Abstract

Atherosclerosis is a chronic inflammatory disease characterized by a complex interplay between innate and adaptive immunity. Dendritic cells (DCs) play a key role in T-cell activation and regulation by promoting a tolerogenic environment through the expression of the immunosuppressive enzyme indoleamine 2,3-dioxygenase (IDO), an intracellular enzyme involved in tryptophan catabolism. IDO expression and activity was analyzed in monocytes derived DCs (MDDCs) from non-ST segment elevation myocardial infarction (NSTEMI) patients, stable angina (SA) patients and healthy controls (HC) by real-time quantitative polymerase chain reaction (RT-qPCR) before and after in vitro maturation with lipopolysaccharide (LPS). The amount of tryptophan catabolite; kynurenine; was evaluated in the culture supernatants of mature-MDDCs by ELISA assay. Autologous mixed lymphocyte reaction (MLR) between mature-MDDCs and naïve T-cells was carried out to study the differentiation towards T-helper 1 (Th1) and induced regulatory T-cells (iTreg). Analysis of IDO mRNA transcripts in mature-MDDCs revealed a significant reduction in cells isolated from NSTEMI (625.0 ± 128.2; mean ± SEM) as compared with those from SA (958.5 ± 218.3; p = 0.041) and from HC (1183.6 ± 231.6; p = 0.034). Furthermore; the concentration of kynurenine was lower in NSTEMI patients (2.78 ± 0.2) and SA (2.98 ± 0.25) as compared with HC (5.1 ± 0.69 ng/mL; p = 0.002 and p = 0.016; respectively). When IDO-competent mature-MDDCs were co-cultured with allogeneic naïve T-cells, the ratio between the percentage of generated Th1 and iTreg was higher in NSTEMI (4.4 ± 2.9) than in SA (1.8 ± 0.6; p = 0.056) and HC (0.9 ± 0.3; p = 0.008). In NSTEMI, the tolerogenic mechanism of the immune response related to IDO production by activated MDDCs is altered, supporting their role in T-cell dysregulation.

Highlights

  • The early outcome of acute coronary syndromes (ACS) has recently considerably improved, cardiovascular diseases still represent the leading cause of mortality worldwide.An adaptive immunity imbalance, mostly involving CD4+ T-cell subsets, has been documented among ACS with systemic evidence of inflammation [1]

  • We studied markers of monocyte-derived Dendritic cells (DCs) (MDDC) maturation, the expression of IDO and the kynurenine pathway in monocytes derived DCs (MDDCs) from patients presenting with non-ST segment elevation myocardial infarction (NSTEMI), stable angina (SA) and healthy controls (HC) after stimulation with lipopolysaccharide (LPS)

  • MDDC Maturation was Altered in NSTEMI Patients 2.1

Read more

Summary

Introduction

Mostly involving CD4+ T-cell subsets, has been documented among ACS with systemic evidence of inflammation [1]. T-cells from ACS patients show T-cell receptor (TCR) signaling abnormalities leading to enhanced immune response and altered T helper differentiation [5,6]. Atherosclerotic lesions contain abundant innate immune cells, including antigen presenting cells (APCs) that take part in initiation, progression and destabilization of the atherosclerotic plaque [7]. Dendritic cells (DCs) are a heterogeneous pool of professional APCs with the ability to sense a wide array of stimuli and translate innate into adaptive immunity by directing an appropriate T-cell response [8,9]

Methods
Results
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call