Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 42
Filter
1.
Int J Mol Sci ; 24(24)2023 Dec 14.
Article in English | MEDLINE | ID: mdl-38139295

ABSTRACT

This review will provide an overview of what is currently known about mechanisms linking poor glycaemic control with increased thrombotic risk. The leading causes of death in people with diabetes are strokes and cardiovascular disease. Significant morbidity is associated with an increased risk of thrombosis, resulting in myocardial infarction, ischaemic stroke, and peripheral vascular disease, along with the sequelae of these events, including loss of functional ability, heart failure, and amputations. While the increased platelet activity, pro-coagulability, and endothelial dysfunction directly impact this risk, the molecular mechanisms linking poor glycaemic control with increased thrombotic risk remain unclear. This review highlights the complex mechanisms underlying thrombosis prevalence in individuals with diabetes and hyperglycaemia. Post-translational modifications, such as O-GlcNAcylation, play a crucial role in controlling protein function in diabetes. However, the role of O-GlcNAcylation remains poorly understood due to its intricate regulation and the potential involvement of multiple variables. Further research is needed to determine the precise impact of O-GlcNAcylation on specific disease processes.


Subject(s)
Brain Ischemia , Diabetes Mellitus , Hyperglycemia , Myocardial Infarction , Stroke , Thrombosis , Humans , Brain Ischemia/complications , Stroke/etiology , Thrombosis/complications , Myocardial Infarction/complications , Hyperglycemia/complications
2.
PLoS One ; 18(2): e0280594, 2023.
Article in English | MEDLINE | ID: mdl-36724150

ABSTRACT

Microplastics (MPs) are ubiquitous in the environment, in the human food chain, and have been recently detected in blood and lung tissues. To undertake a pilot analysis of MP contamination in human vein tissue samples with respect to their presence (if any), levels, and characteristics of any particles identified. This study analysed digested human saphenous vein tissue samples (n = 5) using µFTIR spectroscopy (size limitation of 5 µm) to detect and characterise any MPs present. In total, 20 MP particles consisting of five MP polymer types were identified within 4 of the 5 vein tissue samples with an unadjusted average of 29.28 ± 34.88 MP/g of tissue (expressed as 14.99 ± 17.18 MP/g after background subtraction adjustments). Of the MPs detected in vein samples, five polymer types were identified, of irregular shape (90%), with alkyd resin (45%), poly (vinyl propionate/acetate, PVAc (20%) and nylon-ethylene-vinyl acetate, nylon-EVA, tie layer (20%) the most abundant. While the MP levels within tissue samples were not significantly different than those identified within procedural blanks (which represent airborne contamination at time of sampling), they were comprised of different plastic polymer types. The blanks comprised n = 13 MP particles of four MP polymer types with the most abundant being polytetrafluoroethylene (PTFE), then polypropylene (PP), polyethylene terephthalate (PET) and polyfumaronitrile:styrene (FNS), with a mean ± SD of 10.4 ± 9.21, p = 0.293. This study reports the highest level of contamination control and reports unadjusted values alongside different contamination adjustment techniques. This is the first evidence of MP contamination of human vascular tissues. These results support the phenomenon of transport of MPs within human tissues, specifically blood vessels, and this characterisation of types and levels can now inform realistic conditions for laboratory exposure experiments, with the aim of determining vascular health impacts.


Subject(s)
Microplastics , Water Pollutants, Chemical , Humans , Microplastics/analysis , Plastics/analysis , Pilot Projects , Nylons , Saphenous Vein , Water Pollutants, Chemical/analysis , Environmental Monitoring , Polymers
3.
Cells ; 11(4)2022 02 17.
Article in English | MEDLINE | ID: mdl-35203353

ABSTRACT

Unresolved hyperglycaemia, a hallmark of type 2 diabetes mellitus (T2DM), is a well characterised manifestation of altered fuel homeostasis and our understanding of its role in the pathologic activation of the inflammatory system continues to grow. Metabolic disorders like T2DM trigger changes in the regulation of key cellular processes such as cell trafficking and proliferation, and manifest as chronic inflammatory disorders with severe long-term consequences. Activation of inflammatory pathways has recently emerged as a critical link between T2DM and inflammation. A substantial body of evidence has suggested that this is due in part to increased flux through the hexosamine biosynthetic pathway (HBP). The HBP, a unique nutrient-sensing metabolic pathway, produces the activated amino sugar UDP-GlcNAc which is a critical substrate for protein O-GlcNAcylation, a dynamic, reversible post-translational glycosylation of serine and threonine residues in target proteins. Protein O-GlcNAcylation impacts a range of cellular processes, including inflammation, metabolism, trafficking, and cytoskeletal organisation. As increased HBP flux culminates in increased protein O-GlcNAcylation, we propose that targeting O-GlcNAcylation may be a viable therapeutic strategy for the prevention and management of glucose-dependent pathologies with inflammatory components.


