To Örebro University

oru.seÖrebro University Publications
Change search
Link to record
Permanent link

Direct link
Alternative names
Publications (10 of 33) Show all publications
Hayderi, A., Sirsjö, A., Salihovic, S. & Ljungberg, L. (2023). Interferon gamma reprograms glutamine metabolic pathways in human aortic smooth muscle cells. Paper presented at 91st Annual Meeting of the European-Atherosclerosis-Society (EAS 2023), Mannheim, Germany, May 21-24, 2023. Atherosclerosis, 379(Suppl. 1), S8-S8, Article ID P024.
Open this publication in new window or tab >>Interferon gamma reprograms glutamine metabolic pathways in human aortic smooth muscle cells
2023 (English)In: Atherosclerosis, ISSN 0021-9150, E-ISSN 1879-1484, Vol. 379, no Suppl. 1, p. S8-S8, article id P024Article in journal, Meeting abstract (Other academic) Published
Abstract [en]

Background and Aims: Cells within atherosclerotic lesions have a higher glutamine demand than cells in healthy vessel although glutaminase, the enzyme converting glutamine to glutamate, is significantly downregulated in human carotid lesions. This may suggest rewiring of glutamine metabolic pathways in atherosclerotic lesions, caused by infiltrating immune cells and or their cytokines. Here we aimed at exploring the enzymes and transporters involved in glutamine metabolism in human carotid atherosclerotic tissues and aortic smooth muscle cells (hAoSMCs) exposed to interferon gamma.

Methods: Protein and mRNA from interferon gamma-treated hAoSMCs were subjected to Western blot or qRT-PCR for quantification of enzymes and transporters involved in glutamine metabolism. H2DCFDA probe was utilized for detection of intracellular reactive oxygen species (ROS) using flow cytometry. The expression of these enzymes and transporters was also evaluated in human carotid lesions (GEO accession: GSE43292).

Results: Interferon-treated hAoSMCs display a significantly lower expression of glutaminase followed by an increase in the expression of glutamine transporters, glutamine synthetase and glutamine-fructose-6-phosphate transaminase-1 (GFPT1). The level of ROS and the expression of enzymes involved in de novo synthesis of glutathione are elevated in interferon-treated cells. A similar expression pattern for these genes, expect for GFPT1, is also evident in human carotid lesions where glutaminase mRNA shows a strong positive correlation with SMC markers and a strong negative correlation with macrophage markers.

Conclusions: Glutamine metabolism is disrupted in human carotid lesions and interferon gamma alters glutamine metabolism in hAoSMCs, which may favor the production of UDP-GlcNAc and reactive oxygen species.

Place, publisher, year, edition, pages
Elsevier, 2023
National Category
Cardiac and Cardiovascular Systems
Identifiers
urn:nbn:se:oru:diva-109548 (URN)001060595800310 ()
Conference
91st Annual Meeting of the European-Atherosclerosis-Society (EAS 2023), Mannheim, Germany, May 21-24, 2023
Available from: 2023-11-06 Created: 2023-11-06 Last updated: 2023-11-06Bibliographically approved
Zegeye, M. M., Matic, L., Lengquist, M., Hayderi, A., Grenegård, M., Hedin, U., . . . Kumawat, A. K. (2023). Interleukin-6 trans-signaling induced laminin switch contributes to reduced trans-endothelial migration of granulocytic cells. Atherosclerosis, 371, 41-53
Open this publication in new window or tab >>Interleukin-6 trans-signaling induced laminin switch contributes to reduced trans-endothelial migration of granulocytic cells
Show others...
2023 (English)In: Atherosclerosis, ISSN 0021-9150, E-ISSN 1879-1484, Vol. 371, p. 41-53Article in journal (Refereed) Published
Abstract [en]

BACKGROUND AND AIMS: Laminins are essential components of the endothelial basement membrane, which predominantly contains LN421 and LN521 isoforms. Regulation of laminin expression under pathophysiological conditions is largely unknown. In this study, we aimed to investigate the role of IL-6 in regulating endothelial laminin profile and characterize the impact of altered laminin composition on the phenotype, inflammatory response, and function of endothelial cells (ECs).

