Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 25
1.
Cardiovasc Res ; 119(12): 2179-2189, 2023 10 16.
Article En | MEDLINE | ID: mdl-37309666

AIMS: Atherosclerosis is driven by multiple processes across multiple body systems. For example, the innate immune system drives both atherogenesis and plaque rupture via inflammation, while coronary artery-occluding thrombi formed by the coagulation system cause myocardial infarction and death. However, the interplay between these systems during atherogenesis is understudied. We recently showed that coagulation and immunity are fundamentally linked by the activation of interleukin-1α (IL-1α) by thrombin, and generated a novel knock-in mouse in which thrombin cannot activate endogenous IL-1α [IL-1α thrombin mutant (IL-1αTM)]. METHODS AND RESULTS: Here, we show significantly reduced atherosclerotic plaque formation in IL-1αTM/Apoe-/- mice compared with Apoe-/- and reduced T-cell infiltration. However, IL-1αTM/Apoe-/- plaques have reduced vascular smooth muscle cells, collagen, and fibrous caps, indicative of a more unstable phenotype. Interestingly, the reduced atherogenesis seen with thrombin inhibition was absent in IL-1αTM/Apoe-/- mice, suggesting that thrombin inhibitors can affect atherosclerosis via reduced IL-1α activation. Finally, bone marrow chimeras show that thrombin-activated IL-1α is derived from both vessel wall and myeloid cells. CONCLUSIONS: Together, we reveal that the atherogenic effect of ongoing coagulation is, in part, mediated via thrombin cleavage of IL-1α. This not only highlights the importance of interplay between systems during disease and the potential for therapeutically targeting IL-1α and/or thrombin, but also forewarns that IL-1 may have a role in plaque stabilization.


Atherosclerosis , Plaque, Atherosclerotic , Thrombin , Animals , Mice , Apolipoproteins E/genetics , Atherosclerosis/genetics , Atherosclerosis/prevention & control , Cell Proliferation , Collagen/metabolism , Interleukin-1alpha/genetics , Interleukin-1alpha/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Plaque, Atherosclerotic/metabolism , Thrombin/metabolism
2.
Eur J Immunol ; 50(11): 1663-1675, 2020 11.
Article En | MEDLINE | ID: mdl-32447774

IL-1 is a powerful cytokine that drives inflammation and modulates adaptive immunity. Both IL-1α and IL-1ß are translated as proforms that require cleavage for full cytokine activity and release, while IL-1α is reported to occur as an alternative plasma membrane-associated form on many cell types. However, the existence of cell surface IL-1α (csIL-1α) is contested, how IL-1α tethers to the membrane is unknown, and signaling pathways controlling trafficking are not specified. Using a robust and fully validated system, we show that macrophages present bona fide csIL-1α after ligation of TLRs. Pro-IL-1α tethers to the plasma membrane in part through IL-1R2 or via association with a glycosylphosphatidylinositol-anchored protein, and can be cleaved, activated, and released by proteases. csIL-1α requires de novo protein synthesis and its trafficking to the plasma membrane is exquisitely sensitive to inhibition by IFN-γ, independent of expression level. We also reveal how prior csIL-1α detection could occur through inadvertent cell permeabilisation, and that senescent cells do not drive the senescent-associated secretory phenotype via csIL-1α, but rather via soluble IL-1α. We believe these data are important for determining the local or systemic context in which IL-1α can contribute to disease and/or physiological processes.


Cell Membrane/metabolism , Glycosylphosphatidylinositols/metabolism , Interferon-gamma/metabolism , Interleukin-1alpha/metabolism , Receptors, Interleukin-1 Type II/metabolism , Animals , Humans , Inflammation/metabolism , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Protein Binding/physiology , Protein Transport/physiology
3.
Nat Commun ; 11(1): 307, 2020 01 16.
Article En | MEDLINE | ID: mdl-31949142

Autophagy is an important cellular degradation pathway with a central role in metabolism as well as basic quality control, two processes inextricably linked to ageing. A decrease in autophagy is associated with increasing age, yet it is unknown if this is causal in the ageing process, and whether autophagy restoration can counteract these ageing effects. Here we demonstrate that systemic autophagy inhibition induces the premature acquisition of age-associated phenotypes and pathologies in mammals. Remarkably, autophagy restoration provides a near complete recovery of morbidity and a significant extension of lifespan; however, at the molecular level this rescue appears incomplete. Importantly autophagy-restored mice still succumb earlier due to an increase in spontaneous tumour formation. Thus, our data suggest that chronic autophagy inhibition confers an irreversible increase in cancer risk and uncovers a biphasic role of autophagy in cancer development being both tumour suppressive and oncogenic, sequentially.