Subject(s)
Diabetes Mellitus, Type 2 , Glycosylation , Hexosamines/metabolism , Humans , Inflammation , Protein Processing, Post-Translational , Proteins/metabolism
4.
Clin Sci (Lond) ; 135(13): 1563-1590, 2021 07 16.
Article in English | MEDLINE | ID: mdl-34231841

ABSTRACT

Despite obesity and diabetes markedly increasing the risk of developing cardiovascular diseases, the molecular and cellular mechanisms that underlie this association remain poorly characterised. In the last 20 years it has become apparent that chronic, low-grade inflammation in obese adipose tissue may contribute to the risk of developing insulin resistance and type 2 diabetes. Furthermore, increased vascular pro-inflammatory signalling is a key event in the development of cardiovascular diseases. Overnutrition exacerbates pro-inflammatory signalling in vascular and adipose tissues, with several mechanisms proposed to mediate this. In this article, we review the molecular and cellular mechanisms by which nutrients are proposed to regulate pro-inflammatory signalling in adipose and vascular tissues. In addition, we examine the potential therapeutic opportunities that these mechanisms provide for suppression of inappropriate inflammation in obesity and vascular disease.


Subject(s)
Adipose Tissue/metabolism , Cardiovascular Diseases/metabolism , Cardiovascular System/metabolism , Energy Metabolism , Inflammation Mediators/metabolism , Inflammation/metabolism , Nutritional Status , Obesity/metabolism , Adipokines/metabolism , Adipose Tissue/drug effects , Adipose Tissue/immunology , Adipose Tissue/physiopathology , Animals , Anti-Inflammatory Agents/therapeutic use , Cardiovascular Diseases/immunology , Cardiovascular Diseases/physiopathology , Cardiovascular Diseases/prevention & control , Cardiovascular System/drug effects , Cardiovascular System/immunology , Cardiovascular System/physiopathology , Energy Metabolism/drug effects , Humans , Hypoglycemic Agents/therapeutic use , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology , Inflammation Mediators/antagonists & inhibitors , Obesity/drug therapy , Obesity/immunology , Obesity/physiopathology , Oxidative Stress , Signal Transduction
5.
Nutr Metab Cardiovasc Dis ; 31(5): 1349-1356, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33812732

ABSTRACT

AIM: Coronary artery bypass graft (CABG) using autologous saphenous vein continues to be a gold standard procedure to restore the supply of oxygen-rich blood to the heart muscles in coronary artery disease (CAD) patients with or without type 2 diabetes mellitus (T2DM). However, CAD patients with T2DM are at higher risk of graft failure. While failure rates have been reduced through improvements in procedure-related factors, much less is known about the molecular and cellular mechanisms by which T2DM initiates vein graft failure. This review gives novel insights into these cellular and molecular mechanisms and identifies potential therapeutic targets for development of new medicines to improve vein graft patency. DATA SYNTHESIS: One important cellular process that has been implicated in the pathogenesis of T2DM is protein O-GlcNAcylation, a dynamic, reversible post-translational modification of serine and threonine residues on target proteins that is controlled by two enzymes: O-GlcNAc transferase (OGT) and O-GlcNAcase (OGA). Protein O-GlcNAcylation impacts a range of cellular processes, including trafficking, metabolism, inflammation and cytoskeletal organisation. Altered O-GlcNAcylation homeostasis have, therefore, been linked to a range of human pathologies with a metabolic component, including T2DM. CONCLUSION: We propose that protein O-GlcNAcylation alters vascular smooth muscle and endothelial cell function through modification of specific protein targets which contribute to the vascular re-modelling responsible for saphenous vein graft failure in T2DM.