METHODS: HUVECs and HAECs were used for in vitro experiments. Trans-well migration experiments were performed using leukocytes isolated from peripheral blood of healthy donors. The BiKE cohort was used to assess expression of laminins in atherosclerotic plaques and healthy vessels. Gene and protein expression was analyzed using Microarray/qPCR and proximity extension assay, ELISA, immunostaining or immunoblotting techniques, respectively.

RESULTS: Stimulation of ECs with IL-6+sIL-6R, but not IL-6 alone, reduces expression of laminin α4 (LAMA4) and increases laminin α5 (LAMA5) expression at the mRNA and protein levels. In addition, IL-6+sIL-6R stimulation of ECs differentially regulates the release of several proteins including CXCL8 and CXCL10, which collectively were predicted to inhibit granulocyte transmigration. Experimentally, we demonstrated that granulocyte migration is inhibited across ECs pre-treated with IL-6+sIL-6R. In addition, granulocyte migration across ECs cultured on LN521 was significantly lower compared to LN421. In human atherosclerotic plaques, expression of endothelial LAMA4 and LAMA5 is significantly lower compared to control vessels. Moreover, LAMA5-to-LAMA4 expression ratio was negatively correlated with granulocytic cell markers (CD177 and myeloperoxidase (MPO)) and positively correlated with T-lymphocyte marker CD3.

CONCLUSIONS: We showed that expression of endothelial laminin alpha chains is regulated by IL-6 trans-signaling and contributes to inhibition of trans-endothelial migration of granulocytic cells. Further, expression of laminin alpha chains is altered in human atherosclerotic plaques and is related to intra-plaque abundance of leukocyte subpopulations.

Place, publisher, year, edition, pages
Elsevier, 2023
Keywords
Atherosclerosis, Chronic inflammation, Diapedesis, Endothelial basement membrane, Plaque stability
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:oru:diva-105279 (URN)10.1016/j.atherosclerosis.2023.03.010 (DOI)000994230600001 ()36996622 (PubMedID)2-s2.0-85150807616 (Scopus ID)
Funder
Knowledge Foundation, 2018-0035Stiftelsen Gamla Tjänarinnor, 2019-00851
Available from: 2023-03-31 Created: 2023-03-31 Last updated: 2023-06-07Bibliographically approved
Zegeye, M., Ljungberg, L., Nakka, S., Andersson, J., Söderberg, S., Kumawat, A. & Sirsjö, A. (2023). Soluble LDL-receptor is induced by TNF-Α and inhibits hepatocytic clearance of LDL-cholesterol. Paper presented at 91st Annual Meeting of the European-Atherosclerosis-Society (EAS 2023), Mannheim, Germany, May 21-24, 2023. Atherosclerosis, 379(Suppl. 1), S61-S61, Article ID P195.
Open this publication in new window or tab >>Soluble LDL-receptor is induced by TNF-Α and inhibits hepatocytic clearance of LDL-cholesterol
Show others...
2023 (English)In: Atherosclerosis, ISSN 0021-9150, E-ISSN 1879-1484, Vol. 379, no Suppl. 1, p. S61-S61, article id P195Article in journal, Meeting abstract (Other academic) Published
Abstract [en]

Background and Aims: LDL-c is cleared from the circulation mainly by hepatic LDL-receptor mediated endocytosis. Defective LDL-c clearance and hence its elevation in circulation is one of the risk factors for myocardial infarction (MI). A soluble LDL-R (sLDL-R) exists in human plasma and exhibits strong correlation with circulating LDL-c and conditions that promote chronic inflammation. However, the mechanistic interplay between sLDL-R, inflammation and MI remains to be investigated.

Methods: In vitro studies using HepG2 cells treated with TNF-α, and a nested case-control study was conducted to investigate the relationship between plasma sLDL-R, TNF-α and risk of future MI.

Results: Stimulation of HepG2 cells with TNF-α induces release of sLDL-R with limited effect on surface expression of LDL-R. TNF-α induces gene expression of peptidases ADAM17 and MMP14 in HepG2 cells, and inhibition of ADAM17 and MMP-14 significantly reduces the TNF-α induced sLDL-R release. Although TNF-α treatment of HepG2 cells has limited effect on LDL-c endocytosis, HepG2 cells incubated with recombinant sLDL-R showed reduced LDL-c uptake in a dose-dependent manner. In a nested case-control study, baseline sLDL-R in plasma was positively correlated with plasma total cholesterol level. Further, a 2-fold increase in plasma sLDL-R was associated with 2.1x higher risk of future MI. Using mediation analyses, we determined that significant proportion of the association is mediated by elevation in plasma cholesterol level.