Aging/physiology , Autophagy/drug effects , Autophagy/physiology , Longevity/physiology , Neoplasms , Aging/genetics , Animals , Autophagy/genetics , Autophagy-Related Protein 5/genetics , Bone Marrow Transplantation , Disease Models, Animal , Female , Inflammation , Longevity/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscles , Phenotype , Sequestosome-1 Protein/metabolism , Skin/pathology
4.
Front Immunol ; 11: 613170, 2020.
Article En | MEDLINE | ID: mdl-33391283

Cytokines activate or inhibit immune cell behavior and are thus integral to all immune responses. IL-1α and IL-1ß are powerful apical cytokines that instigate multiple downstream processes to affect both innate and adaptive immunity. Multiple studies show that IL-1ß is typically activated in macrophages after inflammasome sensing of infection or danger, leading to caspase-1 processing of IL-1ß and its release. However, many alternative mechanisms activate IL-1α and IL-1ß in atypical cell types, and IL-1 function is also important for homeostatic processes that maintain a physiological state. This review focuses on the less studied, yet arguably more interesting biology of IL-1. We detail the production by, and effects of IL-1 on specific innate and adaptive immune cells, report how IL-1 is required for barrier function at multiple sites, and discuss how perturbation of IL-1 pathways can drive disease. Thus, although IL-1 is primarily studied for driving inflammation after release from macrophages, it is clear that it has a multifaceted role that extends far beyond this, with various unconventional effects of IL-1 vital for health. However, much is still unknown, and a detailed understanding of cell-type and context-dependent actions of IL-1 is required to truly understand this enigmatic cytokine, and safely deploy therapeutics for the betterment of human health.


Interleukin-1/immunology , Adaptive Immunity/immunology , Animals , Homeostasis/immunology , Humans , Immunity, Innate/immunology , Macrophages/immunology
5.
Nat Rev Cardiol ; 16(12): 727-744, 2019 12.
Article En | MEDLINE | ID: mdl-31243391

Vascular smooth muscle cells (VSMCs) are a major cell type present at all stages of an atherosclerotic plaque. According to the 'response to injury' and 'vulnerable plaque' hypotheses, contractile VSMCs recruited from the media undergo phenotypic conversion to proliferative synthetic cells that generate extracellular matrix to form the fibrous cap and hence stabilize plaques. However, lineage-tracing studies have highlighted flaws in the interpretation of former studies, revealing that these studies had underestimated both the content and functions of VSMCs in plaques and have thus challenged our view on the role of VSMCs in atherosclerosis. VSMCs are more plastic than previously recognized and can adopt alternative phenotypes, including phenotypes resembling foam cells, macrophages, mesenchymal stem cells and osteochondrogenic cells, which could contribute both positively and negatively to disease progression. In this Review, we present the evidence for VSMC plasticity and summarize the roles of VSMCs and VSMC-derived cells in atherosclerotic plaque development and progression. Correct attribution and spatiotemporal resolution of clinically beneficial and detrimental processes will underpin the success of any therapeutic intervention aimed at VSMCs and their derivatives.


Atherosclerosis/pathology , Myocytes, Smooth Muscle/pathology , Animals , Cell Plasticity , Cell Proliferation , Cellular Senescence , Disease Progression , Extracellular Matrix/physiology , Humans , Myocytes, Smooth Muscle/physiology , Phenotype , Plaque, Atherosclerotic/pathology
6.
Immunity ; 50(6): 1339-1341, 2019 06 18.
Article En | MEDLINE | ID: mdl-31216455

Pyroptotic cell death during endotoxemia causes death via unknown mechanisms. In this issue of Immunity, Wu et al. (2019) show that T3SS rod proteins or LPS induces inflammasome activation, macrophage pyroptosis, and accompanying tissue factor release, directly connecting inflammation to coagulation.


