980670

ABSTRACT

AIMS: Aortic adaptive immunity plays a role in atherosclerosis; however, the precise mechanisms leading to T-cell activation in the arterial wall remain poorly understood. METHODS AND RESULTS: Here, we have identified naïve T cells in the aorta of wild-type and T-cell receptor transgenic mice and we demonstrate that naïve T cells can be primed directly in the vessel wall with both kinetics and frequency of T-cell activation found to be similar to splenic and lymphoid T cells. Aortic homing of naïve T cells is regulated at least in part by the P-selectin glycosylated ligand-1 receptor. In experimental atherosclerosis the aorta supports CD4+ T-cell activation selectively driving Th1 polarization. By contrast, secondary lymphoid organs display Treg expansion. CONCLUSION: Our results demonstrate that the aorta can support T-cell priming and that naïve T cells traffic between the circulation and vessel wall. These data underpin the paradigm that local priming of T cells specific for plaque antigens contributes to atherosclerosis progression.


Subject(s)
Adaptive Immunity , Aorta/immunology , Aortic Diseases/immunology , Atherosclerosis/immunology , Cell Proliferation , Lymphocyte Activation , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Animals , Aorta/metabolism , Aorta/pathology , Aortic Diseases/genetics , Aortic Diseases/metabolism , Aortic Diseases/pathology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cytokines/metabolism , Disease Models, Animal , Disease Progression , Genes, T-Cell Receptor , Kinetics , Membrane Glycoproteins/metabolism , Mice, Inbred C57BL , Mice, Knockout, ApoE , Phenotype , Plaque, Atherosclerotic , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/metabolism
8.
Front Immunol ; 10: 1979, 2019.
Article in English | MEDLINE | ID: mdl-31552015

ABSTRACT

Abdominal Aortic Aneurysm (AAA) is a major cause of cardiovascular mortality. Adverse changes in vascular phenotype act in concert with chronic inflammation to promote AAA progression. Perivascular adipose tissue (PVAT) helps maintain vascular homeostasis but when inflamed and dysfunctional, can also promote vascular pathology. Previous studies suggested that PVAT may be an important site of vascular inflammation in AAA; however, a detailed assessment of leukocyte populations in human AAA, their anatomic location in the vessel wall and correlation to AAA size remain undefined. Accordingly, we performed in depth immunophenotyping of cells infiltrating the pathologically altered perivascular tissue (PVT) and vessel wall in AAA samples at the site of maximal dilatation (n = 51 patients). Flow cytometry revealed that T cells, rather than macrophages, are the major leukocyte subset in AAA and that their greatest accumulations occur in PVT. Both CD4+ and CD8+ T cell populations are highly activated in both compartments, with CD4+ T cells displaying the highest activation status within the AAA wall. Finally, we observed a positive relationship between T cell infiltration in PVT and AAA wall. Interestingly, only PVT T cell infiltration was strongly related to tertiles of AAA size. In summary, this study highlights an important role for PVT as a reservoir of T lymphocytes and potentially as a key site in modulating the underlying inflammation in AAA.


Subject(s)
Adipose Tissue/immunology , Aorta, Abdominal/immunology , Aortic Aneurysm, Abdominal/immunology , Inflammation/immunology , T-Lymphocytes/immunology , Adipose Tissue/metabolism , Aged , Aorta, Abdominal/metabolism , Aorta, Abdominal/pathology , Aortic Aneurysm, Abdominal/metabolism , Female , Flow Cytometry , Humans , Immunophenotyping , Inflammation/metabolism , Leukocytes/immunology , Leukocytes/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , Middle Aged , Severity of Illness Index , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
9.
Front Immunol ; 10: 849, 2019.
Article in English | MEDLINE | ID: mdl-31068936