Conclusions: Our study suggests that sLDL-R is generated by TNF-α via membrane shedding. Further, an increase in sLDL-R could inhibit hepatic clearance of LDL-c increasing its half-life in the circulation and contributing to the pathogenesis of MI.

Place, publisher, year, edition, pages
Elsevier, 2023
National Category
Cardiac and Cardiovascular Systems
Identifiers
urn:nbn:se:oru:diva-109565 (URN)001060595800481 ()
Conference
91st Annual Meeting of the European-Atherosclerosis-Society (EAS 2023), Mannheim, Germany, May 21-24, 2023
Available from: 2023-11-06 Created: 2023-11-06 Last updated: 2023-11-06Bibliographically approved
Zegeye, M. M., Nakka, S. S., Andersson, J. S. O., Söderberg, S., Ljungberg, L., Kumawat, A. K. & Sirsjö, A. (2023). Soluble LDL-receptor is induced by TNF-α and inhibits hepatocytic clearance of LDL-cholesterol. Journal of Molecular Medicine, 101(12), 1615-1626
Open this publication in new window or tab >>Soluble LDL-receptor is induced by TNF-α and inhibits hepatocytic clearance of LDL-cholesterol
Show others...
2023 (English)In: Journal of Molecular Medicine, ISSN 0946-2716, E-ISSN 1432-1440, Vol. 101, no 12, p. 1615-1626Article in journal (Refereed) Published
Abstract [en]

Defective LDL-C clearance and hence its elevation in the circulation is an established risk factor for cardiovascular diseases (CVDs) such as myocardial infarction (MI). A soluble LDL-receptor (sLDL-R) has been detected in human plasma which correlates strongly with circulating LDL-C and classical conditions that promote chronic inflammation. However, the mechanistic interplay between sLDL-R, inflammation, and CVDs remains to be investigated. Here, we report that stimulation of HepG2 cells with TNF-α induces the release of sLDL-R into culture supernatants. In addition, TNF-α induces gene expression of peptidases ADAM-17 and MMP-14 in HepG2 cells, and inhibiting these peptidases using TMI 1 significantly reduces the TNF-α induced sLDL-R release. We found that a soluble form of recombinant LDL-R (100 nM) can strongly bind to LDL-C and form a stable complex (KD = E-12). Moreover, incubation of HepG2 cells with this recombinant LDL-R resulted in reduced LDL-C uptake in a dose-dependent manner. In a nested case-control study, we found that baseline sLDL-R in plasma is positively correlated with plasma total cholesterol level. Furthermore, a twofold increase in plasma sLDL-R was associated with a 55% increase in the risk of future MI [AOR = 1.55 (95% CI = 1.10-2.18)]. Nevertheless, mediation analyses revealed that a significant proportion of the association is mediated by elevation in plasma cholesterol level (indirect effect β = 0.21 (95% CI = 0.07-0.38). Collectively, our study shows that sLDL-R is induced by a pro-inflammatory cytokine TNF-α via membrane shedding. Furthermore, an increase in sLDL-R could inhibit hepatic clearance of LDL-C increasing its half-life in the circulation and contributing to the pathogenesis of MI. KEY MESSAGES: TNF-α causes shedding of hepatocytic LDL-R through induction of ADAM-17 and MMP-14. sLDL-R binds strongly to LDL-C and inhibits its uptake by hepatocytic cells. Plasma sLDL-R is positively correlated with TNF-α and cholesterol. Plasma sLDL-R is an independent predictor of myocardial infarction (MI). Plasma cholesterol mediates the association between sLDL-R and MI.