Subject(s)
Blood Glucose/metabolism , Coronary Artery Bypass , Coronary Artery Disease/surgery , Diabetes Mellitus, Type 2/complications , Graft Occlusion, Vascular/etiology , Protein Processing, Post-Translational , Saphenous Vein/transplantation , Animals , Biomarkers/blood , Coronary Artery Bypass/adverse effects , Coronary Artery Disease/diagnosis , Coronary Artery Disease/etiology , Coronary Artery Disease/metabolism , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/drug therapy , Glycosylation , Graft Occlusion, Vascular/metabolism , Graft Occlusion, Vascular/pathology , Graft Occlusion, Vascular/prevention & control , Humans , Protein Processing, Post-Translational/drug effects , Risk Assessment , Risk Factors , Saphenous Vein/metabolism , Saphenous Vein/pathology , Treatment Failure , Vascular Remodeling
6.
Pharmacol Res ; 165: 105467, 2021 03.
Article in English | MEDLINE | ID: mdl-33515704

ABSTRACT

Cardiovascular diseases (CVDs) are the leading cause of death globally. While the major focus of pharmacological and non-pharmacological interventions has been on targeting disease pathophysiology and limiting predisposing factors, our understanding of the cellular and molecular mechanisms underlying the pathogenesis of CVDs remains incomplete. One mechanism that has recently emerged is protein O-GlcNAcylation. This is a dynamic, site-specific reversible post-translational modification of serine and threonine residues on target proteins and is controlled by two enzymes: O-linked ß-N-acetylglucosamine transferase (OGT) and O-linked ß-N-acetylglucosaminidase (OGA). Protein O-GlcNAcylation alters the cellular functions of these target proteins which play vital roles in pathways that modulate vascular homeostasis and cardiac function. Through this review, we aim to give insights on the role of protein O-GlcNAcylation in cardiovascular diseases and identify potential therapeutic targets in this pathway for development of more effective medicines to improve patient outcomes.


Subject(s)
Cardiovascular Agents/administration & dosage , Cardiovascular Diseases/drug therapy , Drug Delivery Systems/methods , Enzyme Inhibitors/administration & dosage , Protein Processing, Post-Translational/drug effects , Acetylglucosamine/antagonists & inhibitors , Acetylglucosamine/metabolism , Acetylglucosaminidase/antagonists & inhibitors , Acetylglucosaminidase/metabolism , Acylation/drug effects , Acylation/physiology , Animals , Antigens, Neoplasm/metabolism , Cardiovascular Diseases/metabolism , Glycosylation/drug effects , Histone Acetyltransferases/antagonists & inhibitors , Histone Acetyltransferases/metabolism , Humans , Hyaluronoglucosaminidase/antagonists & inhibitors , Hyaluronoglucosaminidase/metabolism , N-Acetylglucosaminyltransferases/antagonists & inhibitors , N-Acetylglucosaminyltransferases/metabolism , Protein Processing, Post-Translational/physiology , beta-N-Acetylhexosaminidases/antagonists & inhibitors , beta-N-Acetylhexosaminidases/metabolism
7.
Methods Mol Biol ; 2169: 105-118, 2020.
Article in English | MEDLINE | ID: mdl-32548823

ABSTRACT

The ability of inducible regulator suppressor of cytokine signaling 3 (SOCS3) to inhibit Janus kinase-signal transducer and activator of transcription (JAK-STAT) signaling requires interaction with specific cytokine receptors, JAKs, and components of the cellular ubiquitylation machinery. However, it is now clear that additional protein interactions are essential for effective inhibition of JAK-STAT signaling that have also identified new roles for SOCS3. For example, we have demonstrated that SOCS3 interaction with cavin-1, a core component of caveolae essential for their formation, is required for effective inhibition of interleukin (IL)-6 signaling and maintenance of cellular levels of caveolae. This is achieved through cavin-1 interaction with a discrete motif within the SOCS3 SH2 domain. Here, we describe in detail three methods (coimmunoprecipitation; peptide pull-down; peptide array overlay) we have used to validate and characterize cavin-1/SOCS3 interactions in vitro.