ABSTRACT

Atherosclerosis is a complex inflammatory pathology underpinning cardiovascular diseases (CVD), which are the leading cause of death worldwide. The interplay between vascular stromal cells and immune cells is fundamental to the progression and outcome of atherosclerotic disease, however, the majority of in vitro studies do not consider the implications of these interactions and predominantly use mono-culture approaches. Here we present a simple and robust methodology involving the co-culture of vascular endothelial (ECs) and smooth muscle cells (SMCs) alongside an inflammatory compartment, in our study containing THP-1 macrophages, for studying these complex interactions. Using this approach, we demonstrate that the interaction between vascular stromal and immune cells produces unique cellular phenotypes and soluble mediator profiles not observed in double-cell 2D cultures. Our results highlight the importance of cellular communication and support the growing idea that in vitro research must evolve from mono-culture systems to provide data more representative of the multi-cellular environment found in vivo. The methodology presented, in comparison with established approaches, has the advantage of being technically simple whilst enabling the isolation of pure populations of ECs, SMCs and immune cells directly from the co-culture without cell sorting. The approach described within would be applicable to those studying mechanisms of vascular inflammation, particularly in relation to understanding the impact cellular interaction has on the cumulative immune-vascular response to atherogenic or inflammatory stimuli.


Subject(s)
Atherosclerosis/etiology , Atherosclerosis/pathology , Cell Communication , Cell Culture Techniques , Coculture Techniques , Models, Biological , Biomarkers , Endothelial Cells/immunology , Endothelial Cells/metabolism , Fluorescent Antibody Technique , Humans , Macrophages/immunology , Macrophages/metabolism , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle/immunology , Myocytes, Smooth Muscle/metabolism
11.
Cardiovasc Res ; 114(13): 1714-1727, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30165574

ABSTRACT

Cardiovascular diseases (CVD) account for nearly half of all deaths in Europe and almost 30% of global deaths. Despite the improved clinical management, cardiovascular mortality is predicted to rise in the next decades due to the increasing impact of aging, obesity, and diabetes. The goal of emerging cardiovascular nanomedicine is to reduce the burden of CVD using nanoscale medical products and devices. However, the development of novel multicomponent nano-sized products poses multiple technical, ethical, and regulatory challenges, which often obstruct their road to successful approval and use in clinical practice. This review discusses the rational design of nanoparticles, including safety considerations and regulatory issues, and highlights the steps needed to achieve efficient clinical translation of promising nanomedicinal products for cardiovascular applications.


Subject(s)
Cardiology/standards , Cardiovascular Diseases/therapy , Nanomedicine/standards , Practice Guidelines as Topic/standards , Translational Research, Biomedical/standards , Animals , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/mortality , Disease Models, Animal , Humans , Patient Safety , Risk Assessment , Toxicity Tests/standards
12.
Clin Sci (Lond) ; 132(14): 1509-1512, 2018 07 31.
Article in English | MEDLINE | ID: mdl-30037838

ABSTRACT

The hallmark features of atherosclerosis include accumulation of low-density lipoprotein (LDL) carrying cholesterol in the vessel wall, formation of lipid-laden foam cells, and the creation of a pro-inflammatory microenvironment. To date, no effective treatments are clinically available for increasing cholesterol efflux from vascular macrophages and inducing reverse cholesterol transport (RCT). In an article published recently in Clinical Science (vol 132, issue 6, 1199-1213), Zhang and colleagues identified the extracellular matrix protein mindin/spondin 2 as a positive regulator of atherosclerosis. Genetic knockout of mindin in apolipoprotein-E (apoE)-/- mice attenuated atherosclerosis, foam cell formation, and inflammation within the vessel wall. Conversely, selective overexpression of mindin in macrophages in apoE-/- mice was sufficient to promote the greater severity of atherosclerosis. Interestingly, foam cell formation was closely associated with the expression of cholesterol transporters (ABCA1 and ACBG1) that facilitate cholesterol efflux. Liver X receptor (LXR)-ß is a key modulator of cholesterol transporter expression and formed direct interactions with mindin. Furthermore, the protective effects of mindin deficiency on foam cell formation were blocked by inhibition of LXR-ß. This article highlights a novel role of mindin in modulating foam cell formation and atherosclerosis development in mice through direct regulation of LXR-ß. Thus far, direct targetting of LXR-ß via pharmacological agonists has proven to be problematic due to the lack of subtype selective inhibitors and associated adverse effects. Indirect targetting of LXR-ß, therefore, via mindin inhibition offers a new therapeutic strategy for increasing LXR-ß induced cholesterol efflux, reducing foam cell formation, and preventing or treating atherosclerosis.