Place, publisher, year, edition, pages
Springer, 2023
Keywords
ADAM-17, Chronic inflammation, Hypercholesterolemia, MMP-14, Mediation analyses, Myocardial infarction
National Category
Cardiac and Cardiovascular Systems
Identifiers
urn:nbn:se:oru:diva-109360 (URN)10.1007/s00109-023-02379-4 (DOI)001088058800001 ()37861809 (PubMedID)2-s2.0-85174533259 (Scopus ID)
Funder
Örebro UniversityKnowledge Foundation, 2018-0035 2021-0038
Available from: 2023-10-23 Created: 2023-10-23 Last updated: 2024-01-12Bibliographically approved
Kumawat, A. K., Zegeye, M. M., Paramel Varghese, G., Baumgartner, R., Gisterå, A., Amegavie, O., . . . Sirsjö, A. (2022). Inhibition of IL17A Using an Affibody Molecule Attenuates Inflammation in ApoE-Deficient Mice. Frontiers in Cardiovascular Medicine, 9, Article ID 831039.
Open this publication in new window or tab >>Inhibition of IL17A Using an Affibody Molecule Attenuates Inflammation in ApoE-Deficient Mice
Show others...
2022 (English)In: Frontiers in Cardiovascular Medicine, E-ISSN 2297-055X, Vol. 9, article id 831039Article in journal (Refereed) Published
Abstract [en]

The balance between pro- and anti-inflammatory cytokines released by immune and non-immune cells plays a decisive role in the progression of atherosclerosis. Interleukin (IL)-17A has been shown to accelerate atherosclerosis. In this study, we investigated the effect on pro-inflammatory mediators and atherosclerosis development of an Affibody molecule that targets IL17A. Affibody molecule neutralizing IL17A, or sham were administered in vitro to human aortic smooth muscle cells (HAoSMCs) and murine NIH/3T3 fibroblasts and in vivo to atherosclerosis-prone, hyperlipidaemic ApoE(-/-) mice. Levels of mediators of inflammation and development of atherosclerosis were compared between treatments. Exposure of human smooth muscle cells and murine NIH/3T3 fibroblasts in vitro to alpha IL-17A Affibody molecule markedly reduced IL6 and CXCL1 release in supernatants compared with sham exposure. Treatment of ApoE(-/-) mice with alpha IL-17A Affibody molecule significantly reduced plasma protein levels of CXCL1, CCL2, CCL3, HGF, PDGFB, MAP2K6, QDPR, and splenocyte mRNA levels of Ccxl1, Il6, and Ccl20 compared with sham exposure. There was no significant difference in atherosclerosis burden between the groups. In conclusion, administration of alpha IL17A Affibody molecule reduced levels of pro-inflammatory mediators and attenuated inflammation in ApoE(-/-) mice.

Place, publisher, year, edition, pages
Frontiers Media S.A., 2022
Keywords
CXCL1, Affibody molecule, atherosclerosis, apolipoprotein, E-deficient (ApoE(-/-)) mice, human aortic smooth muscle cells
National Category
Cardiac and Cardiovascular Systems
Identifiers
urn:nbn:se:oru:diva-97689 (URN)10.3389/fcvm.2022.831039 (DOI)000768021300001 ()35282365 (PubMedID)2-s2.0-85138499316 (Scopus ID)
Funder
Knowledge Foundation, KKHOEG 20150245Swedish Heart Lung FoundationNovo Nordisk, NNF15CC0018346Swedish Research CouncilKnut and Alice Wallenberg Foundation
Note

Funding agencies:

ALF/The Stockholm Region

MedTechLabs

Available from: 2022-02-25 Created: 2022-02-25 Last updated: 2023-12-08Bibliographically approved
Lindkvist, M., Zegeye, M. M., Grenegård, M. & Ljungberg, L. (2022). Pleiotropic, Unique and Shared Responses Elicited by IL-6 Family Cytokines in Human Vascular Endothelial Cells. International Journal of Molecular Sciences, 23(3), Article ID 1448.
Open this publication in new window or tab >>Pleiotropic, Unique and Shared Responses Elicited by IL-6 Family Cytokines in Human Vascular Endothelial Cells
2022 (English)In: International Journal of Molecular Sciences, ISSN 1661-6596, E-ISSN 1422-0067, Vol. 23, no 3, article id 1448Article in journal (Refereed) Published
Abstract [en]

Vascular endothelial cells express glycoprotein 130 (gp130), which is utilized as a signaling receptor by cytokines in the interleukin-6 (IL-6) family. Several IL-6 family cytokines can be found in the circulatory system during physiological or pathological conditions, and may influence endothelial function and response. This study evaluated and compared the cellular and molecular responses induced by IL-6 family cytokines in human endothelial cells. A proteomic analysis showed that IL-6 family cytokines induce the release of a range of proteins from endothelial cells, such as C-C motif chemokine ligand 23, hepatocyte growth factor, and IL-6. Pathway analysis indicated that gp130-signaling in endothelial cells regulates several functions related to angiogenesis and immune cell recruitment. The present investigation also disclosed differences and similarities between different IL-6 family cytokines in their ability to induce protein release and regulate gene expression and intracellular signaling, in regards to which oncostatin M showed the most pronounced effect. Further, this study showed that soluble gp130 preferentially blocks trans-signaling-induced responses, but does not affect responses induced by classic signaling. In conclusion, IL-6 family cytokines induce both specific and overlapping molecular responses in endothelial cells, and regulate genes and proteins involved in angiogenesis and immune cell recruitment.