Subject(s)
Immunoprecipitation/methods , Protein Array Analysis/methods , RNA-Binding Proteins/metabolism , Suppressor of Cytokine Signaling Proteins/metabolism , Biotin/chemistry , Caveolae/metabolism , HEK293 Cells , Humans , Janus Kinases/metabolism , Peptides/genetics , Peptides/metabolism , Protein Binding , RNA-Binding Proteins/genetics , STAT Transcription Factors/metabolism , Signal Transduction/genetics , Suppressor of Cytokine Signaling 3 Protein/genetics , Suppressor of Cytokine Signaling 3 Protein/metabolism , Suppressor of Cytokine Signaling Proteins/genetics
8.
Biochem Soc Trans ; 47(4): 1143-1156, 2019 08 30.
Article in English | MEDLINE | ID: mdl-31341036

ABSTRACT

Inflammation has been highlighted as a key factor in pulmonary arterial hypertension (PAH) development, particularly interleukin-6 (IL-6). IL-6 activates JAK-STAT signalling to induce transcription of pro-inflammatory and pro-angiogenic genes, enabling PAH progression, as well as the transcription of suppressor of cytokine signalling 3 (SOCS3) which limits IL-6 signalling. Current PAH therapies include prostanoid drugs which induce vasodilation via stimulating intracellular 3',5'-cyclic adenosine monophosphate (cAMP) levels. cAMP can also inhibit IL-6-mediated endothelial dysfunction via the induction of SOCS3. Thus, we propose that an important mechanism by which cAMP-mobilising prostanoid drugs limit PAH is by inhibiting IL-6-mediated pulmonary inflammation and remodelling via SOCS3 inhibition of IL-6 signalling. Further clarification may result in effective strategies with which to target the IL-6/JAK-STAT signalling pathway in PAH.


Subject(s)
Interleukin-6/antagonists & inhibitors , Interleukin-6/metabolism , Prostaglandins/pharmacology , Pulmonary Arterial Hypertension/therapy , Signal Transduction/drug effects , Animals , Disease Models, Animal , Humans , Pulmonary Arterial Hypertension/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism
9.
Trends Pharmacol Sci ; 40(5): 298-308, 2019 05.
Article in English | MEDLINE | ID: mdl-30948191

ABSTRACT

Defective regulation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling pathway in cancers, haematological diseases, and chronic inflammatory conditions highlights its clinical significance. While several biologic and small molecule therapeutics targeting this pathway have been developed, these have several limitations. Therefore, there is a need to identify new targets for intervention. Suppressor of cytokine signalling (SOCS) proteins are a family of inducible inhibitors of cytokine receptors that activate the JAK-STAT pathway. Here we propose that newly identified mechanisms controlling SOCS function could be exploited to develop molecularly targeted drugs with unique modes of action to inhibit JAK-STAT signalling in disease.


Subject(s)
Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Signal Transduction/drug effects , Suppressor of Cytokine Signaling Proteins/metabolism , Animals , Humans , Janus Kinases/antagonists & inhibitors , Molecular Targeted Therapy , STAT Transcription Factors/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Suppressor of Cytokine Signaling Proteins/antagonists & inhibitors
10.
Ann Rheum Dis ; 78(7): 929-933, 2019 07.
Article in English | MEDLINE | ID: mdl-31018959

ABSTRACT

OBJECTIVE: We investigated whether the normal human spinal enthesis contained resident myeloid cell populations, capable of producing pivotal proinflammatory cytokines including tumour necrosis factor (TNF) and interleukin (IL)-23 and determined whether these could be modified by PDE4 inhibition. METHODS: Normal human enthesis soft tissue (ST) and adjacent perientheseal bone (PEB) (n=15) were evaluated using immunohistochemistry (IHC), digested for myeloid cell phenotyping, sorted and stimulated with different adjuvants (lipopolysaccharide and mannan). Stimulated enthesis fractions were analysed for inducible production of spondyloarthropathy disease-relevant mediators (IL-23 full protein, TNF, IL-1ß and CCL20). Myeloid populations were also compared with matched blood populations for further mRNA analysis and the effect of PDE4 inhibition was assessed. RESULTS: A myeloid cell population (CD45+ HLADR+ CD14+ CD11c+) phenotype was isolated from both the ST and adjacent PEB and termed 'CD14+ myeloid cells' with tissue localisation confirmed by CD14+ IHC. The CD14- fraction contained a CD123+ HLADR+ CD11c- cell population (plasmacytoid dendritic cells). The CD14+ population was the dominant entheseal producer of IL-23, IL-1ß, TNF and CCL20. IL-23 and TNF from the CD14+ population could be downregulated by a PDE4I and other agents (histamine and 8-Bromo-cAMP) which elevate cAMP. Entheseal CD14+ cells had a broadly similar gene expression profile to the corresponding CD14+ population from matched blood but showed significantly lower CCR2 gene expression. CONCLUSIONS: The human enthesis contains a CD14+ myeloid population that produces most of the inducible IL-23, IL-1ß, TNF and CCL20. This population has similar gene expression profile to the matched blood CD14+ population.