Subject(s)
Atherosclerosis , Foam Cells , ATP Binding Cassette Transporter 1 , Animals , Apolipoproteins E , Extracellular Matrix Proteins , Liver X Receptors , Macrophages , Mice
13.
Theranostics ; 8(22): 6195-6209, 2018.
Article in English | MEDLINE | ID: mdl-30613292

ABSTRACT

Vascular immune-inflammatory responses play a crucial role in the progression and outcome of atherosclerosis. The ability to assess localized inflammation through detection of specific vascular inflammatory biomarkers would significantly improve cardiovascular risk assessment and management; however, no multi-parameter molecular imaging technologies have been established to date. Here, we report the targeted in vivo imaging of multiple vascular biomarkers using antibody-functionalized nanoparticles and surface-enhanced Raman scattering (SERS). Methods: A series of antibody-functionalized gold nanoprobes (BFNP) were designed containing unique Raman signals in order to detect intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1) and P-selectin using SERS. Results: SERS and BFNP were utilized to detect, discriminate and quantify ICAM-1, VCAM-1 and P-selectin in vitro on human endothelial cells and ex vivo in human coronary arteries. Ultimately, non-invasive multiplex imaging of adhesion molecules in a humanized mouse model was demonstrated in vivo following intravenous injection of the nanoprobes. Conclusion: This study demonstrates that multiplexed SERS-based molecular imaging can indicate the status of vascular inflammation in vivo and gives promise for SERS as a clinical imaging technique for cardiovascular disease in the future.


Subject(s)
Coronary Vessels/diagnostic imaging , Coronary Vessels/immunology , Human Umbilical Vein Endothelial Cells/chemistry , Molecular Imaging/methods , Spectrum Analysis, Raman/methods , Animals , Female , Gold/chemistry , Human Umbilical Vein Endothelial Cells/immunology , Humans , Intercellular Adhesion Molecule-1/genetics , Intercellular Adhesion Molecule-1/immunology , Male , Mice , Mice, Inbred NOD , Mice, SCID , Molecular Imaging/instrumentation , Nanoparticles/chemistry , P-Selectin/genetics , P-Selectin/immunology , Vascular Cell Adhesion Molecule-1/genetics , Vascular Cell Adhesion Molecule-1/immunology
14.
Heart ; 104(6): 460-467, 2018 03.
Article in English | MEDLINE | ID: mdl-29061690

ABSTRACT

To accurately predict atherosclerotic plaque progression, a detailed phenotype of the lesion at the molecular level is required. Here, we assess the respective merits and limitations of molecular imaging tools. Clinical imaging includes contrast-enhanced ultrasound, an inexpensive and non-toxic technique but with poor sensitivity. CT benefits from high spatial resolution but poor sensitivity coupled with an increasing radiation burden that limits multiplexing. Despite high sensitivity, positron emission tomography and single-photon emission tomography have disadvantages when applied to multiplex molecular imaging due to poor spatial resolution, signal cross talk and increasing radiation dose. In contrast, MRI is non-toxic, displays good spatial resolution but poor sensitivity. Preclinical techniques include near-infrared fluorescence (NIRF), which provides good spatial resolution and sensitivity; however, multiplexing with NIRF is limited, due to photobleaching and spectral overlap. Fourier transform infrared spectroscopy and Raman spectroscopy are label-free techniques that detect molecules based on the vibrations of chemical bonds. Both techniques offer fast acquisition times with Raman showing superior spatial resolution. Raman signals are inherently weak; however, leading to the development of surface-enhanced Raman spectroscopy (SERS) that offers greatly increased sensitivity due to using metallic nanoparticles that can be functionalised with biomolecules targeted against plaque ligands while offering high multiplexing potential. This asset combined with high spatial resolution makes SERS an exciting prospect as a diagnostic tool. The ongoing refinements of SERS technologies such as deep tissue imaging and portable systems making SERS a realistic prospect for translation to the clinic.