Place, publisher, year, edition, pages
MDPI, 2022
Keywords
CCL23, cardiotrophin-1 (CT-1), chemotaxis, ciliary neurotrophic factor (CNTF), hepatocyte growth factor (HGF), interleukin-11 (IL-11), leukemia inhibitory factor (LIF)
National Category
Cell and Molecular Biology
Identifiers
urn:nbn:se:oru:diva-97530 (URN)10.3390/ijms23031448 (DOI)000754525300001 ()35163371 (PubMedID)2-s2.0-85123608892 (Scopus ID)
Available from: 2022-02-15 Created: 2022-02-15 Last updated: 2022-02-21Bibliographically approved
Hayderi, A., Ljungberg, L., Kumawat, A. K. & Sirsjö, A. (2021). Function of viperin in human aortic SMCS and its relevance for atherosclerosis. Atherosclerosis, 331, E88-E88
Open this publication in new window or tab >>Function of viperin in human aortic SMCS and its relevance for atherosclerosis
2021 (English)In: Atherosclerosis, ISSN 0021-9150, E-ISSN 1879-1484, Vol. 331, p. E88-E88Article in journal, Meeting abstract (Other academic) Published
Abstract [en]

Background and Aims: During atherosclerosis, activated vascular cells secrete chemokines to recruit leukocytes to the subendothelial space. Activated leukocytes, in particular activated T lymphocytes express IFN-γ, which affects the expression of numerous genes, including the antiviral protein viperin, in vascular cells. Viperin is expressed in endothelial cells of human carotid plaques but its relevance for vascular physiology and/or pathophysiology has not been established (Olofsson et al., 2005). We aimed at studying the function of viperin in human AoSMCs.

Methods: Immunostaining of human carotid plaques was performed to determine the expression and localization of viperin in carotid plaques. To study the role of viperin in regulation of vascular inflammation, viperin was silenced using siRNA in cultured human AoSMCs, prior to stimulation with IFN-γ. Release of inflammatory mediators was analyzed using OLINK proteomics and ELISA, and the gene expression was analyzed by qRT-PCR.

Results: Viperin is expressed in endothelial cells, macrophages and smooth muscle cells in human carotid plaques. OLINK data analysis revealed reduction in release of CCL3, CXCL9, CXCL10 and CXCL11 by AoSMCs that were targeted with anti-viperin siRNA prior to stimulation with IFN-γ. Using ELISA and qRT-PCR, we could confirm the reduction of IFN-γ-induced CXCL10 and CXCL11 in viperin knockdown AoSMCs.

Conclusions: Viperin is expressed in human carotid plaques and seem to be involved in regulating the expression of CXCL10 and CXCL11 in AoSMCs. Thus, it may play a role in recruitment of T lymphocytes to the subendothelial space in atherosclerotic plaques.

Place, publisher, year, edition, pages
Elsevier, 2021
National Category
Cardiac and Cardiovascular Systems
Identifiers
urn:nbn:se:oru:diva-94655 (URN)10.1016/j.atherosclerosis.2021.06.256 (DOI)000693712700253 ()
Available from: 2021-09-30 Created: 2021-09-30 Last updated: 2021-09-30Bibliographically approved
Zegeye, M. M., Andersson, J. S. O., Wennberg, P., Repsilber, D., Sirsjö, A. & Ljungberg, L. (2021). IL-6 as a Mediator of the Association Between Traditional Risk Factors and Future Myocardial Infarction: A Nested Case-Control Study. Arteriosclerosis, Thrombosis and Vascular Biology, 41(4), 1570-1579
Open this publication in new window or tab >>IL-6 as a Mediator of the Association Between Traditional Risk Factors and Future Myocardial Infarction: A Nested Case-Control Study
Show others...
2021 (English)In: Arteriosclerosis, Thrombosis and Vascular Biology, ISSN 1079-5642, E-ISSN 1524-4636, Vol. 41, no 4, p. 1570-1579Article in journal (Refereed) Published
Abstract [en]

OBJECTIVE: Studies elucidating the importance of IL (interleukin)-6 trans-signaling associated with risk of future myocardial infarction (MI) are scarce. Additionally, whether elevation in IL-6 explains part of the association between traditional risk factors and future MI has not been explored.