Inflammasomes , Pyroptosis , Blood Coagulation , Humans , Inflammation , Macrophages
7.
Aging Cell ; 18(3): e12946, 2019 06.
Article En | MEDLINE | ID: mdl-30916891

Interleukin-1 alpha (IL-1α) is a powerful cytokine that modulates immunity, and requires canonical cleavage by calpain for full activity. Mature IL-1α is produced after inflammasome activation and during cell senescence, but the protease cleaving IL-1α in these contexts is unknown. We show IL-1α is activated by caspase-5 or caspase-11 cleavage at a conserved site. Caspase-5 drives cleaved IL-1α release after human macrophage inflammasome activation, while IL-1α secretion from murine macrophages only requires caspase-11, with IL-1ß release needing caspase-11 and caspase-1. Importantly, senescent human cells require caspase-5 for the IL-1α-dependent senescence-associated secretory phenotype (SASP) in vitro, while senescent mouse hepatocytes need caspase-11 for the SASP-driven immune surveillance of senescent cells in vivo. Together, we identify IL-1α as a novel substrate of noncanonical inflammatory caspases and finally provide a mechanism for how IL-1α is activated during senescence. Thus, targeting caspase-5 may reduce inflammation and limit the deleterious effects of accumulated senescent cells during disease and Aging.


Caspases/metabolism , Cellular Senescence , Inflammasomes/metabolism , Inflammation/metabolism , Interleukin-1alpha/metabolism , Animals , Cells, Cultured , Female , HeLa Cells , Humans , Interleukin-1alpha/analysis , Mice , Mice, Inbred C57BL
8.
Immunity ; 50(4): 1033-1042.e6, 2019 04 16.
Article En | MEDLINE | ID: mdl-30926232

Ancient organisms have a combined coagulation and immune system, and although links between inflammation and hemostasis exist in mammals, they are indirect and slower to act. Here we investigated direct links between mammalian immune and coagulation systems by examining cytokine proproteins for potential thrombin protease consensus sites. We found that interleukin (IL)-1α is directly activated by thrombin. Thrombin cleaved pro-IL-1α at a site perfectly conserved across disparate species, indicating functional importance. Surface pro-IL-1α on macrophages and activated platelets was cleaved and activated by thrombin, while tissue factor, a potent thrombin activator, colocalized with pro-IL-1α in the epidermis. Mice bearing a mutation in the IL-1α thrombin cleavage site (R114Q) exhibited defects in efficient wound healing and rapid thrombopoiesis after acute platelet loss. Thrombin-cleaved IL-1α was detected in humans during sepsis, pointing to the relevance of this pathway for normal physiology and the pathogenesis of inflammatory and thrombotic diseases.


Blood Coagulation/physiology , Immune System/immunology , Interleukin-1alpha/physiology , Thrombin/physiology , Adaptive Immunity , Amino Acid Sequence , Animals , Blood Platelets/metabolism , Humans , Immunity, Innate , Interleukin-1alpha/genetics , Interleukin-1alpha/immunology , Keratinocytes/metabolism , Macrophages/metabolism , Mammals/immunology , Mice , Protein Precursors/metabolism , Selection, Genetic , Sepsis/immunology , Sequence Alignment , Sequence Homology, Amino Acid , Thrombopoiesis/immunology , Wound Healing/immunology
9.
Bio Protoc ; 9(20): e3405, 2019 Oct 20.
Article En | MEDLINE | ID: mdl-33654906

Platelets regulate hemostasis and are the key determinants of pathogenic thrombosis following atherosclerotic plaque rupture. Platelets circulate in an inactive state, but become activated in response to damage to the endothelium, which exposes thrombogenic material such as collagen to the blood flow. Activation results in a number of responses, including secretion of soluble bioactive molecules via the release of alpha and dense granules, activation of membrane adhesion receptors, release of microparticles, and externalization of phosphatidylserine. These processes facilitate firm adhesion to sites of injury and the recruitment and activation of other platelets and leukocytes, resulting in aggregation and thrombus formation. Platelet activation drives the hemostatic response, and also contributes to pathogenic thrombus formation. Thus, quantification of platelet-associated responses is key to many pathophysiologically relevant processes. Here we describe protocols for isolating, counting, and activating platelets, and for the rapid quantification of cell surface proteins using flow cytometry.