Subject(s)
Cardiovascular Diseases/diagnosis , Plaque, Atherosclerotic/diagnostic imaging , Spectrum Analysis, Raman/methods , Disease Progression , Humans , Molecular Imaging/methods
15.
Br J Pharmacol ; 174(2): 210-222, 2017 01.
Article in English | MEDLINE | ID: mdl-27864936

ABSTRACT

BACKGROUND AND PURPOSE: The sphingosine analogue, FTY720 (GilenyaR ), alleviates clinical disease progression in multiple sclerosis. Here, we variously assessed the effects of an azide analogue of (S)-FTY720 vinylphosphonate (compound 5; a sphingosine kinase 1 activator), (R)-FTY720 methyl ether (ROMe, a sphingosine kinase 2 inhibitor) and RB-020 (a sphingosine kinase 1 inhibitor and sphingosine kinase 2 substrate) on IL-1ß formation, sphingosine 1-phosphate levels and expression of S1P1 receptors. We also assessed the effect of compound 5 and ROMe in an experimental autoimmune encephalomyelitis (EAE) model in mice. EXPERIMENTAL APPROACH: We measured IL-1ß formation by macrophages, sphingosine 1-phosphate levels and expression levels of S1P1 receptors in vitro and clinical score in mice with EAE and the extent of inflammatory cell infiltration into the spinal cord in vivo. KEY RESULTS: Treatment of differentiated U937 macrophages with compound 5, RB-020 or sphingosine (but not ROMe) enhanced IL-1ß release. These data suggest that these compounds might be pro-inflammatory in vitro. However, compound 5 or ROMe reduced disease progression and infiltration of inflammatory cells into the spinal cord in EAE, and ROMe induced a reduction in CD4+ and CD8+ T-cell levels in the blood (lymphopenia). Indeed, ROMe induced a marked decrease in expression of cell surface S1P1 receptors in vitro. CONCLUSION AND IMPLICATIONS: This is the first demonstration that an activator of sphingosine kinase 1 (compound 5) and an inhibitor of sphingosine kinase 2 (ROMe, which also reduces cell surface S1P1 receptor expression) have an anti-inflammatory action in EAE.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Interleukin-1beta/metabolism , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Piperidines/pharmacology , Receptors, Lysosphingolipid/biosynthesis , Sphingosine/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Cells, Cultured , Cricetulus , Dose-Response Relationship, Drug , Encephalomyelitis, Autoimmune, Experimental/metabolism , Humans , Mice , Mice, Inbred C57BL , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Piperidines/chemistry , Sphingosine/chemistry , Sphingosine-1-Phosphate Receptors , Structure-Activity Relationship
16.
Cell Signal ; 28(8): 946-55, 2016 08.
Article in English | MEDLINE | ID: mdl-27063355

ABSTRACT

Recent studies have demonstrated that the expression of sphingosine kinase 1, the enzyme that catalyses formation of the bioactive lipid, sphingosine 1-phosphate, is increased in lungs from patients with pulmonary arterial hypertension. In addition, Sk1(-/-) mice are protected from hypoxic-induced pulmonary arterial hypertension. Therefore, we assessed the effect of the sphingosine kinase 1 selective inhibitor, PF-543 and a sphingosine kinase 1/ceramide synthase inhibitor, RB-005 on pulmonary and cardiac remodelling in a mouse hypoxic model of pulmonary arterial hypertension. Administration of the potent sphingosine kinase 1 inhibitor, PF-543 in a mouse hypoxic model of pulmonary hypertension had no effect on vascular remodelling but reduced right ventricular hypertrophy. The latter was associated with a significant reduction in cardiomyocyte death. The protection involves a reduction in the expression of p53 (that promotes cardiomyocyte death) and an increase in the expression of anti-oxidant nuclear factor (erythroid-derived 2)-like 2 (Nrf-2). In contrast, RB-005 lacked effects on right ventricular hypertrophy, suggesting that sphingosine kinase 1 inhibition might be nullified by concurrent inhibition of ceramide synthase. Therefore, our findings with PF-543 suggest an important role for sphingosine kinase 1 in the development of hypertrophy in pulmonary arterial hypertension.