Approach and Results: We conducted a nested case-control study including a total of 584 participants (292 cases and 292 controls) from Västerbotten Intervention Programme and MONICA (Multinational Monitoring of Trends and Determinants in Cardiovascular Disease) cohorts. At baseline, plasma cholesterol levels were measured, and clinical characteristics of participants were collected. In this study, we measured the plasma concentration of IL-6, sIL-6R (soluble IL-6 receptor), and sgp130 (soluble-gp130). To estimate extent of IL-6 trans-signaling, we estimated plasma concentration of a novel biomarker, the IL-6 binary complex. IL-6 binary complex concentration was significantly elevated in participants who experienced MI compared with those who did not. Univariate analyses showed that a 2-fold increase in IL-6 binary complex was associated with 2.45× higher risk of future MI (95% CI relative risk, 1.65-3.66, P<0.001). Receiver operating characteristics analyses revealed that the predictive performance of IL-6 binary complex (area under the curve, 0.614) was equivalent to that of IL-6 (area under the curve, 0.603). Furthermore, using Process mediation analyses tool, we found statistically significant indirect effect of smoking and hypertension on future MI that is mediated through increased IL-6 binary complex or plasma IL-6.

CONCLUSIONS: IL-6 and IL-6 binary complex concentration in plasma were significantly associated with future MI. Our data additionally imply that both the elevated plasma IL-6, and the IL-6 binary complex concentration could partly explain, and, thus, might hypothetically be functionally associated with the increased risk of MI in smokers and hypertensive participants.

Place, publisher, year, edition, pages
Lippincott Williams & Wilkins, 2021
Keywords
Acute coronary syndrome, atherosclerosis, biomarker, hypertension, smoking
National Category
Cardiac and Cardiovascular Systems
Identifiers
urn:nbn:se:oru:diva-90144 (URN)10.1161/ATVBAHA.120.315793 (DOI)000639317700034 ()33657883 (PubMedID)
Funder
Knowledge Foundation, Dnr20180035Swedish Research Council, VR 2017-0650
Note

Funding Agency:

Foundation for Old Servants 2019-00851 2018-00654 2017-00579

Available from: 2021-03-05 Created: 2021-03-05 Last updated: 2021-05-24Bibliographically approved
Zegeye, M. M., Kumawat, A. K., Matic, L., Lengquist, M., Hyderi, A., Hedin, U., . . . Ljungberg, L. (2021). IL-6 trans-signaling regulates vascular endothelial laminin profile and inflammatory responses: Possible mechanism for immune cell recruitment during atherosclerosis?. Atherosclerosis, 331, E61-E61
Open this publication in new window or tab >>IL-6 trans-signaling regulates vascular endothelial laminin profile and inflammatory responses: Possible mechanism for immune cell recruitment during atherosclerosis?
Show others...
2021 (English)In: Atherosclerosis, ISSN 0021-9150, E-ISSN 1879-1484, Vol. 331, p. E61-E61Article in journal, Meeting abstract (Other academic) Published
Abstract [en]

Background and Aims: We aimed to investigate the role of IL-6 trans-signaling in regulating the release of endothelial inflammatory mediators and endothelial basement mebrane proteins, laminins. We also aimed to investigate the laminin composition in atherosclerotic plaques in relation to the immune cell content.

Methods: HUVECs were cultured for in vitro experiments. The BiKE cohort was used to assess expression of laminins in atherosclerotic plaques and healthy vessels. Gene and protein expression was analysed using Microarray/qPCR and proximity extension assay, immunostaining or immunoblotting techniques respectively.