12.
J Biol Chem ; 290(41): 25188-96, 2015 Oct 09.
Article En | MEDLINE | ID: mdl-26324711

Inflammation is a key instigator of the immune responses that drive atherosclerosis and allograft rejection. IL-1α, a powerful cytokine that activates both innate and adaptive immunity, induces vessel inflammation after release from necrotic vascular smooth muscle cells (VSMCs). Similarly, IL-1α released from endothelial cells (ECs) damaged during transplant drives allograft rejection. However, IL-1α requires cleavage for full cytokine activity, and what controls cleavage in necrotic ECs is currently unknown. We find that ECs have very low levels of IL-1α activity upon necrosis. However, TNFα or IL-1 induces significant levels of active IL-1α in EC necrotic lysates without alteration in protein levels. Increased activity requires cleavage of IL-1α by calpain to the more active mature form. Immunofluorescence and proximity ligation assays show that IL-1α associates with interleukin-1 receptor-2, and this association is decreased by TNFα or IL-1 and requires caspase activity. Thus, TNFα or IL-1 treatment of ECs leads to caspase proteolytic activity that cleaves interleukin-1 receptor-2, allowing IL-1α dissociation and subsequent processing by calpain. Importantly, ECs could be primed by IL-1α from adjacent damaged VSMCs, and necrotic ECs could activate neighboring normal ECs and VSMCs, causing them to release inflammatory cytokines and up-regulate adhesion molecules, thus amplifying inflammation. These data unravel the molecular mechanisms and interplay between damaged ECs and VSMCs that lead to activation of IL-1α and, thus, initiation of adaptive responses that cause graft rejection.


Allografts/immunology , Caspase 1/metabolism , Graft Rejection/metabolism , Graft Rejection/pathology , Human Umbilical Vein Endothelial Cells/pathology , Interleukin-1alpha/metabolism , Receptors, Interleukin-1 Type II/metabolism , Calpain/metabolism , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/immunology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Interleukin-1/pharmacology , Necrosis/immunology , Proteolysis , Tumor Necrosis Factor-alpha/pharmacology
13.
Arterioscler Thromb Vasc Biol ; 35(9): 1963-74, 2015 Sep.
Article En | MEDLINE | ID: mdl-26139463

OBJECTIVE: Vascular smooth muscle cells (VSMCs) that become senescent are both present within atherosclerotic plaques and thought to be important to the disease process. However, senescent VSMCs are generally considered to only contribute through inaction, with failure to proliferate resulting in VSMC- and collagen-poor unstable fibrous caps. Whether senescent VSMCs can actively contribute to atherogenic processes, such as inflammation, is unknown. APPROACH AND RESULTS: We find that senescent human VSMCs develop a proinflammatory state known as a senescence-associated secretory phenotype. Senescent human VSMCs release high levels of multiple cytokines and chemokines driven by secreted interleukin-1α acting in an autocrine manner. Consequently, the VSMC senescence-associated secretory phenotype promotes chemotaxis of mononuclear cells in vitro and in vivo. In addition, senescent VSMCs release active matrix metalloproteinase-9, secrete less collagen, upregulate multiple inflammasome components, and prime adjacent endothelial cells and VSMCs to a proadhesive and proinflammatory state. Importantly, maintaining the senescence-associated secretory phenotype places a large metabolic burden on senescent VSMCs, such that they can be selectively killed by inhibiting glucose utilization. CONCLUSIONS: Senescent VSMCs may actively contribute toward the chronic inflammation associated with atherosclerosis through the interleukin-1α-driven senescence-associated secretory phenotype and the priming of adjacent cells to a proatherosclerotic state. These data also suggest that inhibition of this potentially important source of chronic inflammation in atherosclerosis requires blockade of interleukin-1α and not interleukin-1ß.


Cellular Senescence/genetics , Gene Expression Regulation , Inflammation/genetics , Interleukin-1alpha/genetics , Muscle, Smooth, Vascular/metabolism , Plaque, Atherosclerotic/genetics , RNA/genetics , Animals , Cells, Cultured , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Disease Models, Animal , Flow Cytometry , Humans , Inflammation/metabolism , Inflammation/pathology , Interleukin-1alpha/biosynthesis , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Phenotype , Plaque, Atherosclerotic/metabolism , Plaque, Atherosclerotic/pathology , Regulatory Factor X Transcription Factors , Reverse Transcriptase Polymerase Chain Reaction , Transcription Factors/biosynthesis , Transcription Factors/genetics
15.
Cardiovasc Res ; 101(3): 513-21, 2014 Mar 01.
Article En | MEDLINE | ID: mdl-24323316

AIMS: The chemokine receptor CCR5 and its inflammatory ligands have been linked to atherosclerosis, an accelerated form of which occurs in saphenous vein graft disease. We investigated the function of vascular smooth muscle CCR5 in human coronary artery and saphenous vein, vascular tissues susceptible to atherosclerosis, and vasospasm. METHODS AND RESULTS: CCR5 ligands were vasoconstrictors in saphenous vein and coronary artery. In vein, constrictor responses to CCL4 were completely blocked by CCR5 antagonists, including maraviroc. CCR5 antagonists prevented the development of a neointima after 14 days in cultured saphenous vein. CCR5 and its ligands were expressed in normal and diseased coronary artery and saphenous vein and localized to medial and intimal smooth muscle, endothelial, and inflammatory cells. [(125)I]-CCL4 bound to venous smooth muscle with KD = 1.15 ± 0.26 nmol/L and density of 22 ± 9 fmol mg(-1) protein. CONCLUSIONS: Our data support a potential role for CCR5 in vasoconstriction and neointimal formation in vitro and imply that CCR5 chemokines may contribute to vascular remodelling and augmented vascular tone in human coronary artery and vein graft disease. The repurposing of maraviroc for the treatment of cardiovascular disease warrants further investigation.


Muscle, Smooth, Vascular/blood supply , Receptors, CCR5/metabolism , Saphenous Vein/metabolism , Tunica Intima/pathology , Vasoconstriction/physiology , Atherosclerosis/metabolism , Cells, Cultured , Chemokines/metabolism , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Muscle, Smooth, Vascular/pathology , Organ Culture Techniques , Saphenous Vein/pathology , Tunica Intima/metabolism
16.
Circulation ; 128(7): 702-12, 2013 Aug 13.
Article En | MEDLINE | ID: mdl-23841983

BACKGROUND: Mitochondrial DNA (mtDNA) damage occurs in both circulating cells and the vessel wall in human atherosclerosis. However, it is unclear whether mtDNA damage directly promotes atherogenesis or is a consequence of tissue damage, which cell types are involved, and whether its effects are mediated only through reactive oxygen species. METHODS AND RESULTS: mtDNA damage occurred early in the vessel wall in apolipoprotein E-null (ApoE(-/-)) mice, before significant atherosclerosis developed. mtDNA defects were also identified in circulating monocytes and liver and were associated with mitochondrial dysfunction. To determine whether mtDNA damage directly promotes atherosclerosis, we studied ApoE(-/-) mice deficient for mitochondrial polymerase-γ proofreading activity (polG(-/-)/ApoE(-/-)). polG(-/-)/ApoE(-/-) mice showed extensive mtDNA damage and defects in oxidative phosphorylation but no increase in reactive oxygen species. polG(-/-)/ApoE(-/-) mice showed increased atherosclerosis, associated with impaired proliferation and apoptosis of vascular smooth muscle cells, and hyperlipidemia. Transplantation with polG(-/-)/ApoE(-/-) bone marrow increased the features of plaque vulnerability, and polG(-/-)/ApoE(-/-) monocytes showed increased apoptosis and inflammatory cytokine release. To examine mtDNA damage in human atherosclerosis, we assessed mtDNA adducts in plaques and in leukocytes from patients who had undergone virtual histology intravascular ultrasound characterization of coronary plaques. Human atherosclerotic plaques showed increased mtDNA damage compared with normal vessels; in contrast, leukocyte mtDNA damage was associated with higher-risk plaques but not plaque burden. CONCLUSIONS: We show that mtDNA damage in vessel wall and circulating cells is widespread and causative and indicates higher risk in atherosclerosis. Protection against mtDNA damage and improvement of mitochondrial function are potential areas for new therapeutics.