Subject(s)
Connective Tissue Cells/metabolism , Interleukin-23/biosynthesis , Myeloid Cells/metabolism , Chemokine CCL20/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Dendritic Cells/metabolism , Humans , Immunohistochemistry , Interleukin-1beta/biosynthesis , Lipopolysaccharide Receptors/metabolism , Tumor Necrosis Factor-alpha/biosynthesis
11.
Int J Mol Sci ; 19(12)2018 Dec 05.
Article in English | MEDLINE | ID: mdl-30563079

ABSTRACT

Protein kinase B (Akt) is a key enzyme in the insulin signalling cascade, required for insulin-stimulated NO production in endothelial cells (ECs). Previous studies have suggested that AMP-activated protein kinase (AMPK) activation stimulates NO synthesis and enhances insulin-stimulated Akt activation, yet these studies have largely used indirect activators of AMPK. The effects of the allosteric AMPK activator A769662 on insulin signalling and endothelial function was therefore examined in cultured human macrovascular ECs. Surprisingly, A769662 inhibited insulin-stimulated NO synthesis and Akt phosphorylation in human ECs from umbilical veins (HUVECs) and aorta (HAECs). In contrast, the AMPK activators compound 991 and AICAR had no substantial inhibitory effect on insulin-stimulated Akt phosphorylation in ECs. Inhibition of AMPK with SBI-0206965 had no effect on the inhibition of insulin-stimulated Akt phosphorylation by A769662, suggesting the inhibitory action of A769662 is AMPK-independent. A769662 decreased IGF1-stimulated Akt phosphorylation yet had no effect on VEGF-stimulated Akt signalling in HUVECs, suggesting that A769662 attenuates early insulin/IGF1 signalling. The effects of A769662 on insulin-stimulated Akt phosphorylation were specific to human ECs, as no effect was observed in the human cancer cell lines HepG2 or HeLa, as well as in mouse embryonic fibroblasts (MEFs). A769662 inhibited insulin-stimulated Erk1/2 phosphorylation in HAECs and MEFs, an effect that was independent of AMPK in MEFs. Therefore, despite being a potent AMPK activator, A769662 has effects unlikely to be mediated by AMPK in human macrovascular ECs that reduce insulin sensitivity and eNOS activation.


Subject(s)
AMP-Activated Protein Kinases/antagonists & inhibitors , Aorta/enzymology , Human Umbilical Vein Endothelial Cells/enzymology , MAP Kinase Signaling System/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Pyrones/pharmacology , Thiophenes/pharmacology , AMP-Activated Protein Kinases/metabolism , Aorta/cytology , Biphenyl Compounds , Enzyme Activation/drug effects , HeLa Cells , Hep G2 Cells , Human Umbilical Vein Endothelial Cells/cytology , Humans , Proto-Oncogene Proteins c-akt/metabolism
12.
Sci Rep ; 8(1): 5276, 2018 03 27.
Article in English | MEDLINE | ID: mdl-29588466

ABSTRACT

Recent clinical trials of the hypoglycaemic sodium-glucose co-transporter-2 (SGLT2) inhibitors, which inhibit renal glucose reabsorption, have reported beneficial cardiovascular outcomes. Whether SGLT2 inhibitors directly affect cardiovascular tissues, however, remains unclear. We have previously reported that the SGLT2 inhibitor canagliflozin activates AMP-activated protein kinase (AMPK) in immortalised cell lines and murine hepatocytes. As AMPK has anti-inflammatory actions in vascular cells, we examined whether SGLT2 inhibitors attenuated inflammatory signalling in cultured human endothelial cells. Incubation with clinically-relevant concentrations of canagliflozin, but not empagliflozin or dapagliflozin activated AMPK and inhibited IL-1ß-stimulated adhesion of pro-monocytic U937 cells and secretion of IL-6 and monocyte chemoattractant protein-1 (MCP-1). Inhibition of MCP-1 secretion was attenuated by expression of dominant-negative AMPK and was mimicked by the direct AMPK activator, A769662. Stimulation of cells with either canagliflozin or A769662 had no effect on IL-1ß-stimulated cell surface levels of adhesion molecules or nuclear factor-κB signalling. Despite these identical effects of canagliflozin and A769662, IL-1ß-stimulated IL-6/MCP-1 mRNA was inhibited by canagliflozin, but not A769662, whereas IL-1ß-stimulated c-jun N-terminal kinase phosphorylation was inhibited by A769662, but not canagliflozin. These data indicate that clinically-relevant canagliflozin concentrations directly inhibit endothelial pro-inflammatory chemokine/cytokine secretion by AMPK-dependent and -independent mechanisms without affecting early IL-1ß signalling.