Results: The release of 20 inflammatory proteins from ECs was significantly altered (3 downregulated, 17 upregulated) in response to stimulation with IL-6 and sIL-6R (p-value <0.05, FDR 5%). Ingenuity pathway analyses predicted that these proteins enhance lymphocyte binding and transmigration while inhibiting transmigration of granulocytes. ECs treated with combination of IL-6 and sIL-6R upregulated expression of LAMA5 while downregulating LAMA4. Transcriptomic and proteomic analyses showed that both endothelial LAMA4 and LAMA5 were significantly lower in atherosclerotic plaques compared to healthy vessels. In addition, expression of endothelial LAMA5 was significantly lower in plaques from symptomatic patients compared to those from asymptomatic patients. We also showed that endothelial laminins are negatively correlated with immune cell markers.

Conclusions: Our findings suggest that IL-6 trans-signaling regulates endothelial inflammatory proteins and laminin production that enhance binding and trans-migration of lymphocytes. In the context of atherosclerosis, expression of laminin alpha chains is altered and also appeared to be associated with plaque stability. Our data also revealed a relationship between laminin chains and immune cell content in atherosclerotic plaques.

Place, publisher, year, edition, pages
Elsevier, 2021
National Category
Cardiac and Cardiovascular Systems
Identifiers
urn:nbn:se:oru:diva-94663 (URN)10.1016/j.atherosclerosis.2021.06.175 (DOI)000693712700176 ()
Available from: 2021-09-30 Created: 2021-09-30 Last updated: 2021-09-30Bibliographically approved
Paramel Varghese, G., Karadimou, G., Göthlin Eremo, A., Ljungberg, L., Hedin, U., Olofsson, P. S., . . . Fransén, K. (2020). Expression of CARD8 in human atherosclerosis and its regulation of inflammatory proteins in human endothelial cells. Scientific Reports, 10(1), Article ID 19108.
Open this publication in new window or tab >>Expression of CARD8 in human atherosclerosis and its regulation of inflammatory proteins in human endothelial cells
Show others...
2020 (English)In: Scientific Reports, E-ISSN 2045-2322, Vol. 10, no 1, article id 19108Article in journal (Refereed) Published
Abstract [en]

The Caspase activation and recruitment domain 8 (CARD8) protein is a component of innate immunity and overexpression of CARD8 mRNA was previously identified in atherosclerosis. However, very little is known about the regulation of CARD8 in endothelial cells and atherosclerosis. The aim of this study was to investigate CARD8 in the regulation of cytokine and chemokine expression in endothelial cells. Sections of human atherosclerotic lesions and non-atherosclerotic arteries were immunostained for CARD8 protein. Expression of CARD8 was correlated to mediators of inflammation in atherosclerotic lesions using Biobank of Karolinska Endarterectomies microarray data. The CARD8 mRNA was knocked-down in human umbilical vein endothelial cells (HUVECs) in vitro, followed by quantitative RT-PCR analysis and OLINK Proteomics. Endothelial and smooth muscle cells in arterial tissue expressed CARD8 and CARD8 correlated with vWF, CD163 and the expression of inflammatory genes, such as CXCL1, CXCL6 and PDGF-A in plaque. Knock-down of CARD8 in HUVECs significantly altered proteins involved in inflammatory response, such as CXCL1, CXCL6, PDGF-A, MCP-1 and IL-6. The present study suggest that CARD8 regulate the expression of cytokines and chemokines in endothelial cells and atherosclerotic lesions, suggesting that CARD8 plays a significant role in endothelial activation.

Place, publisher, year, edition, pages
Nature Publishing Group, 2020
Keywords
CARD8, atherosclerosis, chemokines, chronic inflammation, diagnostic markers, innate immunity, interleukins, mechanisms of disease
National Category
Cell and Molecular Biology Cardiac and Cardiovascular Systems
Research subject
Medicine
Identifiers
urn:nbn:se:oru:diva-87251 (URN)10.1038/s41598-020-73600-4 (DOI)000587146400001 ()33154409 (PubMedID)2-s2.0-85095132771 (Scopus ID)
Funder
Knowledge FoundationSwedish Heart Lung Foundation
Note

An Author Correction to this article was published in Sci Rep. 2021 Apr 19;11(1):8852. doi: 10.1038/s41598-021-88467-2. 

Available from: 2020-11-07 Created: 2020-11-07 Last updated: 2022-09-15Bibliographically approved
Organisations
Identifiers
ORCID iD: ORCID iD iconorcid.org/0000-0003-4253-3369

Search in DiVA

Show all publications