Subject(s)
Enzyme Inhibitors/pharmacology , Hypertension, Pulmonary/physiopathology , Hypoxia/physiopathology , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Pyrrolidines/pharmacology , Sulfones/pharmacology , Ventricular Remodeling/drug effects , Animals , Biomarkers/metabolism , Body Weight/drug effects , Cells, Cultured , Disease Models, Animal , Female , HEK293 Cells , Heart Ventricles/drug effects , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Humans , Hypertension, Pulmonary/blood , Hypertrophy, Right Ventricular/pathology , Hypertrophy, Right Ventricular/physiopathology , Hypoxia/blood , Methanol , Mice, Inbred C57BL , Models, Biological , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Piperidines/blood , Piperidines/chemistry , Piperidines/pharmacology , Pressure , Pulmonary Artery/drug effects , Pulmonary Artery/enzymology , Pulmonary Artery/pathology , Pyrrolidines/blood , Pyrrolidines/chemistry , Signal Transduction/drug effects , Sulfones/blood , Sulfones/chemistry
17.
Adv Biol Regul ; 60: 151-159, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26429117

ABSTRACT

Sphingosine kinase (there are two isoforms, SK1 and SK2) catalyses the formation of sphingosine 1-phosphate (S1P), a bioactive lipid that can be released from cells to activate a family of G protein-coupled receptors, termed S1P1-5. In addition, S1P can bind to intracellular target proteins, such as HDAC1/2, to induce cell responses. There is increasing evidence of a role for S1P receptors (e.g. S1P4) and SK1 in cancer, where high expression of these proteins in ER negative breast cancer patient tumours is linked with poor prognosis. Indeed, evidence will be presented here to demonstrate that S1P4 is functionally linked with SK1 and the oncogene HER2 (ErbB2) to regulate mitogen-activated protein kinase pathways and growth of breast cancer cells. Although much emphasis is placed on SK1 in terms of involvement in oncogenesis, evidence will also be presented for a role of SK2 in both T-cell and B-cell acute lymphoblastic leukemia. In patient T-ALL lymphoblasts and T-ALL cell lines, we have demonstrated that SK2 inhibitors promote T-ALL cell death via autophagy and induce suppression of c-myc and PI3K/AKT pathways. We will also present evidence demonstrating that certain SK inhibitors promote oxidative stress and protein turnover via proteasomal degradative pathways linked with induction of p53-and p21-induced growth arrest. In addition, the SK1 inhibitor, PF-543 exacerbates disease progression in an experimental autoimmune encephalomyelitis mouse model indicating that SK1 functions in an anti-inflammatory manner. Indeed, sphingosine, which accumulates upon inhibition of SK1 activity, and sphingosine-like compounds promote activation of the inflammasome, which is linked with multiple sclerosis, to stimulate formation of the pro-inflammatory mediator, IL-1ß. Such compounds could be exploited to produce antagonists that diminish exaggerated inflammation in disease. The therapeutic potential of modifying the SK-S1P receptor pathway in cancer and inflammation will therefore, be reviewed.


Subject(s)
Inflammation/enzymology , Neoplasms/enzymology , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Receptors, Lysosphingolipid/metabolism , Sphingosine/metabolism , Animals , Humans , Inflammation/genetics , Inflammation/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Phosphotransferases (Alcohol Group Acceptor)/genetics , Receptors, Lysosphingolipid/genetics
18.
PeerJ ; 3: e1192, 2015.
Article in English | MEDLINE | ID: mdl-26312183

ABSTRACT

Objective. Emerging evidence suggests an important role for mast cells in vein graft failure. This study addressed the hypothesis that perivascular mast cells regulate in situ vascular inflammatory and proliferative responses and subsequent vein graft neointimal lesion formation, using an optimized local mast cell reconstitution method. Methods and Results. Neointimal hyperplasia was induced by insertion of a vein graft into the right carotid artery in wild type and mast cell deficient Kit(W-sh/W-sh) mice. In some experiments, mast cells were reconstituted systemically (tail vein injection of bone marrow-derived mast cells) or locally (directly into the right neck area) prior to vein grafting. Vein graft neointimal lesion formation was significantly (P < 0.05) reduced in Kit(W-sh/W-sh) mice. Mast cell deficiency reduced the number of proliferating cells, and inhibited L-selectin, CCL2, M-CSF and MIP-3α expression in the vein grafts. Local but not systemic mast cell reconstitution restored a perivascular mast cell population that subsequently promoted neointimal formation in mast cell deficient mice. Conclusion. Our data demonstrate that perivascular mast cells play a key role in promoting neointima formation by inducing local acute inflammatory and proliferative responses. These results suggest that ex vivo intraoperative targeting of mast cells may have therapeutic potential for the prevention of pathological vein graft remodeling.