Atherosclerosis/etiology , DNA Damage , DNA, Mitochondrial/chemistry , Mitochondria/pathology , Monocytes/pathology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Plaque, Atherosclerotic/pathology , Adiposity , Adult , Aged , Animals , Apolipoproteins E/genetics , Apoptosis , Atherosclerosis/genetics , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cells, Cultured , Cytokines/metabolism , DNA Adducts/analysis , DNA Polymerase gamma , DNA-Directed DNA Polymerase/deficiency , DNA-Directed DNA Polymerase/genetics , Electron Transport , Female , Humans , Hyperlipidemias/genetics , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Mitochondria/chemistry , Mitochondria/physiology , Monocytes/metabolism , Muscle, Smooth, Vascular/metabolism , Oxygen Consumption , Radiation Chimera , Reactive Oxygen Species , Risk
17.
Immunity ; 38(2): 285-95, 2013 Feb 21.
Article En | MEDLINE | ID: mdl-23395675

Necrosis can induce profound inflammation or be clinically silent. However, the mechanisms underlying such tissue specificity are unknown. Interleukin-1α (IL-1α) is a key danger signal released upon necrosis that exerts effects on both innate and adaptive immunity and is considered to be constitutively active. In contrast, we have shown that necrosis-induced IL-1α activity is tightly controlled in a cell type-specific manner. Most cell types examined expressed a cytosolic IL-1 receptor 2 (IL-1R2) whose binding to pro-IL-1α inhibited its cytokine activity. In cell types exhibiting a silent necrotic phenotype, IL-1R2 remained associated with pro-IL-1α. Cell types possessing inflammatory necrotic phenotypes either lacked IL-1R2 or had activated caspase-1 before necrosis, which degraded and dissociated IL-1R2 from pro-IL-1α. Full IL-1α activity required cleavage by calpain after necrosis, which increased its affinity for IL-1 receptor 1. Thus, we report a cell type-dependent process that fundamentally governs IL-1α activity postnecrosis and the mechanism allowing conditional release of this blockade.


Inflammation/metabolism , Interleukin-1alpha/genetics , Necrosis/metabolism , Protein Precursors/genetics , Receptors, Interleukin-1 Type II/genetics , Animals , Calpain/genetics , Calpain/immunology , Caspase 1/genetics , Caspase 1/immunology , Cell Line , Gene Expression Regulation , Humans , Inflammation/genetics , Inflammation/immunology , Interleukin-1alpha/immunology , Interleukin-1alpha/metabolism , Mice , Necrosis/genetics , Necrosis/immunology , Organ Specificity , Protein Binding , Protein Precursors/immunology , Protein Precursors/metabolism , Proteolysis , Receptors, Interleukin-1 Type II/immunology , Receptors, Interleukin-1 Type II/metabolism , Signal Transduction
18.
Arterioscler Thromb Vasc Biol ; 31(12): 2781-6, 2011 Dec.
Article En | MEDLINE | ID: mdl-22096097

Cell death and inflammation are ancient processes of fundamental biological importance in both normal physiology and pathology. This is evidenced by the profound conservation of mediators, with ancestral homologues identified from plants to humans, and the number of diseases driven by aberrant control of either process. Apoptosis is the most well-studied cell death, but many forms exist, including autophagy, necrosis, pyroptosis, paraptosis, and the obscure dark cell death. Cell death occurs throughout the cardiovascular system, from initial shaping of the heart and vasculature during development to involvement in pathologies, including atherosclerosis, aneurysm, cardiomyopathy, restenosis, and vascular graft rejection. However, determining whether cell death primarily drives pathology or is a secondary bystander effect is difficult. Inflammation, the primary response of innate immunity, is considered essential in initiating and driving vascular diseases. Cell death and inflammation are inextricably linked with their effectors modulating the other process. Indeed, an evolutionary link between cell death and inflammation occurs at caspase-1 (which activates interleukin-1ß), which can induce death by pyroptosis, and is a member of the caspase family vital for apoptosis. This review examines cell death in vascular disease, how it can induce inflammation, and finally the emergence of inflammasomes in vascular pathology.


Apoptosis/physiology , Cardiovascular Diseases/physiopathology , Inflammasomes/physiology , Inflammation/physiopathology , Cardiovascular Diseases/pathology , Cell Death/physiology , Humans , Immunity, Innate/physiology , Inflammation/pathology , Macrophages/pathology
19.
Arterioscler Thromb Vasc Biol ; 31(11): 2402-9, 2011 Nov.
Article En | MEDLINE | ID: mdl-21885847

OBJECTIVE: Although vascular smooth muscle cell (VSMC) apoptosis occurs after vessel injury and during remodeling, the direct role of VSMC death in determining final vessel structure is unclear. We sought to determine the role of VSMC apoptosis in vessel remodeling, medial repair, and neointima formation and to identify the mediators involved. METHODS AND RESULTS: The left common carotid artery was ligated in SM22α-human diphtheria toxin receptor mice, in which diphtheria toxin treatment selectively induces VSMC apoptosis. Apoptosis induced from day 7 to day 14 after ligation significantly increased neointimal and medial areas, cell proliferation, migration, and vessel size. Neointima formation depended on VSMCs, as VSMC depletion before ligation significantly reduced neointimal area and cellularity. In culture, conditioned media from apoptotic VSMCs promoted VSMC migration, proliferation, and collagen synthesis. Interleukin-6 (IL-6) secretion increased 5-fold and IL-1α 1.5-fold after apoptosis, whereas IL-6 inhibition negated the effect of apoptotic VSMC supernatants on VSMC migration, proliferation, and matrix synthesis. CONCLUSION: Signaling from apoptotic VSMCs directly promotes vessel remodeling, medial repair, and neointima formation after flow reduction. Although lumen size appears to depend on flow, VSMC apoptosis is an important determinant of vessel, medial, and neointimal size after flow reduction.


Apoptosis/physiology , Blood Vessels/pathology , Cell Movement/physiology , Cell Proliferation , Collagen/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Animals , Blood Vessels/metabolism , Female , Heparin-binding EGF-like Growth Factor , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-6/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Animal , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Neointima/physiopathology
20.
Circ Res ; 106(2): 363-72, 2010 Feb 05.
Article En | MEDLINE | ID: mdl-19926874

RATIONALE: Atherosclerosis is characterized by lipid accumulation in the vessel wall, inflammation, and both macrophage and vascular smooth muscle cell (VSMC) apoptosis. However, whereas VSMC apoptosis in mice with established atherosclerotic plaques or hyperlipidemia increases serum levels of the proatherogenic cytokines monocyte chemotactic protein (MCP)-1, tumor necrosis factor alpha, and interleukin (IL)-6, the link between hyperlipidemia, apoptosis and inflammation, and the mechanisms by which apoptotic cells promote inflammation in atherosclerosis are unknown. OBJECTIVE: To determine whether hyperlipidemia affects apoptotic cell clearance, and identify the molecular pathways downstream of VSMC apoptosis that may promote inflammation. METHODS AND RESULTS: We find that human VSMCs are potent and efficient phagocytes of apoptotic human VSMCs, but phagocytosis is significantly reduced by oxidized low-density lipoprotein in vitro or hyperlipidemia in vivo. Necrotic human aortic VSMCs release IL-1alpha, which induces IL-6 and MCP-1 production from viable human VSMCs in vitro. In contrast, secondary necrotic VSMCs release both IL-1alpha and caspase-activated IL-1beta, augmenting IL-6 and MCP-1 production. Conditionally inducing VSMC apoptosis in situ in hyperlipidemic SM22alpha-hDTR/ApoE(-/-) mice to levels seen in human plaques increases serum MCP-1, tumor necrosis factor alpha, and IL-6, which is prevented by blocking IL-1. CONCLUSIONS: We conclude that VSMC necrosis releases IL-1alpha, whereas secondary necrosis of apoptotic VSMCs releases both IL-1alpha and beta. IL-1 from necrotic VSMCs induces the surrounding viable VSMCs to produce proinflammatory cytokines. Thus, failed clearance of apoptotic VSMCs caused by hyperlipidemia in vivo may promote the increased serum cytokines and chronic inflammation associated with atherosclerosis.


Apoptosis/physiology , Inflammation/physiopathology , Interleukin-1alpha/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Apolipoproteins E/genetics , Apolipoproteins E/metabolism , Blotting, Western , Cells, Cultured , Chemokine CCL2/blood , Chemokine CCL2/metabolism , Heparin-binding EGF-like Growth Factor , Humans , Hyperlipidemias/physiopathology , Immunohistochemistry , Inflammation/metabolism , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Interleukin-6/blood , Interleukin-6/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/parasitology , Phagocytosis/physiology
...