Try a new search

Format these results:

Searched for:

in-biosketch:yes

person:giannc02

Total Results:

92


Macrophage-Derived 25-Hydroxycholesterol Promotes Vascular Inflammation, Atherogenesis, and Lesion Remodeling

Canfrán-Duque, Alberto; Rotllan, Noemi; Zhang, Xinbo; Andrés-Blasco, Irene; Thompson, Bonne M; Sun, Jonathan; Price, Nathan L; Fernández-Fuertes, Marta; Fowler, Joseph W; Gómez-Coronado, Diego; Sessa, William C; Giannarelli, Chiara; Schneider, Robert J; Tellides, George; McDonald, Jeffrey G; Fernández-Hernando, Carlos; Suárez, Yajaira
BACKGROUND:Cross-talk between sterol metabolism and inflammatory pathways has been demonstrated to significantly affect the development of atherosclerosis. Cholesterol biosynthetic intermediates and derivatives are increasingly recognized as key immune regulators of macrophages in response to innate immune activation and lipid overloading. 25-Hydroxycholesterol (25-HC) is produced as an oxidation product of cholesterol by the enzyme cholesterol 25-hydroxylase (CH25H) and belongs to a family of bioactive cholesterol derivatives produced by cells in response to fluctuating cholesterol levels and immune activation. Despite the major role of 25-HC as a mediator of innate and adaptive immune responses, its contribution during the progression of atherosclerosis remains unclear. METHODS: RESULTS:We found that 25-HC accumulated in human coronary atherosclerotic lesions and that macrophage-derived 25-HC accelerated atherosclerosis progression, promoting plaque instability through autocrine and paracrine actions. 25-HC amplified the inflammatory response of lipid-loaded macrophages and inhibited the migration of smooth muscle cells within the plaque. 25-HC intensified inflammatory responses of lipid-laden macrophages by modifying the pool of accessible cholesterol in the plasma membrane, which altered Toll-like receptor 4 signaling, promoted nuclear factor-κB-mediated proinflammatory gene expression, and increased apoptosis susceptibility. These effects were independent of 25-HC-mediated modulation of liver X receptor or SREBP (sterol regulatory element-binding protein) transcriptional activity. CONCLUSIONS:Production of 25-HC by activated macrophages amplifies their inflammatory phenotype, thus promoting atherogenesis.
PMID: 36416142
ISSN: 1524-4539
CID: 5384202

Editorial: Immune and autoimmune mechanisms in cardiovascular disease

Winkels, Holger; Gerdes, Norbert; Kahles, Florian; Dutta, Partha; Giannarelli, Chiara; Wolf, Dennis
SCOPUS:85146891062
ISSN: 2297-055x
CID: 5424012

Publisher Correction: Systems immunology-based drug repurposing framework to target inflammation in atherosclerosis (Nature Cardiovascular Research, (2023), 2, 6, (550-571), 10.1038/s44161-023-00278-y)

Amadori, Letizia; Calcagno, Claudia; Fernandez, Dawn M.; Koplev, Simon; Fernandez, Nicolas; Kaur, Ravneet; Mury, Pauline; Khan, Nayaab S.; Sajja, Swathy; Shamailova, Roza; Cyr, Yannick; Jeon, Minji; Hill, Christopher A.; Chong, Peik Sean; Naidu, Sonum; Sakurai, Ken; Ghotbi, Adam Ali; Soler, Raphael; Eberhardt, Natalia; Rahman, Adeeb; Faries, Peter; Moore, Kathryn J.; Fayad, Zahi A.; Ma"™ayan, Avi; Giannarelli, Chiara
Correction to: Nature Cardiovascular Research. Published online 8 June 2023. In the version of this article initially published, a protein (pMAPKAPK2) was misspelled in Fig. 1 and Extended Data Fig. 7; a colored box for "AKT" was missing from the second column of regulators in Fig. 5a; Extended Data Fig. 2f was missing a header above the color key; and typographical errors (extraneous citations to refs. 1, 3 and 5) were present in the "Analysis of RNA-seq data from saracatinib-treated tissue" section of Methods. In addition, the Reporting Summary and the legends for Supplementary Figs. 5 and 8 were outdated versions. The errors have been corrected in the HTML and PDF versions of the article.
SCOPUS:85164454863
ISSN: 2731-0590
CID: 5549612

Loss of PRMT2 in myeloid cells in normoglycemic mice phenocopies impaired regression of atherosclerosis in diabetic mice

Vurusaner, Beyza; Thevkar-Nages, Prashanth; Kaur, Ravneet; Giannarelli, Chiara; Garabedian, Michael J; Fisher, Edward A
The regression, or resolution, of inflammation in atherosclerotic plaques is impaired in diabetes. However, the factors mediating this effect remain incomplete. We identified protein arginine methyltransferase 2 (PRMT2) as a protein whose expression in macrophages is reduced in hyperglycemia and diabetes. PRMT2 catalyzes arginine methylation to target proteins to modulate gene expression. Because PRMT2 expression is reduced in cells in hyperglycemia, we wanted to determine whether PRMT2 plays a causal role in the impairment of atherosclerosis regression in diabetes. We, therefore, examined the consequence of deleting PRMT2 in myeloid cells during the regression of atherosclerosis in normal and diabetic mice. Remarkably, we found significant impairment of atherosclerosis regression under normoglycemic conditions in mice lacking PRMT2 (Prmt2-/-) in myeloid cells that mimic the decrease in regression of atherosclerosis in WT mice under diabetic conditions. This was associated with increased plaque macrophage retention, as well as increased apoptosis and necrosis. PRMT2-deficient plaque CD68+ cells under normoglycemic conditions showed increased expression of genes involved in cytokine signaling and inflammation compared to WT cells. Consistently, Prmt2-/- bone marrow-derived macrophages (BMDMs) showed an increased response of proinflammatory genes to LPS and a decreased response of inflammation resolving genes to IL-4. This increased response to LPS in Prmt2-/- BMDMs occurs via enhanced NF-kappa B activity. Thus, the loss of PRMT2 is causally linked to impaired atherosclerosis regression via a heightened inflammatory response in macrophages. That PRMT2 expression was lower in myeloid cells in plaques from human subjects with diabetes supports the relevance of our findings to human atherosclerosis.
PMCID:9283439
PMID: 35835907
ISSN: 2045-2322
CID: 5278482

Systematically evaluating DOTATATE and FDG as PET immuno-imaging tracers of cardiovascular inflammation

Toner, Yohana C; Ghotbi, Adam A; Naidu, Sonum; Sakurai, Ken; van Leent, Mandy M T; Jordan, Stefan; Ordikhani, Farideh; Amadori, Letizia; Sofias, Alexandros Marios; Fisher, Elizabeth L; Maier, Alexander; Sullivan, Nathaniel; Munitz, Jazz; Senders, Max L; Mason, Christian; Reiner, Thomas; Soultanidis, Georgios; Tarkin, Jason M; Rudd, James H F; Giannarelli, Chiara; Ochando, Jordi; Pérez-Medina, Carlos; Kjaer, Andreas; Mulder, Willem J M; Fayad, Zahi A; Calcagno, Claudia
In recent years, cardiovascular immuno-imaging by positron emission tomography (PET) has undergone tremendous progress in preclinical settings. Clinically, two approved PET tracers hold great potential for inflammation imaging in cardiovascular patients, namely FDG and DOTATATE. While the former is a widely applied metabolic tracer, DOTATATE is a relatively new PET tracer targeting the somatostatin receptor 2 (SST2). In the current study, we performed a detailed, head-to-head comparison of DOTATATE-based radiotracers and [18F]F-FDG in mouse and rabbit models of cardiovascular inflammation. For mouse experiments, we labeled DOTATATE with the long-lived isotope [64Cu]Cu to enable studying the tracer's mode of action by complementing in vivo PET/CT experiments with thorough ex vivo immunological analyses. For translational PET/MRI rabbit studies, we employed the more widely clinically used [68Ga]Ga-labeled DOTATATE, which was approved by the FDA in 2016. DOTATATE's pharmacokinetics and timed biodistribution were determined in control and atherosclerotic mice and rabbits by ex vivo gamma counting of blood and organs. Additionally, we performed in vivo PET/CT experiments in mice with atherosclerosis, mice subjected to myocardial infarction and control animals, using both [64Cu]Cu-DOTATATE and [18F]F-FDG. To evaluate differences in the tracers' cellular specificity, we performed ensuing ex vivo flow cytometry and gamma counting. In mice subjected to myocardial infarction, in vivo [64Cu]Cu-DOTATATE PET showed higher differential uptake between infarcted (SUVmax 1.3, IQR, 1.2-1.4, N = 4) and remote myocardium (SUVmax 0.7, IQR, 0.5-0.8, N = 4, p = 0.0286), and with respect to controls (SUVmax 0.6, IQR, 0.5-0.7, N = 4, p = 0.0286), than [18F]F-FDG PET. In atherosclerotic mice, [64Cu]Cu-DOTATATE PET aortic signal, but not [18F]F-FDG PET, was higher compared to controls (SUVmax 1.1, IQR, 0.9-1.3 and 0.5, IQR, 0.5-0.6, respectively, N = 4, p = 0.0286). In both models, [64Cu]Cu-DOTATATE demonstrated preferential accumulation in macrophages with respect to other myeloid cells, while [18F]F-FDG was taken up by macrophages and other leukocytes. In a translational PET/MRI study in atherosclerotic rabbits, we then compared [68Ga]Ga-DOTATATE and [18F]F-FDG for the assessment of aortic inflammation, combined with ex vivo radiometric assays and near-infrared imaging of macrophage burden. Rabbit experiments showed significantly higher aortic accumulation of both [68Ga]Ga-DOTATATE and [18F]F-FDG in atherosclerotic (SUVmax 0.415, IQR, 0.338-0.499, N = 32 and 0.446, IQR, 0.387-0.536, N = 27, respectively) compared to control animals (SUVmax 0.253, IQR, 0.197-0.285, p = 0.0002, N = 10 and 0.349, IQR, 0.299-0.423, p = 0.0159, N = 11, respectively). In conclusion, we present a detailed, head-to-head comparison of the novel SST2-specific tracer DOTATATE and the validated metabolic tracer [18F]F-FDG for the evaluation of inflammation in small animal models of cardiovascular disease. Our results support further investigations on the use of DOTATATE to assess cardiovascular inflammation as a complementary readout to the widely used [18F]F-FDG.
PMCID:9007951
PMID: 35418569
ISSN: 2045-2322
CID: 5202012

False Utopia of One Unifying Description of the Vulnerable Atherosclerotic Plaque: A Call for Recalibration That Appreciates the Diversity of Mechanisms Leading to Atherosclerotic Disease

Pasterkamp, Gerard; den Ruijter, Hester M; Giannarelli, Chiara
Atherosclerosis is a complex disease characterized by the formation of arterial plaques with a broad diversity of morphological phenotypic presentations. Researchers often apply one description of the vulnerable plaque as a gold standard in preclinical and clinical research that could be applied as a surrogate measure of a successful therapeutic intervention, despite the variability in lesion characteristics that may underly a thrombotic occlusion. The complex mechanistic interplay underlying progression of atherosclerotic disease is a consequence of the broad range of determinants such as sex, risk factors, hemodynamics, medications, and the genetic landscape. Currently, we are facing an overwhelming amount of data based on genetic, transcriptomic, proteomic, and metabolomic studies that all point to heterogeneous molecular profiles of atherosclerotic lesions that lead to a myocardial infarction or stroke. The observed molecular diversity implies that one unifying model cannot fully recapitulate the natural history of atherosclerosis. Despite emerging data obtained from -omics studies, a description of a natural history of atherosclerotic disease in which cell-specific expression of proteins or genes are included is still lacking. This also applies to the insights provided by genome-wide association studies. This review will critically discuss the dogma that the progression of atherosclerotic disease can be captured in one unifying natural history model of atherosclerosis.
PMID: 35139657
ISSN: 1524-4636
CID: 5176082

How Single-Cell Technologies Have Provided New Insights Into Atherosclerosis

Eberhardt, Natalia; Giannarelli, Chiara
The development of innovative single-cell technologies has allowed the high-dimensional transcriptomic and proteomic profiling of individual blood and tissue cells. Recent single-cell studies revealed a new cellular heterogeneity of atherosclerotic plaque tissue and allowed a better understanding of distinct immune functional states in the context of atherosclerosis. In this brief review, we describe how single-cell technologies have shed a new light on the cellular composition of atherosclerotic plaques, and their response to diet perturbations or genetic manipulation in mouse models of atherosclerosis. We discuss how single-cell RNA sequencing, cellular indexing of transcriptomes and epitopes by sequencing, transposase-accessible chromatin with high-throughput sequencing, and cytometry by time-of-flight platforms have empowered the identification of discrete immune, endothelial, and smooth muscle cells alterations in atherosclerosis progression and regression. Finally, we review how single-cell approaches have allowed mapping the cellular and molecular composition of human atherosclerotic plaques and the discovery of new immune alterations in plaques from patients with stroke.
PMID: 35109673
ISSN: 1524-4636
CID: 5153632

A mechanistic framework for cardiometabolic and coronary artery diseases

Koplev, Simon; Seldin, Marcus; Sukhavasi, Katyayani; Ermel, Raili; Pang, Shichao; Zeng, Lingyao; Bankier, Sean; Di Narzo, Antonio; Cheng, Haoxiang; Meda, Vamsidhar; Ma, Angela; Talukdar, Husain; Cohain, Ariella; Amadori, Letizia; Argmann, Carmen; Houten, Sander M; Franzén, Oscar; Mocci, Giuseppe; Meelu, Omar A; Ishikawa, Kiyotake; Whatling, Carl; Jain, Anamika; Jain, Rajeev Kumar; Gan, Li-Ming; Giannarelli, Chiara; Roussos, Panos; Hao, Ke; Schunkert, Heribert; Michoel, Tom; Ruusalepp, Arno; Schadt, Eric E; Kovacic, Jason C; Lusis, Aldon J; Björkegren, Johan L M
Coronary atherosclerosis results from the delicate interplay of genetic and exogenous risk factors, principally taking place in metabolic organs and the arterial wall. Here we show that 224 gene-regulatory coexpression networks (GRNs) identified by integrating genetic and clinical data from patients with (n = 600) and without (n = 250) coronary artery disease (CAD) with RNA-seq data from seven disease-relevant tissues in the Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task (STARNET) study largely capture this delicate interplay, explaining >54% of CAD heritability. Within 89 cross-tissue GRNs associated with clinical severity of CAD, 374 endocrine factors facilitated inter-organ interactions, primarily along an axis from adipose tissue to the liver (n = 152). This axis was independently replicated in genetically diverse mouse strains and by injection of recombinant forms of adipose endocrine factors (EPDR1, FCN2, FSTL3 and LBP) that markedly altered blood lipid and glucose levels in mice. Altogether, the STARNET database and the associated GRN browser (http://starnet.mssm.edu) provide a multiorgan framework for exploration of the molecular interplay between cardiometabolic disorders and CAD.
PMCID:9583458
PMID: 36276926
ISSN: 2731-0590
CID: 5359232

Immune cell profiling in atherosclerosis: role in research and precision medicine

Fernandez, Dawn M; Giannarelli, Chiara
Inflammation is intimately involved at all stages of atherosclerosis and remains a substantial residual cardiovascular risk factor in optimally treated patients. The proof of concept that targeting inflammation reduces cardiovascular events in patients with a history of myocardial infarction has highlighted the urgent need to identify new immunotherapies to treat patients with atherosclerotic cardiovascular disease. Importantly, emerging data from new clinical trials show that successful immunotherapies for atherosclerosis need to be tailored to the specific immune alterations in distinct groups of patients. In this Review, we discuss how single-cell technologies - such as single-cell mass cytometry, single-cell RNA sequencing and cellular indexing of transcriptomes and epitopes by sequencing - are ideal for mapping the cellular and molecular composition of human atherosclerotic plaques and how these data can aid in the discovery of new precise immunotherapies. We also argue that single-cell data from studies in humans need to be rigorously validated in relevant experimental models, including rapidly emerging single-cell CRISPR screening technologies and mouse models of atherosclerosis. Finally, we discuss the importance of implementing single-cell immune monitoring tools in early phases of drug development to aid in the precise selection of the target patient population for data-driven translation into randomized clinical trials and the successful translation of new immunotherapies into the clinic.
PMID: 34267377
ISSN: 1759-5010
CID: 4938942

Single cell analyses to understand the immune continuum in atherosclerosis

Hill, Christopher A; Fernandez, Dawn M; Giannarelli, Chiara
Atherosclerosis is initiated by the accumulation of lipids in the arterial wall that trigger a complex and poorly understood network of inflammatory processes. At the same time, recent clinical findings reveal that targeting specific immune alterations in patients with cardiovascular disease (CVD) represents a promising approach to preventing recurrent cardiovascular events. In order to achieve these tailored therapies, it is critical to resolve the heterogenous environment of the atherosclerotic lesion and decipher the complex structural and functional changes which immune cells undergo throughout disease progression. Recently, single-cell approaches including single cell mass cytometry by time of flight (CyTOF), single cell RNA sequencing (scRNA-seq) and Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq) have emerged as valuable tools in resolving cellular plasticity within atherosclerotic lesions. In this review, we will discuss the most important insights that have been gleaned from the application of these single-cell approaches to validated experimental models of atherosclerosis. Additionally, as clinical progress in treatment of the disease depends on the translation of discoveries to human tissues, we will also examine the challenges associated with the application of single-cell approaches to human vascular tissue and the discoveries made by the initial efforts in this direction. Finally, we will analyze the advantages and limitations of dissociative single-cell approaches and how novel in-situ technologies could advance the field by allowing for the investigation of individual cells while preserving the heterogenous architecture of the atherosclerotic lesion.
PMID: 33934886
ISSN: 1879-1484
CID: 4865862