Subject(s)
AMP-Activated Protein Kinases/immunology , Anti-Inflammatory Agents/pharmacology , Canagliflozin/pharmacology , Cytokines/immunology , Endothelial Cells/drug effects , Interleukin-1beta/antagonists & inhibitors , Sodium-Glucose Transporter 2 Inhibitors/pharmacology , Animals , Cells, Cultured , Chemokines/immunology , Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells , Humans , Interleukin-1beta/immunology , Mice
13.
Nat Commun ; 9(1): 168, 2018 01 12.
Article in English | MEDLINE | ID: mdl-29330478

ABSTRACT

Effective suppression of JAK-STAT signalling by the inducible inhibitor "suppressor of cytokine signalling 3" (SOCS3) is essential for limiting signalling from cytokine receptors. Here we show that cavin-1, a component of caveolae, is a functionally significant SOCS3-interacting protein. Biochemical and confocal imaging demonstrate that SOCS3 localisation to the plasma membrane requires cavin-1. SOCS3 is also critical for cavin-1 stabilisation, such that deletion of SOCS3 reduces the expression of cavin-1 and caveolin-1 proteins, thereby reducing caveola abundance in endothelial cells. Moreover, the interaction of cavin-1 and SOCS3 is essential for SOCS3 function, as loss of cavin-1 enhances cytokine-stimulated STAT3 phosphorylation and abolishes SOCS3-dependent inhibition of IL-6 signalling by cyclic AMP. Together, these findings reveal a new functionally important mechanism linking SOCS3-mediated inhibition of cytokine signalling to localisation at the plasma membrane via interaction with and stabilisation of cavin-1.


Subject(s)
Membrane Proteins/metabolism , RNA-Binding Proteins/metabolism , Suppressor of Cytokine Signaling 3 Protein/metabolism , Animals , Caveolae/physiology , Gene Deletion , Gene Expression Regulation , HEK293 Cells , Humans , Janus Kinases/genetics , Janus Kinases/metabolism , Membrane Proteins/genetics , Mice , Protein Binding , RNA-Binding Proteins/genetics , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Suppressor of Cytokine Signaling 3 Protein/genetics
14.
Pharmacol Res ; 128: 88-100, 2018 02.
Article in English | MEDLINE | ID: mdl-29037480

ABSTRACT

Exaggerated Janus kinase-signal transducer and activator of transcription (JAK-STAT) signalling is key to the pathogenesis of pro-inflammatory disorders, such as rheumatoid arthritis and cardiovascular diseases. Mutational activation of JAKs is also responsible for several haematological malignancies, including myeloproliferative neoplasms and acute lymphoblastic leukaemia. Accumulating evidence links adenosine 5'-monophosphate (AMP)-activated protein kinase (AMPK), an energy sensor and regulator of organismal and cellular metabolism, with the suppression of immune and inflammatory processes. Recent studies have shown that activation of AMPK can limit JAK-STAT-dependent signalling pathways via several mechanisms. These novel findings support AMPK activation as a strategy for management of an array of disorders characterised by hyper-activation of the JAK-STAT pathway. This review discusses the pivotal role of JAK-STAT signalling in a range of disorders and how both established clinically used and novel AMPK activators might be used to treat these conditions.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Janus Kinases/metabolism , STAT Transcription Factors/metabolism , Animals , Humans , Inflammation/metabolism , Interleukin-6/metabolism , Signal Transduction
15.
Mol Cell Endocrinol ; 440: 44-56, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27840174

ABSTRACT

Inflammation of adipose tissue in obesity is associated with increased IL-1ß, IL-6 and TNF-α secretion and proposed to contribute to insulin resistance. AMP-activated protein kinase (AMPK) regulates nutrient metabolism and is reported to have anti-inflammatory actions in adipose tissue, yet the mechanisms underlying this remain poorly characterised. The effect of AMPK activation on cytokine-stimulated proinflammatory signalling was therefore assessed in cultured adipocytes. AMPK activation inhibited IL-1ß-stimulated CXCL10 secretion, associated with reduced interleukin-1 receptor associated kinase-4 (IRAK4) phosphorylation and downregulated MKK4/JNK and IKK/IκB/NFκB signalling. AMPK activation inhibited TNF-α-stimulated IKK/IκB/NFκB signalling but had no effect on JNK phosphorylation. The JAK/STAT3 pathway was also suppressed by AMPK after IL-6 stimulation and during adipogenesis. Adipose tissue from AMPKα1-/- mice exhibited increased JNK and STAT3 phosphorylation, supporting suppression of these distinct proinflammatory pathways by AMPK in vivo. The inhibition of multiple pro-inflammatory signalling pathways by AMPK may underlie the reported beneficial effects of AMPK activation in adipose tissue.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Adipocytes/enzymology , Adipocytes/pathology , Inflammation/enzymology , Inflammation/pathology , Interleukin-1 Receptor-Associated Kinases/metabolism , 3T3-L1 Cells , Adipogenesis/drug effects , Animals , Biphenyl Compounds , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Enzyme Activation/drug effects , Female , Humans , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Mice , NF-kappa B/metabolism , Phosphorylation/drug effects , Pyrones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Thiophenes/pharmacology
16.
Sci Signal ; 9(453): ra109, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27919027

ABSTRACT

Adenosine 5'-monophosphate-activated protein kinase (AMPK) is a pivotal regulator of metabolism at cellular and organismal levels. AMPK also suppresses inflammation. We found that pharmacological activation of AMPK rapidly inhibited the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway in various cells. In vitro kinase assays revealed that AMPK directly phosphorylated two residues (Ser515 and Ser518) within the Src homology 2 domain of JAK1. Activation of AMPK enhanced the interaction between JAK1 and 14-3-3 proteins in cultured vascular endothelial cells and fibroblasts, an effect that required the presence of Ser515 and Ser518 and was abolished in cells lacking AMPK catalytic subunits. Mutation of Ser515 and Ser518 abolished AMPK-mediated inhibition of JAK-STAT signaling stimulated by either the sIL-6Rα/IL-6 complex or the expression of a constitutively active V658F-mutant JAK1 in human fibrosarcoma cells. Clinically used AMPK activators metformin and salicylate enhanced the inhibitory phosphorylation of endogenous JAK1 and inhibited STAT3 phosphorylation in primary vascular endothelial cells. Therefore, our findings reveal a mechanism by which JAK1 function and inflammatory signaling may be suppressed in response to metabolic stress and provide a mechanistic rationale for the investigation of AMPK activators in a range of diseases associated with enhanced activation of the JAK-STAT pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Endothelial Cells/metabolism , Janus Kinase 1/metabolism , Signal Transduction/physiology , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , AMP-Activated Protein Kinases/genetics , Amino Acid Substitution , Animals , Endothelial Cells/cytology , Enzyme Activation , Janus Kinase 1/genetics , Mice , Mice, Knockout , Mutation, Missense , Phosphorylation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism
17.
Biochem J ; 473(24): 4681-4697, 2016 Dec 15.
Article in English | MEDLINE | ID: mdl-27784766

ABSTRACT

The key metabolic regulator, AMP-activated protein kinase (AMPK), is reported to be down-regulated in metabolic disorders, but the mechanisms are poorly characterised. Recent studies have identified phosphorylation of the AMPKα1/α2 catalytic subunit isoforms at Ser487/491, respectively, as an inhibitory regulation mechanism. Vascular endothelial growth factor (VEGF) stimulates AMPK and protein kinase B (Akt) in cultured human endothelial cells. As Akt has been demonstrated to be an AMPKα1 Ser487 kinase, the effect of VEGF on inhibitory AMPK phosphorylation in cultured primary human endothelial cells was examined. Stimulation of endothelial cells with VEGF rapidly increased AMPKα1 Ser487 phosphorylation in an Akt-independent manner, without altering AMPKα2 Ser491 phosphorylation. In contrast, VEGF-stimulated AMPKα1 Ser487 phosphorylation was sensitive to inhibitors of protein kinase C (PKC) and PKC activation using phorbol esters or overexpression of PKC-stimulated AMPKα1 Ser487 phosphorylation. Purified PKC and Akt both phosphorylated AMPKα1 Ser487 in vitro with similar efficiency. PKC activation was associated with reduced AMPK activity, as inhibition of PKC increased AMPK activity and phorbol esters inhibited AMPK, an effect lost in cells expressing mutant AMPKα1 Ser487Ala. Consistent with a pathophysiological role for this modification, AMPKα1 Ser487 phosphorylation was inversely correlated with insulin sensitivity in human muscle. These data indicate a novel regulatory role of PKC to inhibit AMPKα1 in human cells. As PKC activation is associated with insulin resistance and obesity, PKC may underlie the reduced AMPK activity reported in response to overnutrition in insulin-resistant metabolic and vascular tissues.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Protein Kinase C/metabolism , Cell Line , HEK293 Cells , HeLa Cells , Human Umbilical Vein Endothelial Cells , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Serine/metabolism , Signal Transduction/drug effects , Vascular Endothelial Growth Factor A/pharmacology
18.
Trends Pharmacol Sci ; 36(4): 203-14, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25744542

ABSTRACT

Pharmaceutical manipulation of cAMP levels exerts beneficial effects through the regulation of the exchange protein activated by cAMP (EPAC) and protein kinase A (PKA) signalling routes. Recent attention has turned to the specific regulation of EPAC isoforms (EPAC1 and EPAC2) as a more targeted approach to cAMP-based therapies. For example, EPAC2-selective agonists could promote insulin secretion from pancreatic ß cells, whereas EPAC1-selective agonists may be useful in the treatment of vascular inflammation. By contrast, EPAC1 and EPAC2 antagonists could both be useful in the treatment of heart failure. Here we discuss whether the best way forward is to design EPAC-selective agonists or antagonists and the current strategies being used to develop isoform-selective, small-molecule regulators of EPAC1 and EPAC2 activity.


Subject(s)
Drug Delivery Systems/trends , Guanine Nucleotide Exchange Factors/agonists , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Animals , Cyclic AMP/agonists , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Forecasting , Guanine Nucleotide Exchange Factors/metabolism , Humans , Protein Structure, Secondary , Protein Structure, Tertiary
19.
Cells ; 3(2): 546-62, 2014 May 30.
Article in English | MEDLINE | ID: mdl-24886706

ABSTRACT

The realisation that unregulated activation of the Janus kinase-signal transducer and activator of transcription (JAK-STAT) pathway is a key driver of a wide range of diseases has identified its components as targets for therapeutic intervention by small molecule inhibitors and biologicals. In this review, we discuss JAK-STAT signalling pathway inhibition by the inducible inhibitor "suppressor of cytokine signaling 3 (SOCS3), its role in diseases such as myeloproliferative disorders, and its function as part of a multi-subunit E3 ubiquitin ligase complex. In addition, we highlight potential applications of these insights into SOCS3-based therapeutic strategies for management of conditions such as vascular re-stenosis associated with acute vascular injury, where there is strong evidence that multiple processes involved in disease progression could be attenuated by localized potentiation of SOCS3 expression levels.

20.
Biochem Soc Trans ; 42(2): 284-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24646232

ABSTRACT

Caveolae are curved lipid raft regions rich in cholesterol and sphingolipids found abundantly in vascular endothelial cells, adipocytes, smooth muscle cells and fibroblasts. They are multifunctional organelles with roles in clathrin-independent endocytosis, cholesterol transport, mechanosensing and signal transduction. Caveolae provide an environment where multiple receptor signalling components are sequestered, clustered and compartmentalized for efficient signal transduction. Many of these receptors, including cytokine signal transducer gp130 (glycoprotein 130), are mediators of chronic inflammation during atherogenesis. Subsequently, disruption of these organelles is associated with a broad range of disease states including cardiovascular disease and cancer. Cavin-1 is an essential peripheral component of caveolae that stabilizes caveolin-1, the main structural/integral membrane protein of caveolae. Caveolin-1 is an essential regulator of eNOS (endothelial nitric oxide synthase) and its disruption leads to endothelial dysfunction which initiates a range of cardiovascular and pulmonary disorders. Although dysfunctional cytokine signalling is also a hallmark of cardiovascular disease, knowledge of caveolae-dependent cytokine signalling is lacking as is the role of cavin-1 independent of caveolae. The present review introduces caveolae, their structural components, the caveolins and cavins, their regulation by cAMP, and their potential role in cardiovascular disease.


Subject(s)
Cardiovascular Diseases/metabolism , Caveolae/metabolism , Caveolin 1/metabolism , Animals , Cyclic AMP/metabolism , Humans , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...