19.
Immunity ; 42(6): 1100-15, 2015 Jun 16.
Article in English | MEDLINE | ID: mdl-26084025

ABSTRACT

Tertiary lymphoid organs (TLOs) emerge during nonresolving peripheral inflammation, but their impact on disease progression remains unknown. We have found in aged Apoe(-/-) mice that artery TLOs (ATLOs) controlled highly territorialized aorta T cell responses. ATLOs promoted T cell recruitment, primed CD4(+) T cells, generated CD4(+), CD8(+), T regulatory (Treg) effector and central memory cells, converted naive CD4(+) T cells into induced Treg cells, and presented antigen by an unusual set of dendritic cells and B cells. Meanwhile, vascular smooth muscle cell lymphotoxin ß receptors (VSMC-LTßRs) protected against atherosclerosis by maintaining structure, cellularity, and size of ATLOs though VSMC-LTßRs did not affect secondary lymphoid organs: Atherosclerosis was markedly exacerbated in Apoe(-/-)Ltbr(-/-) and to a similar extent in aged Apoe(-/-)Ltbr(fl/fl)Tagln-cre mice. These data support the conclusion that the immune system employs ATLOs to organize aorta T cell homeostasis during aging and that VSMC-LTßRs participate in atherosclerosis protection via ATLOs.


Subject(s)
Aging/immunology , Atherosclerosis/immunology , Lymphotoxin beta Receptor/metabolism , Myocytes, Smooth Muscle/physiology , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Adventitia/immunology , Aging/genetics , Animals , Aorta/pathology , Apolipoproteins E/genetics , Atherosclerosis/genetics , Cell Differentiation/genetics , Cell Movement/genetics , Cells, Cultured , Choristoma/immunology , Immunologic Memory , Lymphocyte Activation/genetics , Lymphoid Tissue/immunology , Lymphotoxin beta Receptor/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Muscle Proteins/genetics
20.
Biomed Res Int ; 2014: 949845, 2014.
Article in English | MEDLINE | ID: mdl-25136640

ABSTRACT

In the present study aortic murine smooth muscle cell (SMC) antigen presentation capacity was evaluated using the Eα-GFP/Y-Ae system to visualize antigen uptake through a GFP tag and tracking of Eα peptide/MHCII presentation using the Y-Ae Ab. Stimulation with IFN-γ (100 ng/mL) for 72 h caused a significant (P < 0.01) increase in the percentage of MHC class II positive SMCs, compared with unstimulated cells. Treatment with Eα-GFP (100 µg/mL) for 48 h induced a significant (P < 0.05) increase in the percentage of GFP positive SMCs while it did not affect the percentage of Y-Ae positive cells, being indicative of antigen uptake without its presentation in the context of MHC class II. After IFN-γ-stimulation, ovalbumin- (OVA, 1 mg/mL) or OVA323-339 peptide-(0.5 µg/mL) treated SMCs failed to induce OT-II CD4(+) T cell activation/proliferation; this was also accompanied by a lack of expression of key costimulatory molecules (OX40L, CD40, CD70, and CD86) on SMCs. Finally, OVA-treated SMCs failed to induce DO11.10-GFP hybridoma activation, a process independent of costimulation. Our results demonstrate that while murine primary aortic SMCs express MHC class II and can acquire exogenous antigens, they fail to activate T cells through a failure in antigen presentation and a lack of costimulatory molecule expression.


Subject(s)
Antigen Presentation , Aorta/immunology , Histocompatibility Antigens Class II/immunology , Myocytes, Smooth Muscle/immunology , Animals , Aorta/cytology , Flow Cytometry , Histocompatibility Antigens Class I/immunology , Lymphocyte Activation/immunology , Mice , Peptides/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL