501
|
Newman JL, Stone JR. Immune checkpoint inhibition alters the inflammatory cell composition of human coronary artery atherosclerosis. Cardiovasc Pathol 2019; 43:107148. [PMID: 31518915 DOI: 10.1016/j.carpath.2019.107148] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 07/19/2019] [Accepted: 08/09/2019] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibition (ICI) has emerged as a promising new approach to treat malignancy. Such therapies can result in autoimmune-related complications such as myocarditis and hepatitis. The impact of ICI on sites of preexisting chronic inflammation has been less clear. Atherosclerosis is a chronic vascular disease with a significant inflammatory component. METHODS To determine the effect of ICI on the inflammatory infiltrate in coronary artery atherosclerotic plaques, 11 patients who had recently been treated with ICI and subsequently underwent autopsy were matched with 11 cancer patients who had not received ICI treatment. The amount of CD3+ T-lymphocytes, CD8+ cytotoxic T-lymphocytes, and CD68+ macrophages and the ratios of the various cell types in the coronary artery atherosclerotic plaques were compared. RESULTS There was no significant difference in the absolute numbers of CD3+, CD8+, or CD68+ cells in the atherosclerotic plaques. In the plaques of the ICI-treated patients, there was a significant increase in the ratio of CD3+ cells to CD68+ cells (CD3/CD68) (P=.002) and a trend towards an increased CD8/CD68 ratio. The increased CD3/CD68 ratio in the ICI-treated patients resulted in 6 of the 11 patients having lymphocyte-predominate inflammation in contrast to the macrophage-predominate inflammation typically found in atherosclerotic plaques. CONCLUSIONS These findings indicate that ICI alters the inflammatory composition of human atherosclerotic plaque and, thus, may influence plaque progression and/or clinical coronary events. SUMMARY In cancer patients, treatment with immune checkpoint inhibition is associated with an altered inflammatory cell composition in coronary artery atherosclerotic plaques with an increased ratio of CD3+ T cells to CD68+ macrophages. Thus, immune checkpoint inhibition may influence plaque progression and/or clinical coronary events.
Collapse
Affiliation(s)
- Justine L Newman
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA.
| |
Collapse
|
502
|
Wang Y, Zhang Y, Wang Z, Zhang J, Qiao RR, Xu M, Yang N, Gao L, Qiao H, Gao M, Cao F. Optical/MRI dual-modality imaging of M1 macrophage polarization in atherosclerotic plaque with MARCO-targeted upconversion luminescence probe. Biomaterials 2019; 219:119378. [PMID: 31382209 DOI: 10.1016/j.biomaterials.2019.119378] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2019] [Revised: 07/15/2019] [Accepted: 07/24/2019] [Indexed: 02/09/2023]
Abstract
Pro-inflammatory M1 macrophage is identified as a prominent component initializing the progress of vulnerable atherosclerotic plaque. Here, we constructed anti-MARCO NaGdF4:Yb,Er@NaGdF4 upconversion nanoparticles (UCNPs) by conjugating polyclonal MARCO antibody to the surface of NaGdF4:Yb,Er@NaGdF4via condensation reaction. UCNPs displayed highly mono-dispersion with average sizes of 26.7 ± 0.8 nm and favorable biocompatibility. In vivo upconversion optical imaging revealed that distinctive fluorescence signal could be observed in the regions of carotid artery 10 min post-injection, reached peak value at 1 h and decreased back to baseline at 24 h post-injection. The carotid artery wall demonstrated high signal intensity on T1-weighted MR images after anti-MARCO UCNPs injection, as determined by 7.0T MRI. Immunofluorescence staining of tissue section of carotid artery revealed that MARCO was highly abundant in shoulder regions of plaque. Anti-MARCO UCNPs is a promising optical/MRI dual-modality imaging probe which can non-invasively reflect M1 phenotype macrophages behavior in vivo.
Collapse
Affiliation(s)
- Yabin Wang
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China; Department of Cardiology, Chinese Eastern Theatre Naval Hospital, Zhejiang, 316000, China
| | - Yan Zhang
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China; TEDA International Cardiovascular Hospital, Tianjin, 300457, China
| | - Zhao Wang
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Jibin Zhang
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Rui Rui Qiao
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China
| | - Mengqi Xu
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Ning Yang
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Lei Gao
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Hongyu Qiao
- Department of Pediatric, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Mingyuan Gao
- Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing, 100190, China.
| | - Feng Cao
- Department of Cardiology & National Clinical Research Center of Geriatric Disease, 2nd Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
503
|
Del Porto F, Cifani N, Proietta M, Dezi T, Tritapepe L, Raffa S, Micaloni A, Taurino M. Lag3 + regulatory T lymphocytes in critical carotid artery stenosis. Clin Exp Med 2019; 19:463-468. [PMID: 31302842 DOI: 10.1007/s10238-019-00570-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 07/08/2019] [Indexed: 12/13/2022]
Abstract
The aim of this study was to evaluate CD25+ and Lag3+ T regulatory subpopulations in patients with critical carotid artery stenosis (CAS) and Stanford-A acute aortic dissection (AAD). CD25+ and Lag3+ were measured in 36 patients affected by CAS and 24 patients with Stanford type A AAD. Based on neurological symptoms, patients affected by CAS were further divided in 25 asymptomatic (CAS-A) and 11 symptomatic (CAS-S) subjects. Twenty-five patients with traditional cardiovascular risk factors (RF), matched for age and sex, were used as control group. Interleukin (IL)-10, IL-6 and transforming growth factor-β-levels were also measured. CD25+ T cells were significantly increased in CAS-S versus CAS-A (p > 0.05), AAD (p > 0.05) and RF (p > 0.05). Moreover, a significant increase in Lag3+ Tregs was observed in CAS e CAS-S versus AAD (p < 0.05) and RF (p < 0.05), whereas no significant difference was observed between CAS-S and CAS-A. IL-6 was higher in AAD compared to the other groups. Patients with neurological symptoms display a peculiar expansion of CD25+ T cells, strongly confirming a relationship between ischemic brain damage and this regulatory subpopulation, whereas Lag3+ Tregs early distinguish CAS from AAD and probably exert protective actions against aortic wall rupture throughout their anti-inflammatory functions.
Collapse
Affiliation(s)
- F Del Porto
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Medicina Interna, Sapienza Università di Roma, Rome, Italy.
| | - N Cifani
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Medicina Interna, Sapienza Università di Roma, Rome, Italy
| | - M Proietta
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Medicina Interna, Sapienza Università di Roma, Rome, Italy
| | - T Dezi
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Chirurgia Vascolare, Sapienza Università di Roma, Rome, Italy
| | - L Tritapepe
- Dipartimento di Scienze Anestesiologiche, Facoltà di Medicina e Odontoiatria, Medicina Critica e Terapia del Dolore, Policlinico Umberto I, Sapienza Università di Roma, Rome, Italy
| | - S Raffa
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Diagnostica Cellulare, Sapienza Università di Roma, Rome, Italy
| | - A Micaloni
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Diagnostica Cellulare, Sapienza Università di Roma, Rome, Italy
| | - M Taurino
- Dipartimento di Medicina Clinica e Molecolare, Facoltà di Medicina e Psicologia, Ospedale Sant'Andrea, UOC Chirurgia Vascolare, Sapienza Università di Roma, Rome, Italy
| |
Collapse
|
504
|
Kim J, Yoo JY, Suh JM, Park S, Kang D, Jo H, Bae YS. The flagellin-TLR5-Nox4 axis promotes the migration of smooth muscle cells in atherosclerosis. Exp Mol Med 2019; 51:1-13. [PMID: 31292433 PMCID: PMC6802658 DOI: 10.1038/s12276-019-0275-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 03/29/2019] [Accepted: 04/10/2019] [Indexed: 01/22/2023] Open
Abstract
We hypothesized that NADPH oxidase 4 (Nox4) is involved in the formation of neointimal atherosclerotic plaques through the migration of smooth muscle cells (SMCs) in response to flagellin. Here, we demonstrate that TLR5-mediated Nox4 activation regulates the migration of SMCs, leading to neointimal plaque formation in atherosclerosis. To investigate the molecular mechanism by which the TLR5-Nox4 cascade mediates SMC migration, we analyzed the signaling cascade in primary vascular SMCs (VSMCs) from wild-type (WT) or Nox4 KO mice. Stimulation of VSMCs from Nox4 KO mice with flagellin failed to induce H2O2 production and Rac activation compared with stimulation of VSMCs from WT mice. Moreover, the migration of Nox4-deficient VSMCs was attenuated in response to flagellin in transwell migration and wound healing assays. Finally, we performed partial carotid artery ligation in ApoE KO and Nox4ApoE DKO mice fed a high-fat diet (HFD) with or without recombinant FliC (rFliC) injection. Injection of rFliC into ApoE KO mice fed a HFD resulted in significantly increased SMC migration into the intimal layer, whereas SMC accumulation was not detected in Nox4ApoE DKO mice. We conclude that activation of the TLR5-Nox4 cascade plays an important role in the formation of neointimal atherosclerotic plaques.
Collapse
Affiliation(s)
- Jinoh Kim
- 0000 0001 2171 7754grid.255649.9Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Jung-Yeon Yoo
- 0000 0001 2171 7754grid.255649.9Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Jung Min Suh
- 0000 0001 2171 7754grid.255649.9Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Sujin Park
- 0000 0001 2171 7754grid.255649.9Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Dongmin Kang
- 0000 0001 2171 7754grid.255649.9Department of Life Science, Ewha Womans University, Seoul, Korea
| | - Hanjoong Jo
- 0000 0001 0941 6502grid.189967.8Department of Biotechnology, Emory University, Atlanta, GA USA
| | - Yun Soo Bae
- 0000 0001 2171 7754grid.255649.9Department of Life Science, Ewha Womans University, Seoul, Korea
| |
Collapse
|
505
|
Kusters PJH, Lutgens E, Seijkens TTP. Exploring immune checkpoints as potential therapeutic targets in atherosclerosis. Cardiovasc Res 2019; 114:368-377. [PMID: 29309533 DOI: 10.1093/cvr/cvx248] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 12/21/2017] [Indexed: 12/20/2022] Open
Abstract
In the past decades, the inflammatory nature of atherosclerosis has been well-recognized and despite the development of therapeutic strategies targeted at its classical risk factors such as dyslipidemia and hypertension, atherosclerosis remains a major cause of morbidity and mortality. Additional strategies targeting the chronic inflammatory pathways underlying the development of atherosclerosis are therefore required. Interactions between different immune cells result in the secretion of inflammatory mediators, such as cytokines and chemokines, and fuel atherogenesis. Immune checkpoint proteins have a critical role in facilitating immune cell interactions and play an essential role in the development of atherosclerosis. Although the therapeutic potential of these molecules is well-recognized in clinical oncology, the use of immune checkpoint modulators in atherosclerosis is still limited to experimental models. Here, we review recent insights on the role of immune checkpoint proteins in atherosclerosis. Additionally, we explore the therapeutic potential and challenges of immune checkpoint modulating strategies in cardiovascular medicine and we discuss novel therapeutic approaches to target these proteins in atherosclerosis.
Collapse
Affiliation(s)
- Pascal J H Kusters
- Department of Medical Biochemistry, Experimental Vascular Biology, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 15, 1105 CZ Amsterdam, The Netherlands
| | - Esther Lutgens
- Department of Medical Biochemistry, Experimental Vascular Biology, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 15, 1105 CZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University (LMU), Pettenkoferstraße 8a, 80336 Munich, Germany
| | - Tom T P Seijkens
- Department of Medical Biochemistry, Experimental Vascular Biology, Academic Medical Center (AMC), University of Amsterdam, Meibergdreef 15, 1105 CZ Amsterdam, The Netherlands.,Institute for Cardiovascular Prevention (IPEK), Ludwig Maximilian's University (LMU), Pettenkoferstraße 8a, 80336 Munich, Germany
| |
Collapse
|
506
|
Libby P, Hansson GK. Taming Immune and Inflammatory Responses to Treat Atherosclerosis. J Am Coll Cardiol 2019; 71:173-176. [PMID: 29325641 DOI: 10.1016/j.jacc.2017.10.081] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2017] [Accepted: 10/02/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| | - Göran K Hansson
- Department of Medicine and Center for Molecular Medicine, Karolinska University Hospital, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
507
|
Wu Z, Zhang Z, Lei Z, Lei P. CD14: Biology and role in the pathogenesis of disease. Cytokine Growth Factor Rev 2019; 48:24-31. [PMID: 31296363 DOI: 10.1016/j.cytogfr.2019.06.003] [Citation(s) in RCA: 143] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 06/21/2019] [Indexed: 12/22/2022]
Abstract
Human monocyte differentiation antigen CD14 is a pattern recognition receptor (PRR) that enhances innate immune responses. CD14 was first identified as a marker of monocytes to signal intracellular responses upon bacterial encounters. Given the absence of an intracellular tail, CD14 was doubted to have the signaling capacities. Later CD14 was confirmed as the TLR co-receptor for the detection of pathogen-associated molecular patterns. However, CD14 has been revealed as a multi-talented receptor. In last decade, CD14 was identified to activate NFAT to regulate the life cycle of myeloid cells in a TLR4-independent manner and to transport inflammatory lipids to induce phagocyte hyperactivation. And its influences on multiple related diseases have been further considered. In this review, we summarize advancements in the basic biology of the CD14 including its structure, binding ligands, signaling pathways, and its roles in the pathogenesis of inflammation, atherosclerosis, tumor and metabolic diseases. We also discuss the therapeutic potential of targeting the CD14 in related diseases.
Collapse
Affiliation(s)
- Zhenghao Wu
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhenxiong Zhang
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Biliary-Pancreatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030 China
| | - Zehua Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China; Department of Orthopaedic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ping Lei
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
508
|
Zirak MR, Mehri S, Karimani A, Zeinali M, Hayes AW, Karimi G. Mechanisms behind the atherothrombotic effects of acrolein, a review. Food Chem Toxicol 2019; 129:38-53. [DOI: 10.1016/j.fct.2019.04.034] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/18/2019] [Accepted: 04/18/2019] [Indexed: 12/31/2022]
|
509
|
Novikova OA, Nazarkina ZK, Cherepanova AV, Laktionov PP, Chelobanov BP, Murashov IS, Deev RV, Pokushalov EA, Karpenko AA, Laktionov PP. Isolation, culturing and gene expression profiling of inner mass cells from stable and vulnerable carotid atherosclerotic plaques. PLoS One 2019; 14:e0218892. [PMID: 31242269 PMCID: PMC6594632 DOI: 10.1371/journal.pone.0218892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
The connective tissue components that form the atherosclerotic plaque body are produced by the plaque inner mass cells (PIMC), located inside the plaque. We report an approach to isolate and culture cells from the connective tissue of stable and vulnerable human atherosclerotic plaques based on elimination of non-connective tissue cells such as blood and non-plaque intima cells with a lysis buffer. The resulting plaque cells were characterized by growth capacity, morphology, transcriptome profiling and specific protein expression. Plaque cells slowly proliferated for up to three passages unaffected by the use of proliferation stimulants or changes of culture media composition. Stable plaques yielded more cells than vulnerable ones. Plaque cell cultures also contained several morphological cellular types. RNA-seq profiles of plaque cells were different from any of the cell types known to be involved in atherogenesis. The expression of the following proteins was observed in cultured plaque cells: smooth muscle cells marker α-SMA, macrophage marker CD14, extracellular matrix proteins aggrecan, fibronectin, neovascularisation markers VEGF-A, CD105, cellular adhesion receptor CD31 and progenitor/dedifferentiation receptor CD34. Differential expression of several notable transcripts in cells from stable and vulnerable plaques suggests the value of plaque cell culture studies for the search of plaque vulnerability markers.
Collapse
Affiliation(s)
- Olga A. Novikova
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Zhanna K. Nazarkina
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Anna V. Cherepanova
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
- * E-mail:
| | - Petr P. Laktionov
- Laboratory of Genomics, SB RAS Institute of Molecular and Cellular Biology, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Boris P. Chelobanov
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Ivan S. Murashov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | | | - Evgeny A. Pokushalov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Andrey A. Karpenko
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Pavel P. Laktionov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| |
Collapse
|
510
|
Schnitzler JG, Dallinga-Thie GM, Kroon J. The Role of (Modified) Lipoproteins in Vascular Function: A Duet Between Monocytes and the Endothelium. Curr Med Chem 2019; 26:1594-1609. [PMID: 29546830 DOI: 10.2174/0929867325666180316121015] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 12/05/2017] [Accepted: 12/06/2017] [Indexed: 12/24/2022]
Abstract
Over the last century, many studies have demonstrated that low-density lipoprotein (LDL) is a key risk factor of cardiovascular diseases (CVD) related to atherosclerosis. Thus, for these CVD patients, LDL lowering agents are commonly used in the clinic to reduce the risk for CVD. LDL, upon modification, will develop distinct inflammatory and proatherogenic potential, leading to impaired endothelial integrity, influx of immune cells and subsequent increased foam cell formation. LDL can also directly affect peripheral monocyte composition, rendering them in a more favorable position to migrate and accumulate in the subendothelial space. It has become apparent that other lipoprotein particles, such as triglyceride- rich lipoproteins or remnants (TRL) and lipoprotein(a) [Lp(a)] may also impact on atherogenic pathways. Evidence is accumulating that Lp(a) can promote peripheral monocyte activation, eventually leading to increased transmigration through the endothelium. Similarly, remnant cholesterol has been identified to play a key role in endothelial dysfunction and monocyte behavior. In this review, we will discuss recent developments in understanding the role of different lipoproteins in the context of inflammation at both the level of the monocyte and the endothelium.
Collapse
Affiliation(s)
- Johan G Schnitzler
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Geesje M Dallinga-Thie
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands.,Department of Experimental Vascular Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
511
|
Danvin A, Quillard T, Espitia O, Charrier C, Guyomarch B, Gouëffic Y, Maurel B. Impact of Femoral Ossification on Local and Systemic Cardiovascular Patients' Condition. Ann Vasc Surg 2019; 60:335-345. [PMID: 31200045 DOI: 10.1016/j.avsg.2019.03.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 02/22/2019] [Accepted: 03/01/2019] [Indexed: 12/22/2022]
Abstract
BACKGROUND Vascular calcifications are associated with a high cardiovascular morbi-mortality in the coronary territory. In parallel, femoral arteries are more calcified and develop osteoid metaplasia (OM). This study was conducted to assess the predictive value of OM and local inflammation on the occurrence of mid- and long-term adverse cardiovascular events. METHOD Between 2008 and 2015, 86 atheromatous samples were harvested during femoral endarterectomy on 81 patients and processed for histomorphological analyses of calcifications and inflammation (monocytes and B cells). Histological findings were compared with the long-term follow-up of patients, including major adverse cardiac event (MACE), major adverse limb event (MALE), and mortality. Frequencies were presented as percentage, and continuous data, as mean and standard deviation. A P-value < 0.05 was considered statistically significant. RESULTS Median follow-up was 42.4 months (26.9-58.8). Twenty-eight percent of patients underwent a MACE; a MALE occurred in 18 (21%) limbs. Survival rate was 87.2% at 36 months. OM was found in 41 samples (51%), without any significant impact on the occurrence of MACE, MALE, or mortality. Preoperative white blood cell formulae revealed a higher rate of neutrophils associated with MACE (P = 0.04) and MALE (P = 0.0008), correlated with higher B cells counts in plaque samples. CONCLUSIONS OM is part of femoral calcifications in almost 50% of the cases but does not seem to be an independent predictive variable for MACE or MALE. However, a higher rate of B cell infiltration of the plaque and preoperative neutrophil blood count may be predictive of adverse events during follow-up.
Collapse
Affiliation(s)
- Aurore Danvin
- CHU Nantes, l'institut du thorax, service de chirurgie vasculaire, Nantes, France
| | - Thibaut Quillard
- Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France
| | - Olivier Espitia
- Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France; CHU Nantes, unité de médicine vasculaire, Nantes, France; Université de Nantes, Nantes, France
| | - Céline Charrier
- Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France
| | - Béatrice Guyomarch
- CHU Nantes, institut du thorax, INSERM, CNRS, UNIV Nantes, Nantes, France
| | - Yann Gouëffic
- CHU Nantes, l'institut du thorax, service de chirurgie vasculaire, Nantes, France; Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France; Université de Nantes, Nantes, France
| | - Blandine Maurel
- CHU Nantes, l'institut du thorax, service de chirurgie vasculaire, Nantes, France; Laboratoire de Physiopathologie de la Résorption Osseuse, Inserm UMR S 1238, Nantes, France.
| |
Collapse
|
512
|
Ji YP, Shi TY, Zhang YY, Lin D, Linghu KG, Xu YN, Tao L, Lu Q, Shen XC. Essential oil from Fructus Alpinia zerumbet (fruit of Alpinia zerumbet (Pers.) Burtt.et Smith) protected against aortic endothelial cell injury and inflammation in vitro and in vivo. JOURNAL OF ETHNOPHARMACOLOGY 2019; 237:149-158. [PMID: 30880260 DOI: 10.1016/j.jep.2019.03.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 03/05/2019] [Accepted: 03/05/2019] [Indexed: 06/09/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Fructus Alpinia zerumbet (FAZ), a dry and ripe fruit of Alpinia zerumbet (Pers.) Burtt. et Smith, is widely used as a spice to treat cardiovascular diseases in clinic as a miao folk medicine in Guizhou Province of China. Essential oil extracted from FAZ (EOFAZ) is the key bioactive ingredients. AIM OF THE STUDY This study aimed to examine the effects and mechanisms of EOFAZ on lipopolysaccharide (LPS)-induced endothelial cell injury, inflammation and apoptosis in vitro and in vivo. MATERIALS AND METHODS For the in vitro study, LPS-treated human aortic endothelial cells were used to perform PCR, western blot analysis and immunofluorescence. For the in vivo study, male mouse were divided into four groups, vehicle control group and LPS group received 0.5% Tween-80 in saline; and two EOFAZ groups receive different dose of EOFAZ (90 mg kg -1·day-1, 180 mg kg -1·day-1) respectively. Each group was fed for 7 days by intragastrical administration at daily base. Then, except vehicle control group received saline, mice in other three groups were administered with LPS (1 mg kg -1, dissolved in saline) by intraperitoneal injection. 24 h later, Aorta tissue was collected and frozen immediately in liquid N2, stored at -80 °C for western blot analysis. RESULTS We found that EOFAZ completely prevented LPS-induced HAEC activation and inflammation in vitro and in vivo, as assessed by expression of endothelial adhesion molecules, ICAM-1 and VCAM-1. Similarly, EOFAZ significantly blunted LPS-induced endothelial injury, as tested by MTT assay, LDH release and caspase-3 activation. We further demonstrated that TLR4-dependent NF-κB signaling may be involved in the process. CONCLUSION EOFAZ protected against LPS-induced endothelial cell injury and inflammation likely via inhibition of TLR4-dependent NF-κB signaling.
Collapse
Affiliation(s)
- Yun-Peng Ji
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China; Department of Pharmacy, Guizhou Provincial People's Hospital, Guiyang City, Guizhou Province, China
| | - Ting-Yu Shi
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China; Department of Neonatal Disease Screening, Shenyang Maternity and Child Health Hospital, Shenyang City, Liaoning Province, China
| | - Yan-Yan Zhang
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Dan Lin
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Ke-Gang Linghu
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Yi-Ni Xu
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Ling Tao
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China
| | - Qing Lu
- Vascular Research Laboratory, Providence Veterans Affairs Medical Center/ Alpert Medical School of Brown University, Providence, RI, USA.
| | - Xiang-Chun Shen
- The Department of Pharmacology of Materia Medica (the State Key Laboratory of Functions and Applications of Medicinal Plants, The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, The High Educational Key Laboratory of Guizhou Province for Natural Medicianl Pharmacology and Drug Ability, The Union Key Laboratory of Guiyang City-Guizhou Medical Univeristy, The Key Laboratory of Optimal Utilizaiton of Natural Medicine Resources), School of Pharmaceutical Sciences, Guizhou Medical University, University Town, Guian New District, Guizhou, 550025, China.
| |
Collapse
|
513
|
Baumer Y, McCurdy S, Jin X, Weatherby TM, Dey AK, Mehta NN, Yap JK, Kruth HS, Boisvert WA. Ultramorphological analysis of plaque advancement and cholesterol crystal formation in Ldlr knockout mouse atherosclerosis. Atherosclerosis 2019; 287:100-111. [PMID: 31247346 DOI: 10.1016/j.atherosclerosis.2019.05.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 04/25/2019] [Accepted: 05/29/2019] [Indexed: 01/17/2023]
Abstract
BACKGOUND AND AIMS The low-density lipoprotein receptor-deficient (Ldlr-/-) mouse has been utilized by cardiovascular researchers for more than two decades to study atherosclerosis. However, there has not yet been a systematic effort to document the ultrastructural changes that accompany the progression of atherosclerotic plaque in this model. METHODS Employing several different staining and microscopic techniques, including immunohistochemistry, as well as electron and polarized microscopy, we analyzed atherosclerotic lesion development in Ldlr-/- mice fed an atherogenic diet over time. RESULTS Lipid-like deposits occurred in the subendothelial space after only one week of atherogenic diet. At two weeks, cholesterol crystals (CC) formed and increased thereafter. Lipid, CC, vascular smooth muscles cells, and collagen progressively increased over time, while after 4 weeks, relative macrophage content decreased. Accelerated accumulation of plate- and needle-shaped CC accompanied plaque core necrosis. Lastly, CC were surrounded by cholesterol microdomains, which co-localized with CC through all stages of atherosclerosis, indicating that the cholesterol microdomains may be a source of CC. CONCLUSIONS Here, we have documented, for the first time in a comprehensive way, atherosclerotic plaque morphology and composition from early to advanced stages in the Ldlr-/- mouse, one of the most commonly used animal models utilized in atherosclerosis research.
Collapse
Affiliation(s)
- Yvonne Baumer
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI, 96813, USA
| | - Sara McCurdy
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI, 96813, USA
| | - Xueting Jin
- Section of Experimental Atherosclerosis, National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Tina M Weatherby
- Pacific Biosciences Research Center, Biological Electron Microscope Facility, University of Hawaii, 2538 The Mall, Snyder Hall, Honolulu, HI, 96822, USA
| | - Amit K Dey
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Nehal N Mehta
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - Jonathan K Yap
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI, 96813, USA
| | - Howard S Kruth
- Section of Experimental Atherosclerosis, National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD, 20892, USA
| | - William A Boisvert
- Center for Cardiovascular Research, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo Street, Honolulu, HI, 96813, USA; Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan, Russia.
| |
Collapse
|
514
|
Analysis of Genetic Variation in CD40 and CD40L: Relationship with mRNA Relative Expression and Soluble Proteins in Acute Coronary Syndrome. J Immunol Res 2019; 2019:8063983. [PMID: 31183392 PMCID: PMC6515173 DOI: 10.1155/2019/8063983] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 03/13/2019] [Accepted: 04/10/2019] [Indexed: 11/27/2022] Open
Abstract
Acute coronary syndrome (ACS) can be triggered by the presence of inflammatory factors which promote the activation of immune cells by costimulatory molecules such as CD40 and its ligand CD40L. Environmental and genetic factors are involved in the etiology of the ACS. The aim of this study was to explore the gene and protein expression associated with CD40 and CD40L genetic variants in ACS patients from the western Mexican population. A total of 620 individuals from western Mexico were recruited: 320 ACS patients and 300 individuals without a history of ischemic cardiopathy were evaluated. The genotype was determined using TaqMan SNP genotyping assays. CD40 and CD40L expressions at the mRNA level were quantified using TaqMan Gene Expression Assays. Soluble protein isoforms were measured by enzyme-linked immunosorbent assay. We did not find evidence of association between CD40 (rs1883832, rs4810485, and rs11086998) and CD40L (rs3092952 and rs3092920) genetic variants and susceptibility to ACS, although rs1883832 and rs4810485 were significantly associated with high sCD40 plasma levels. Plasma levels of sCD40L can be affected by gender and the clinical spectrum of acute coronary syndrome. Our results do not suggest a functional role of CD40 and CD40L genetic variants in ACS. However, they could reflect the inflammatory process and platelet activation in ACS patients, even when they are under pharmacological therapy. Due to the important roles of the CD40-CD40L system in the pathogenesis of ACS, longitudinal studies are required to determine if soluble levels of CD40 and CD40L could be clinically useful markers of a recurrent cardiovascular event after an ACS.
Collapse
|
515
|
Shahi A, Aslani S, Ataollahi M, Mahmoudi M. The role of magnesium in different inflammatory diseases. Inflammopharmacology 2019; 27:649-661. [PMID: 31172335 DOI: 10.1007/s10787-019-00603-7] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 05/11/2019] [Indexed: 12/26/2022]
Abstract
Magnesium deficiency (MgD) can cause inflammation in human body. The known mechanisms of inflammation caused by MgD include activation of phagocytic cells, opening of calcium channels, activation of the N-methyl-D-aspartate (NMDA) receptor, and activation of nuclear factor (NF)-κB. In addition, MgD causes systemic stress response through neuroendocrinological pathways. The inflammation caused by MgD can result in pro-atherogenic changes in the metabolism of lipoproteins, endothelial dysfunction, and high blood pressure. Studies suggest that magnesium may play an important role in the pathophysiology of some inflammatory diseases. Several clinical trials and laboratory studies have been done on the functional role of magnesium. In this study, we review some inflammatory diseases, in which the magnesium has a role in their pathophysiology. Among these diseases, diabetes, asthma, preeclampsia, atherosclerosis, heart damage, and rheumatoid arthritis have been highlighted.
Collapse
Affiliation(s)
- Abbas Shahi
- Rheumatology Research Center, Tehran University of Medical Sciences, P.O.Box: 14117-13137, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences, P.O.Box: 14117-13137, Tehran, Iran
| | - MohammadReza Ataollahi
- Department of Medical Immunology, School of Medicine, Fasa University of Medical Sciences, P.O. Box: 74616-86688, Fasa, Iran.
| | - Mahdi Mahmoudi
- Rheumatology Research Center, Tehran University of Medical Sciences, P.O.Box: 14117-13137, Tehran, Iran.
| |
Collapse
|
516
|
LI TT, WANG ZB, LI Y, CAO F, YANG BY, KUANG HX. The mechanisms of traditional Chinese medicine underlying the prevention and treatment of atherosclerosis. Chin J Nat Med 2019; 17:401-412. [DOI: 10.1016/s1875-5364(19)30048-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Indexed: 02/07/2023]
|
517
|
Xue Y, Wu Y, Wang Q, Xue L, Su Z, Zhang C. Cellular Vehicles Based on Neutrophils Enable Targeting of Atherosclerosis. Mol Pharm 2019; 16:3109-3120. [PMID: 31082253 DOI: 10.1021/acs.molpharmaceut.9b00342] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Given the multiple interactions between neutrophils (NEs) and atherosclerosis (AS), in this study, we exploited NEs as cellular vehicles loaded with cationic liposomes for actively targeting atherosclerotic sites. The cellular vehicles based on NEs possess efficient internalization of cationic liposomes and sensitive response to the chemotaxis of atherosclerotic inflammatory cells, which ultimately realize the targeted delivery of the cargos into the target cells in vitro. Moreover, these effects also translated to significant enhancement of the accumulation of NEs' cargos into the atherosclerotic plaque in vivo after administering NE vehicles to the AS animal model. Consequently, cellular vehicles based on NEs could be a novel strategy for targeted delivery of payloads into atherosclerotic plaque, which would facilitate theranostics for AS and the development of anti-AS drugs to manage the disease.
Collapse
Affiliation(s)
- Yanan Xue
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Yue Wu
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Qianqian Wang
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Lingjing Xue
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Zhigui Su
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| | - Can Zhang
- State Key Laboratory of Natural Medicines Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases Center of Advanced Pharmaceuticals and Biomaterials , China Pharmaceutical University , No. 24 Tongjiaxiang , Nanjing 210009 , China
| |
Collapse
|
518
|
Vieceli Dalla Sega F, Fortini F, Aquila G, Campo G, Vaccarezza M, Rizzo P. Notch Signaling Regulates Immune Responses in Atherosclerosis. Front Immunol 2019; 10:1130. [PMID: 31191522 PMCID: PMC6540611 DOI: 10.3389/fimmu.2019.01130] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Accepted: 05/03/2019] [Indexed: 01/05/2023] Open
Abstract
Atherosclerosis is a chronic autoimmune inflammatory disease that can cause coronary artery disease, stroke, peripheral artery disease, depending on which arteries are affected. At the beginning of atherosclerosis plasma lipoproteins accumulate in the sub-endothelial space. In response, monocytes migrate from the circulation through the endothelium into the intima where they differentiate into macrophages. These early events trigger a complex immune response that eventually involves many cellular subtypes of both innate and adaptive immunity. The Notch signaling pathway is an evolutionary conserved cell signaling system that mediates cell-to-cell communication. Recent studies have revealed that Notch modulate atherosclerosis by controlling macrophages polarization into M1 or M2 subtypes. Furthermore, it is known that Notch signaling controls differentiation and activity of T-helper and cytotoxic T-cells in inflammatory diseases. In this review, we will discuss the role of Notch in modulating immunity in the context of atherosclerosis and whether targeting Notch may represent a therapeutic strategy.
Collapse
Affiliation(s)
| | - Francesca Fortini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy
| | - Giorgio Aquila
- Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Gianluca Campo
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy.,Cardiovascular Center, Azienda Ospedaliero-Universitaria di Ferrara, Cona, Italy
| | - Mauro Vaccarezza
- Faculty of Health Sciences, School of Pharmacy and Biomedical Sciences, Curtin University, Perth, WA, Australia
| | - Paola Rizzo
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, Cotignola, Italy.,Department of Morphology, Surgery, and Experimental Medicine, University of Ferrara, Ferrara, Italy.,Laboratory for Technologies of Advanced Therapies, University of Ferrara, Ferrara, Italy
| |
Collapse
|
519
|
Neupane R, Jin X, Sasaki T, Li X, Murohara T, Cheng XW. Immune Disorder in Atherosclerotic Cardiovascular Disease - Clinical Implications of Using Circulating T-Cell Subsets as Biomarkers. Circ J 2019; 83:1431-1438. [PMID: 31092769 DOI: 10.1253/circj.cj-19-0114] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Atherosclerotic cardiovascular disease (ACVD) is an inflammatory phenomenon that leads to structural abnormality in the vascular lumen due to the formation of atheroma by the deposition of lipid particles and inflammatory cytokines. There is a close interaction between innate immune cells (neutrophils, monocyte, macrophages, dendritic cells) and adaptive immune cells (T and B lymphocytes) in the initiation and progression of atherosclerosis. According to novel insights into the role of adaptive immunity in atherosclerosis, the activation of CD4+T cells in response to oxidized low-density lipoprotein-antigen initiates the formation and facilitates the propagation of atheroma, whereas CD8+T cells cause the rupture of a developed atheroma by their cytotoxic nature. Peripheral CD4+and CD8+T-cell counts were altered in patients with other cardiovascular risk factors. Furthermore, on evaluation of the feasibility of immune cells as a diagnostic tool, the blood CD4+(helper), CD8+(cytotoxic), and CD4+CD25+Foxp3+(regulatory) T cells and the ratio of CD4 to CD8 cells hold promise as biomarkers of coronary artery disease and their subtypes. T cells also could be a therapeutic target for cardiovascular diseases. The goal of this review was therefore to summarize the available information regarding immune disorders in ACVD with a special focus on the clinical implications of circulating T-cell subsets as biomarkers.
Collapse
Affiliation(s)
- Rajib Neupane
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Xiongjie Jin
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Takeshi Sasaki
- Department of Anatomy and Neuroscience, Hamamatsu University School of Medicine
| | - Xiang Li
- Department of Cardiology and Hypertension, Yanbian University Hospital
| | - Toyoaki Murohara
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Xian Wu Cheng
- Department of Cardiology and Hypertension, Yanbian University Hospital.,Department of Cardiology, Nagoya University Graduate School of Medicine
| |
Collapse
|
520
|
Puhm F, Afonyushkin T, Resch U, Obermayer G, Rohde M, Penz T, Schuster M, Wagner G, Rendeiro AF, Melki I, Kaun C, Wojta J, Bock C, Jilma B, Mackman N, Boilard E, Binder CJ. Mitochondria Are a Subset of Extracellular Vesicles Released by Activated Monocytes and Induce Type I IFN and TNF Responses in Endothelial Cells. Circ Res 2019; 125:43-52. [PMID: 31219742 DOI: 10.1161/circresaha.118.314601] [Citation(s) in RCA: 179] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
RATIONALE Extracellular vesicles, including microvesicles, are increasingly recognized as important mediators in cardiovascular disease. The cargo and surface proteins they carry are considered to define their biological activity, including their inflammatory properties. Monocyte to endothelial cell signaling is a prerequisite for the propagation of inflammatory responses. However, the contribution of microvesicles in this process is poorly understood. OBJECTIVE To elucidate the mechanisms by which microvesicles derived from activated monocytic cells exert inflammatory effects on endothelial cells. METHODS AND RESULTS LPS (lipopolysaccharide)-stimulated monocytic cells release free mitochondria and microvesicles with mitochondrial content as demonstrated by flow cytometry, quantitative polymerase chain reaction, Western Blot, and transmission electron microscopy. Using RNAseq analysis and quantitative reverse transcription-polymerase chain reaction, we demonstrated that both mitochondria directly isolated from and microvesicles released by LPS-activated monocytic cells, as well as circulating microvesicles isolated from volunteers receiving low-dose LPS-injections, induce type I IFN (interferon), and TNF (tumor necrosis factor) responses in endothelial cells. Depletion of free mitochondria significantly reduced the ability of these microvesicles to induce type I IFN and TNF-dependent genes. We identified mitochondria-associated TNFα and RNA from stressed mitochondria as major inducers of these responses. Finally, we demonstrated that the proinflammatory potential of microvesicles and directly isolated mitochondria were drastically reduced when they were derived from monocytic cells with nonrespiring mitochondria or monocytic cells cultured in the presence of pyruvate or the mitochondrial reactive oxygen species scavenger MitoTEMPO. CONCLUSIONS Mitochondria and mitochondria embedded in microvesicles constitute a major subset of extracellular vesicles released by activated monocytes, and their proinflammatory activity on endothelial cells is determined by the activation status of their parental cells. Thus, mitochondria may represent critical intercellular mediators in cardiovascular disease and other inflammatory settings associated with type I IFN and TNF signaling.
Collapse
Affiliation(s)
- Florian Puhm
- From the Department of Laboratory Medicine (F.P., T.A., G.O., G.W., C.B., C.J.B.).,Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| | - Taras Afonyushkin
- From the Department of Laboratory Medicine (F.P., T.A., G.O., G.W., C.B., C.J.B.).,Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| | | | - Georg Obermayer
- From the Department of Laboratory Medicine (F.P., T.A., G.O., G.W., C.B., C.J.B.).,Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany (M.R.)
| | - Thomas Penz
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| | - Michael Schuster
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| | - Gabriel Wagner
- From the Department of Laboratory Medicine (F.P., T.A., G.O., G.W., C.B., C.J.B.)
| | - Andre F Rendeiro
- Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| | - Imene Melki
- Department of Infectious Diseases and Immunity, Faculty of Medicine, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Quebec City, Canada (I.M., E.B.)
| | | | - Johann Wojta
- Department of Internal Medicine II (C.K., J.W.).,Core Facilities (J.W.).,Ludwig Boltzmann Cluster for Cardiovascular Research, Vienna, Austria (J.W.)
| | - Christoph Bock
- From the Department of Laboratory Medicine (F.P., T.A., G.O., G.W., C.B., C.J.B.).,Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| | - Bernd Jilma
- Department of Clinical Pharmacology (B.J.), Medical University of Vienna, Austria
| | - Nigel Mackman
- Division of Hematology and Oncology, Department of Medicine, University of North Carolina at Chapel Hill (N.M.)
| | - Eric Boilard
- Department of Infectious Diseases and Immunity, Faculty of Medicine, Centre de Recherche du Centre Hospitalier Universitaire de Québec, Université Laval, Quebec City, Canada (I.M., E.B.)
| | - Christoph J Binder
- From the Department of Laboratory Medicine (F.P., T.A., G.O., G.W., C.B., C.J.B.).,Research Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna (F.P., T.A., G.O., T.P., M.S., A.F.R., C.B., C.J.B.)
| |
Collapse
|
521
|
Raddatz MA, Madhur MS, Merryman WD. Adaptive immune cells in calcific aortic valve disease. Am J Physiol Heart Circ Physiol 2019; 317:H141-H155. [PMID: 31050556 DOI: 10.1152/ajpheart.00100.2019] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Calcific aortic valve disease (CAVD) is highly prevalent and has no pharmaceutical treatment. Surgical replacement of the aortic valve has proved effective in advanced disease but is costly, time limited, and in many cases not optimal for elderly patients. This has driven an increasing interest in noninvasive therapies for patients with CAVD. Adaptive immune cell signaling in the aortic valve has shown potential as a target for such a therapy. Up to 15% of cells in the healthy aortic valve are hematopoietic in origin, and these cells, which include macrophages, T lymphocytes, and B lymphocytes, are increased further in calcified specimens. Additionally, cytokine signaling has been shown to play a causative role in aortic valve calcification both in vitro and in vivo. This review summarizes the physiological presence of hematopoietic cells in the valve, innate and adaptive immune cell infiltration in disease states, and the cytokine signaling pathways that play a significant role in CAVD pathophysiology and may prove to be pharmaceutical targets for this disease in the near future.
Collapse
Affiliation(s)
- Michael A Raddatz
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee.,Vanderbilt University School of Medicine , Nashville, Tennessee
| | - Meena S Madhur
- Department of Medicine, Vanderbilt University Medical Center , Nashville, Tennessee.,Department of Molecular Physiology and Biophysics, Vanderbilt University , Nashville, Tennessee.,Division of Clinical Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee
| | - W David Merryman
- Department of Biomedical Engineering, Vanderbilt University , Nashville, Tennessee
| |
Collapse
|
522
|
Park S, Lee IK. Progression of Multifaceted Immune Cells in Atherosclerotic Development. J Lipid Atheroscler 2019; 8:15-25. [PMID: 32821696 PMCID: PMC7379084 DOI: 10.12997/jla.2019.8.1.15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/29/2019] [Accepted: 04/16/2019] [Indexed: 12/16/2022] Open
Abstract
Atherosclerosis is a major cause of morbidity and mortality due to cardiovascular diseases, such as coronary artery disease, stroke, and peripheral vascular disease, that are associated with thrombosis-induced organ infarction. In Westernized countries, the high prevalence of obesity-induced insulin resistance is predicted to be a major factor leading to atherosclerotic vascular disease. Both genetic and environmental factors interfere with immune responses in atherosclerosis development with chronic and non-resolving states. The most known autoimmune disease therapy is cytokine-targeted therapy, which targets tumor necrosis factor-α and interleukin (IL)-17 antagonists. Recently, a clinical trial with the anti-IL-1β antibody (canakinumab) had shown that the anti-inflammatory effects in canakinumab-treated subjects play a critical role in reducing cardiovascular disease prevalence. Recent emerging data have suggested effective therapeutics involving anti-obesity and anti-diabetic agents, as well as statin and anti-platelet drugs, for atherothrombosis prevention. It is well-known that specialized immune differentiation and activation completely depends on metabolic reprogramming mediated by mitochondrial dynamics in distinct immune cells. Therefore, there is a strong mechanistic link between metabolism and immune function mediated by mitochondrial function. In this review, we describe that cellular metabolism in immune cells is strongly interconnected with systemic metabolism in terms of diverse phenotypes and activation.
Collapse
Affiliation(s)
- Sungmi Park
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Korea
| | - In-Kyu Lee
- Leading-edge Research Center for Drug Discovery and Development for Diabetes and Metabolic Disease, Kyungpook National University Hospital, Daegu, Korea
| |
Collapse
|
523
|
Mathematical Modelling and Simulation of Atherosclerosis Formation and Progress: A Review. Ann Biomed Eng 2019; 47:1764-1785. [PMID: 31020444 DOI: 10.1007/s10439-019-02268-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/10/2019] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) is a major threat to human health since it is the leading cause of death in western countries. Atherosclerosis is a type of CVD related to hypertension, diabetes, high levels of cholesterol, smoking, oxidative stress, and age. Atherosclerosis primarily occurs in medium and large arteries, such as coronary and the carotid artery and, in particular, at bifurcations and curvatures. Atherosclerosis is compared to an inflammatory disease where a thick, porous material comprising cholesterol fat, saturated sterols, proteins, fatty acids, calcium etc., is covered by an endothelial membrane and a fragile fibrous tissue which makes atheromatic plaque prone to rupture that could lead to the blockage of the artery due to the released plaque material. Despite the great progress achieved, the nature of the disease is not fully understood. This paper reviews the current state of modelling of all levels of atherosclerosis formation and progress and discusses further challenges in atherosclerosis modelling. The objective is to pave a way towards more precise computational tools to predict and eventually reengineer the fate of atherosclerosis.
Collapse
|
524
|
Libby P, Kobold S. Inflammation: a common contributor to cancer, aging, and cardiovascular diseases-expanding the concept of cardio-oncology. Cardiovasc Res 2019; 115:824-829. [PMID: 30830168 PMCID: PMC6452304 DOI: 10.1093/cvr/cvz058] [Citation(s) in RCA: 95] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2019] [Accepted: 02/28/2019] [Indexed: 12/19/2022] Open
Abstract
Inflammation participates in the pathogenesis of both cancer and cardiovascular disease. This review examines the mechanistic commonalities between these two scourges of humanity through the lens of inflammation biology. Inflammatory pathways contribute to the initiation, the progression, and the complication of both malignant tumours and atherosclerotic plaques. Modulation of inflammatory pathways have proven transformative in the treatment of cancers and have crossed the threshold of clinical reality as treatments to reduce the risk of cardiovascular events. The finding that clonal haematopoiesis drives both leukaemia and cardiovascular events provides yet another link between these two seemingly disparate diseases. The nascent specialty of cardio-oncology has initially focused on the cardiovascular complications of cancer therapies. The recognition of a more profound pathophysiologic connection between cancer and cardiovascular diseases should expand the concept of cardio-oncology. Embracing the mechanistic connection and transcending traditional barriers between disciplines offers immense opportunities for speeding innovative research that can address the growing burden of both cancer and cardiovascular disease.
Collapse
Affiliation(s)
- Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, USA
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, Center of Integrated Protein Science Munich (CIPS-M), Klinikum der Universität München, Munich, Germany
| |
Collapse
|
525
|
Hillmeister P, Buschmann I, Bondke Persson A. Listen to your physiologist! Acta Physiol (Oxf) 2019; 225:e13265. [PMID: 30762943 DOI: 10.1111/apha.13265] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/09/2019] [Accepted: 02/10/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Philipp Hillmeister
- Department for Angiology Brandenburg Medical School, Campus Clinic Brandenburg, DAZB Deutsches Angiologie Zentrum Brandenburg-Berlin Brandenburg an der Havel Germany
| | - Ivo Buschmann
- Department for Angiology Brandenburg Medical School, Campus Clinic Brandenburg, DAZB Deutsches Angiologie Zentrum Brandenburg-Berlin Brandenburg an der Havel Germany
| | - Anja Bondke Persson
- Charité– Universitätsmedizin Berlin Corporate Member of Freie Universität Berlin, Humboldt‐Universität zu Berlin, and Berlin Institute of Health Berlin Germany
| |
Collapse
|
526
|
Vaccination against atherosclerosis. Curr Opin Immunol 2019; 59:15-24. [PMID: 30928800 DOI: 10.1016/j.coi.2019.02.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/11/2019] [Accepted: 02/22/2019] [Indexed: 12/30/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease that causes most heart attacks and strokes, making it the biggest killer in the world. Although cholesterol-lowering drugs have dramatically reduced these major adverse cardiovascular events, there remains a high residual risk called inflammatory risk. Atherosclerosis has an autoimmune component that can be manipulated by immunologic approaches including vaccination. Vaccination is attractive, because it is antigen-specific, does not impair host defense, and provides long-term protection. Several candidate antigens for atherosclerosis vaccine development have been identified and have been shown to reduce atherosclerosis in animal models. In this review, we focus on two different types of atherosclerosis vaccines: antibody-inducing and regulatory T cell-inducing.
Collapse
|
527
|
Park J, Gao H, Wang Y, Hu H, Simon DI, Steinmetz NF. S100A9-targeted tobacco mosaic virus nanoparticles exhibit high specificity toward atherosclerotic lesions in ApoE -/- mice. J Mater Chem B 2019; 7:1842-1846. [PMID: 32255046 PMCID: PMC7147689 DOI: 10.1039/c8tb02276c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We integrate a biocompatible plant virus-based nanotechnology (tobacco mosaic virus, TMV) with S100A9-targeting peptides for its application in imaging and diagnosis of atherosclerosis. S100A9-targeted TMV nanoparticles exhibit remarkable specificity to S100A9 and targeting of atherosclerosis lesions in ApoE-/- mice.
Collapse
Affiliation(s)
- Jooneon Park
- Department of Nanoengineering, University of California San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | |
Collapse
|
528
|
Wang CL, Kuo CF, Yeh YH, Hsieh MY, Kuo CT, Chang SH. Familial aggregation of myocardial infarction and coaggregation of myocardial infarction and autoimmune disease: a nationwide population-based cross-sectional study in Taiwan. BMJ Open 2019; 9:e023614. [PMID: 30898803 PMCID: PMC6475449 DOI: 10.1136/bmjopen-2018-023614] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVE This study examined how a history of myocardial infarction (MI) in a person's first-degree relatives affects that person's risk of developing MI and autoimmune diseases. DESIGN Nationwide population-based cross-sectional study SETTING: All healthcare facilities in Taiwan. PARTICIPANTS A total of 24 361 345 individuals were enrolled. METHODS Using data from the National Health Insurance Research Database in Taiwan, we conducted a nationwide cross-sectional study of data collected from all beneficiaries in the Taiwan National Health Insurance system in 2015, of whom 259 360 subjects had at least one first-degree relative affected by MI in 2015. We estimated the absolute risks and relative risks (RRs) of MI and autoimmune disease in those subjects, and the relative contribution of genetic and environmental factors to their MI susceptibility. RESULTS The absolute risks of MI for subjects with at least one affected first-degree relative and the general population were 0.87% and 0.56%, respectively, in 2015. Patients with affected first-degree relatives were significantly associated with a higher RR of MI (1.76, 95% CI: 1.68 to 1.85) compared with the general population. There was no association with a higher RR of autoimmune disease. The sibling, offspring and parental MI history conferred RRs (95% CI) for MI of 2.35 (1.96 to 2.83), 2.21 (2.05 to 2.39) and 1.60 (1.52 to 1.68), respectively. The contributions of heritability, shared environmental factors and non-shared environmental factors to MI susceptibility were 19.6%, 3.4% and 77.0%, respectively. CONCLUSIONS Individuals who have first-degree relatives with a history of MI have a higher risk of developing MI than the general population. Non-shared environmental factors contributed more significantly to MI susceptibility than did heritability and shared environmental factors. A family history of MI was not associated with an increased risk of autoimmune disease.
Collapse
Affiliation(s)
- Chun-Li Wang
- Cardiovascular Department, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Chang-Fu Kuo
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Division of Rheumatology, Orthopaedics, and Dermatology, School of Medicine, University of Nottingham, Nottingham, UK
- Division of Rheumatology, Allergy, and Immunology, Chang Gung Memorial Hospital, Taoyuan, Taiwan
| | - Yung-Hsin Yeh
- Cardiovascular Department, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Mei-Yun Hsieh
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chi-Tai Kuo
- Cardiovascular Department, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Shang-Hung Chang
- Cardiovascular Department, Chang Gung Memorial Hospital, Taoyuan, Taiwan
- School of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, Linkou, Taiwan
- Graduate Institute of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| |
Collapse
|
529
|
Abstract
The intricate interplay between the immune and the nervous systems has been steadily unveiled at both cellular and molecular levels. In this issue of Immunity, Vasamsetti et al. (2018) show that sympathetic nerves drive catecholamine signaling from leukocytes, thereby promoting splenic granulocyte macrophage progenitor (GMP) proliferation and differentiation.
Collapse
Affiliation(s)
- Zhongsheng Hu
- Center for Life Sciences, Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China
| | - Wenwen Zeng
- Center for Life Sciences, Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China; Beijing Key Laboratory for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
530
|
Huang LH, Zinselmeyer BH, Chang CH, Saunders BT, Elvington A, Baba O, Broekelmann TJ, Qi L, Rueve JS, Swartz MA, Kim BS, Mecham RP, Wiig H, Thomas MJ, Sorci-Thomas MG, Randolph GJ. Interleukin-17 Drives Interstitial Entrapment of Tissue Lipoproteins in Experimental Psoriasis. Cell Metab 2019; 29:475-487.e7. [PMID: 30415924 PMCID: PMC6365189 DOI: 10.1016/j.cmet.2018.10.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 07/12/2018] [Accepted: 10/17/2018] [Indexed: 12/17/2022]
Abstract
Lipoproteins trapped in arteries drive atherosclerosis. Extravascular low-density lipoprotein undergoes receptor uptake, whereas high-density lipoprotein (HDL) interacts with cells to acquire cholesterol and then recirculates to plasma. We developed photoactivatable apoA-I to understand how HDL passage through tissue is regulated. We focused on skin and arteries of healthy mice versus those with psoriasis, which carries cardiovascular risk in man. Our findings suggest that psoriasis-affected skin lesions program interleukin-17-producing T cells in draining lymph nodes to home to distal skin and later to arteries. There, these cells mediate thickening of the collagenous matrix, such that larger molecules including lipoproteins become entrapped. HDL transit was rescued by depleting CD4+ T cells, neutralizing interleukin-17, or inhibiting lysyl oxidase that crosslinks collagen. Experimental psoriasis also increased vascular stiffness and atherosclerosis via this common pathway. Thus, interleukin-17 can reduce lipoprotein trafficking and increase vascular stiffness by, at least in part, remodeling collagen.
Collapse
Affiliation(s)
- Li-Hao Huang
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | - Bernd H Zinselmeyer
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | - Chih-Hao Chang
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | - Brian T Saunders
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | - Andrew Elvington
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | - Osamu Baba
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | | | - Lina Qi
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | - Joseph S Rueve
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA
| | - Melody A Swartz
- Division of Dermatology, Department of Medicine, Washington University, St Louis, MO 63110, USA
| | - Brian S Kim
- Institute for Molecular Engineering, University of Chicago, Chicago, IL 60637, USA
| | - Robert P Mecham
- Department of Cell Biology, Washington University, St Louis, MO 63110, USA
| | - Helge Wiig
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, Bergen 5009, Norway
| | - Michael J Thomas
- Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Mary G Sorci-Thomas
- Department of Medicine, Division of Endocrinology, Pharmacology and Toxicology, and Blood Research Institute, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Gwendalyn J Randolph
- Department of Pathology & Immunology, Washington University, St Louis, MO 63110, USA.
| |
Collapse
|
531
|
Rodriguez-Duarte J, Galliussi G, Dapueto R, Rossello J, Malacrida L, Kamaid A, Schopfer FJ, Escande C, López GV, Batthyány C. A novel nitroalkene-α-tocopherol analogue inhibits inflammation and ameliorates atherosclerosis in Apo E knockout mice. Br J Pharmacol 2019; 176:757-772. [PMID: 30588602 DOI: 10.1111/bph.14561] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 11/30/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND AND PURPOSE Atherosclerosis is characterized by chronic low-grade inflammation with concomitant lipid accumulation in the arterial wall. Anti-inflammatory and anti-atherogenic properties have been described for a novel class of endogenous nitroalkenes (nitrated-unsaturated fatty acids), formed during inflammation and digestion/absorption processes. The lipid-associated antioxidant α-tocopherol is transported systemically by LDL particles including to the atheroma lesions. To capitalize on the overlapping and complementary salutary properties of endogenous nitroalkenes and α-tocopherol, we designed and synthesized a novel nitroalkene-α-tocopherol analogue (NATOH) to address chronic inflammation and atherosclerosis, particularly at the lesion sites. EXPERIMENTAL APPROACH We synthesized NATOH, determined its electrophilicity and antioxidant capacity and studied its effects over pro-inflammatory and cytoprotective pathways in macrophages in vitro. Moreover, we demonstrated its incorporation into lipoproteins and tissue both in vitro and in vivo, and determined its effect on atherosclerosis and inflammatory responses in vivo using the Apo E knockout mice model. KEY RESULTS NATOH exhibited similar antioxidant capacity to α-tocopherol and, due to the presence of the nitroalkenyl group, like endogenous nitroalkenes, it exerted electrophilic reactivity. NATOH was incorporated in vivo into the VLDL/LDL lipoproteins particles to reach the atheroma lesions. Furthermore, oral administration of NATOH down-regulated NF-κB-dependent expression of pro-inflammatory markers (including IL-1β and adhesion molecules) and ameliorated atherosclerosis in Apo E knockout mice. CONCLUSIONS AND IMPLICATIONS In toto, the data demonstrate a novel pharmacological strategy for the prevention of atherosclerosis based on a creative, natural and safe drug delivery system of a non-conventional anti-inflammatory compound (NATOH) with significant potential for clinical application.
Collapse
Affiliation(s)
- Jorge Rodriguez-Duarte
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Germán Galliussi
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Rosina Dapueto
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Jessica Rossello
- Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Leonel Malacrida
- Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Pathophysiology Department, Hospital de Clínicas, Facultad de Medicina, Universidad de la República, Montevideo, Uruguay
| | - Andrés Kamaid
- Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Francisco J Schopfer
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Carlos Escande
- Laboratory of Metabolic Diseases and Aging, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay
| | - Gloria V López
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Departmento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Carlos Batthyány
- Laboratory of Vascular Biology and Drug Development, INDICYO Program, Institut Pasteur de Montevideo, Montevideo, Uruguay.,Analytical Biochemistry and Proteomics Unit, Institut Pasteur de Montevideo, Montevideo, Uruguay
| |
Collapse
|
532
|
de Winther MP, Dallinga-Thie GM. Introduction to the thematic review series on different levels of genetic regulation of cardiovascular disease. Atherosclerosis 2019; 281:148-149. [DOI: 10.1016/j.atherosclerosis.2018.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 12/19/2018] [Indexed: 10/27/2022]
|
533
|
Kritikou E, van der Heijden T, Swart M, van Duijn J, Slütter B, Wezel A, Smeets HJ, Maffia P, Kuiper J, Bot I. Hypercholesterolemia Induces a Mast Cell-CD4 + T Cell Interaction in Atherosclerosis. THE JOURNAL OF IMMUNOLOGY 2019; 202:1531-1539. [PMID: 30683705 DOI: 10.4049/jimmunol.1800648] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 12/20/2018] [Indexed: 11/19/2022]
Abstract
Mast cells (MCs) are potent innate immune cells that aggravate atherosclerosis through the release of proinflammatory mediators inside atherosclerotic plaques. Similarly, CD4+ T cells are constituents of the adaptive immune response and accumulate within the plaques following lipid-specific activation by APCs. Recently it has been proposed that these two cell types can interact in a direct manner. However, no indication of such an interaction has been investigated in the context of atherosclerosis. In our study, we aimed to examine whether MCs can act as APCs in atherosclerosis, thereby modulating CD4+ T cell responses. We observed that MCs increased their MHC class II expression under hyperlipidemic conditions both in vivo and in vitro. Furthermore, we showed that MCs can present Ags in vivo via MHC class II molecules. Serum from high-fat diet-fed mice also enhanced the expression of the costimulatory molecule CD86 on cultured MCs, whereas OVA peptide-loaded MCs increased OT-II CD4+ T cell proliferation in vitro. The aortic CD4+ and TH1 cell content of atherosclerotic mice that lack MCs was reduced as compared with their wild-type counterparts. Importantly, we identified MCs that express HLA-DR in advanced human atheromata, indicating that these cells are capable of Ag presentation within human atherosclerotic plaques. Therefore, in this artice, we show that MCs may directly modulate adaptive immunity by acting as APCs in atherosclerosis.
Collapse
Affiliation(s)
- Eva Kritikou
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands;
| | - Thomas van der Heijden
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Maarten Swart
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Janine van Duijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Bram Slütter
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Anouk Wezel
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Harm J Smeets
- Department of Surgery, Haaglanden Medical Center Westeinde, 2501 CK The Hague, the Netherlands
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom.,Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom; and.,Department of Pharmacy, University of Naples Federico II, 80138 Naples, Italy
| | - Johan Kuiper
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| | - Ilze Bot
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, 2300 RA Leiden, the Netherlands
| |
Collapse
|
534
|
Liu Y, Wang X, Pang J, Zhang H, Luo J, Qian X, Chen Q, Ling W. Attenuation of Atherosclerosis by Protocatechuic Acid via Inhibition of M1 and Promotion of M2 Macrophage Polarization. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:807-818. [PMID: 30592218 DOI: 10.1021/acs.jafc.8b05719] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Macrophage polarization has a vital impact on the progression of atherosclerosis (AS). Protocatechuic acid (PCA), a flavonol, displays notable atheroprotective effects, but its mechanisms have not been clearly defined. We investigated whether PCA attenuated AS by regulating macrophage polarization. PCA consumption inhibited HCD-induced plaque formation (17.84 and 8.21% in the HCD and HCD with PCA groups, respectively, p < 0.05) and inflammatory responses in apolipoprotein E deficient (ApoE-/-) mice. Moreover, PCA suppressed classically activated macrophage (M1) polarization, which decreased the secretion of nitric oxide synthase (54.63 and 32.86% in the HCD and HCD with PCA groups, respectively, p < 0.05) and proinflammatory factors. PCA promoted alternatively activated macrophage (M2) activation, which increased the expression of arginine I (6.97 and 26.19% in the HCD and HCD with PCA groups, respectively, p < 0.001) and anti-inflammatory factors. PCA also regulated M1-M2 polarization in J774 cells and mouse-bone-marrow-derived macrophages. Finally, PCA reduced PI3K-Akt-mediated nuclear-factor-κB activation, thereby suppressing M1 polarization, and provoked signal-transducers-and-activators-of-transcription-6 phosphorylation and peroxisome-proliferator-activated-receptor-γ activation, leading to enhanced M2 activation. Our data revealed that PCA alleviated AS by regulating M1-M2 conversion.
Collapse
Affiliation(s)
- Yao Liu
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Xu Wang
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Juan Pang
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Hanyue Zhang
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Jing Luo
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Xiaoyun Qian
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Qian Chen
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
| | - Wenhua Ling
- Department of Nutrition, School of Public Health , Sun Yat-Sen University (North Campus) , Guangzhou 510080 , PR China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health , Guangzhou 510080 , PR China
- Guangdong Engineering Technology Center of Nutrition Transformation , Guangzhou 510080 , PR China
| |
Collapse
|
535
|
Akhi R, Wang C, Nissinen AE, Kankaanpää J, Bloigu R, Paju S, Mäntylä P, Buhlin K, Sinisalo J, Pussinen PJ, Hörkkö S. Salivary IgA to MAA-LDL and Oral Pathogens Are Linked to Coronary Disease. J Dent Res 2019; 98:296-303. [PMID: 30669938 DOI: 10.1177/0022034518818445] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
A large body of literature has established the link between periodontal disease and cardiovascular disease. Oxidized low-density lipoproteins (OxLDLs) have a crucial role in atherosclerosis progression through initiation of immunological response. Monoclonal IgM antibodies to malondialdehyde-modified low-density lipoprotein (MDA-LDL) and to malondialdehyde acetaldehyde-modified low-density lipoprotein (MAA-LDL) have been shown to cross-react with the key virulence factors of periodontal pathogens Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans. We have previously shown that salivary IgA antibodies to MAA-LDL cross-react with P. gingivalis in healthy humans. In this study, we aim to assess whether oral mucosal immune response represented by salivary IgA to MAA-LDL and oral pathogens is associated with coronary artery disease (CAD). Also, the molecular mimicry through antibody cross-reaction between salivary IgA to MAA-LDL and oral pathogens was evaluated. The study subjects consisted of 451 patients who underwent a coronary angiography with no CAD ( n = 133), stable CAD ( n = 169), and acute coronary syndrome (ACS, n = 149). Elevated salivary IgA antibody levels to MAA-LDL, Rgp44 (gingipain A hemagglutinin domain of P. gingivalis), and Aa-HSP60 (heat shock protein 60 of A. actinomycetemcomitans) were discovered in stable-CAD and ACS patients when compared to no-CAD patients. In a multinomial regression model adjusted for known cardiovascular risk factors, stable CAD and ACS were associated with IgA to MAA-LDL ( P = 0.016, P = 0.043), Rgp44 ( P = 0.012, P = 0.004), Aa-HSP60 ( P = 0.032, P = 0.030), Tannerella forsythia ( P = 0.002, P = 0.004), Porphyromonas endodontalis ( P = 0.016, P = 0.020), Prevotella intermedia ( P = 0.038, P = 0.005), and with total IgA antibody concentration ( P = 0.002, P = 0.016). Salivary IgA to MAA-LDL showed cross-reactivity with the oral pathogens tested in the study patients. The study highlights an association between salivary IgA to MAA-LDL and atherosclerosis. However, whether salivary IgA to MAA-LDL and the related oral humoral responses play a causal role in the development in the CAD should be elucidated in the future.
Collapse
Affiliation(s)
- R Akhi
- 1 Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,2 Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland.,3 Nordlab, Oulu University Hospital, Oulu, Finland
| | - C Wang
- 1 Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,2 Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland.,3 Nordlab, Oulu University Hospital, Oulu, Finland
| | - A E Nissinen
- 1 Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,2 Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland.,3 Nordlab, Oulu University Hospital, Oulu, Finland
| | - J Kankaanpää
- 1 Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,2 Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland.,3 Nordlab, Oulu University Hospital, Oulu, Finland
| | - R Bloigu
- 4 Medical Informatics and Statistics Research Group Oulu, University of Oulu, Oulu, Finland
| | - S Paju
- 5 Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - P Mäntylä
- 5 Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,6 Institute of Dentistry, University of Eastern Finland, Kuopio, Finland.,7 Kuopio University Hospital, Oral and Maxillofacial Diseases, Kuopio, Finland
| | - K Buhlin
- 5 Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,8 Division of Periodontology, Department of Dental Medicine, Karolinska Institutet, Huddinge, Sweden
| | - J Sinisalo
- 9 HUCH Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - P J Pussinen
- 5 Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - S Hörkkö
- 1 Medical Microbiology and Immunology, Research Unit of Biomedicine, University of Oulu, Oulu, Finland.,2 Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland.,3 Nordlab, Oulu University Hospital, Oulu, Finland
| |
Collapse
|
536
|
Sánchez-Duffhues G, García de Vinuesa A, van de Pol V, Geerts ME, de Vries MR, Janson SG, van Dam H, Lindeman JH, Goumans MJ, Ten Dijke P. Inflammation induces endothelial-to-mesenchymal transition and promotes vascular calcification through downregulation of BMPR2. J Pathol 2019; 247:333-346. [PMID: 30430573 PMCID: PMC6590480 DOI: 10.1002/path.5193] [Citation(s) in RCA: 124] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 10/04/2018] [Accepted: 10/30/2018] [Indexed: 12/27/2022]
Abstract
Endothelial‐to‐mesenchymal transition (EndMT) has been unveiled as a common cause for a multitude of human pathologies, including cancer and cardiovascular disease. Vascular calcification is a risk factor for ischemic vascular disorders and slowing calcification may reduce mortality in affected patients. The absence of early biomarkers hampers the identification of patients at risk. EndMT and vascular calcification are induced upon cooperation between distinct stimuli, including inflammatory cytokines and transforming growth factor beta (TGF‐β) family members. However, how these signaling pathways interplay to promote cell differentiation and eventually vascular calcification is not well understood. Using in vitro and ex vivo analysis in animal models and patient‐derived tissues, we have identified that the pro‐inflammatory cytokines tumor necrosis factor alpha (TNF‐α) and interleukin‐1 beta (IL‐1β) induce EndMT in human primary aortic endothelial cells, thereby sensitizing them for BMP‐9‐induced osteogenic differentiation. Downregulation of the BMP type II receptor BMPR2 is a key event in this process. Rather than compromising BMP canonical signal transduction, loss of BMPR2 results in decreased JNK signaling in ECs, thus enhancing BMP‐9‐induced mineralization. Altogether, our results point at the BMPR2–JNK signaling axis as a key pathway regulating inflammation‐induced EndMT and contributing to calcification. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Gonzalo Sánchez-Duffhues
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Amaya García de Vinuesa
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Vera van de Pol
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Marlieke E Geerts
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Margreet R de Vries
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Stef Gt Janson
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Hans van Dam
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Jan H Lindeman
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Cell and Chemical Biology, Oncode Institute, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
537
|
Gao S, Zhang W, Zhao Q, Zhou J, Wu Y, Liu Y, Yuan Z, Wang L. Curcumin ameliorates atherosclerosis in apolipoprotein E deficient asthmatic mice by regulating the balance of Th2/Treg cells. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 52:129-135. [PMID: 30599892 DOI: 10.1016/j.phymed.2018.09.194] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Revised: 08/31/2018] [Accepted: 09/17/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Allergic asthma and atherosclerosis represent different directions of inflammatory responses of CD4+ T cells, and allergic asthma accelerates atherosclerosis formation. Curcumin could ameliorate the progression of both atherosclerosis and allergic asthma. PURPOSE We aimed to investigate the roles of curcumin in asthma-accelerated atherosclerosis plaque formation, and the change of CD4+ T-cell subsets in this process. METHODS Six to eight-week-old apolipoprotein E-/- (apoE-/-) mice were sensitized and challenged by ovalbumin (OVA) to establish an allergic asthma model, and then received curcumin or vehicle treatment for 8 weeks. RESULTS The accelerated atherosclerosis was induced by allergic asthma accompanied by increased T helper cell (Th)2 and Th17 cells and decreased regulatory T cells (Tregs) in the spleen. After the 8-week treatment with curcumin, the lesion areas in the aortic root in asthmatic mice significantly improved, and the elevated Th2 and Th17 cells significantly decreased, but Tregs markedly increased. Although curcumin treatment markedly reduced the interleukin (IL)-4 and IL-13 in serum and spleen, the elevated IL-17A did not decrease. Moreover, Th1 cells showed no significant change between different groups. The mRNA expression levels of M1 macrophage-related inflammatory factors IL-6, iNOS and IL-1β were markedly elevated in the spleens of asthmatic mice, but significantly decreased after the 8-week treatment with curcumin. CONCLUSION Curcumin ameliorated the aggravation of atherosclerotic lesions and stabilised plaque by modulating the balance of Th2/Tregs in asthmatic apoE-/- mice.
Collapse
Affiliation(s)
- Shanshan Gao
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Weiping Zhang
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Qiang Zhao
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Juan Zhou
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China; Key Laboratory of Molecular Cardiology, Shaanxi Province, China
| | - Yue Wu
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China; Key Laboratory of Molecular Cardiology, Shaanxi Province, China
| | - Yan Liu
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China; Key Laboratory of Environment and Genes Related to Diseases, (Xi'an Jiaotong University), Ministry of Education, China; Key Laboratory of Molecular Cardiology, Shaanxi Province, China; Cardiovascular Research Center, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| | - Lijun Wang
- Department of Cardiovascular Medicine First Affiliated Hospital of Xi'an Jiaotong University, China; Key Laboratory of Molecular Cardiology, Shaanxi Province, China.
| |
Collapse
|
538
|
Nie S, Zeng J, Qin H, Xu X, Zeng J, Yang C, Luo J. Improvement in the blood compatibility of polyvinylidene fluoride membranes via in situ cross-linking polymerization. POLYM ADVAN TECHNOL 2018. [DOI: 10.1002/pat.4525] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Shengqiang Nie
- Engineering Technology Research Center for Materials Protection of Wear and Corrosion of Guizhou Province, University of Guizhou Province, College of Chemistry and Materials Engineering; Guiyang University; Guiyang 550000 China
| | - Jiazhou Zeng
- Engineering Technology Research Center for Materials Protection of Wear and Corrosion of Guizhou Province, University of Guizhou Province, College of Chemistry and Materials Engineering; Guiyang University; Guiyang 550000 China
| | - Hui Qin
- Engineering Technology Research Center for Materials Protection of Wear and Corrosion of Guizhou Province, University of Guizhou Province, College of Chemistry and Materials Engineering; Guiyang University; Guiyang 550000 China
| | - Xiaolu Xu
- Engineering Technology Research Center for Materials Protection of Wear and Corrosion of Guizhou Province, University of Guizhou Province, College of Chemistry and Materials Engineering; Guiyang University; Guiyang 550000 China
| | - Jia Zeng
- Engineering Technology Research Center for Materials Protection of Wear and Corrosion of Guizhou Province, University of Guizhou Province, College of Chemistry and Materials Engineering; Guiyang University; Guiyang 550000 China
| | - Chunlin Yang
- Engineering Technology Research Center for Materials Protection of Wear and Corrosion of Guizhou Province, University of Guizhou Province, College of Chemistry and Materials Engineering; Guiyang University; Guiyang 550000 China
| | - Jun Luo
- Engineering Technology Research Center for Materials Protection of Wear and Corrosion of Guizhou Province, University of Guizhou Province, College of Chemistry and Materials Engineering; Guiyang University; Guiyang 550000 China
| |
Collapse
|
539
|
Affiliation(s)
- Catherine S. Birch
- Institute for Agri‐Food Research & Innovation, School of Natural & Environmental Sciences Newcastle University Newcastle NE1 7RU UK
| | - Graham A. Bonwick
- Institute for Agri‐Food Research & Innovation, School of Natural & Environmental Sciences Newcastle University Newcastle NE1 7RU UK
- Fera Science Limited Sand Hutton, York YO41 1LZ UK
| |
Collapse
|
540
|
McCracken IR, Taylor RS, Henderson NC, Sluimer JC, Baker AH. Unravelling atherosclerotic heterogeneity by single cell RNA sequencing. Curr Opin Lipidol 2018; 29:488-489. [PMID: 30379738 DOI: 10.1097/mol.0000000000000559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Affiliation(s)
| | - Richard S Taylor
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Neil C Henderson
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Judith C Sluimer
- Centre for Cardiovascular Science
- Department of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Andrew H Baker
- Centre for Cardiovascular Science
- Department of Pathology, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
541
|
Mitchell RN. When More Is Less. Circulation 2018; 138:2527-2529. [PMID: 30571358 DOI: 10.1161/circulationaha.118.036650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
542
|
Fernandes M, Patel A, Husi H. C/VDdb: A multi-omics expression profiling database for a knowledge-driven approach in cardiovascular disease (CVD). PLoS One 2018; 13:e0207371. [PMID: 30419069 PMCID: PMC6231654 DOI: 10.1371/journal.pone.0207371] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/30/2018] [Indexed: 12/15/2022] Open
Abstract
The cardiovascular disease (C/VD) database is an integrated and clustered information resource that covers multi-omic studies (microRNA, genomics, proteomics and metabolomics) of cardiovascular-related traits with special emphasis on coronary artery disease (CAD). This resource was built by mining existing literature and public databases and thereafter manual biocuration was performed. To enable integration of omic data from distinct platforms and species, a specific ontology was applied to tie together and harmonise multi-level omic studies based on gene and protein clusters (CluSO) and mapping of orthologous genes (OMAP) across species. CAD continues to be a leading cause of death in the population worldwide, and it is generally thought to be an age-related disease. However, CAD incidence rates are now known to be highly influenced by environmental factors and interactions, in addition to genetic determinants. With the complexity of CAD aetiology, there is a difficulty in research studies to elucidate general elements compared to other cardiovascular diseases. Data from 92 studies, covering 13945 molecular entries (4353 unique molecules) is described, including data descriptors for experimental setup, study design, discovery-validation sample size and associated fold-changes of the differentially expressed molecular features (p-value<0.05). A dedicated interactive web interface, equipped with a multi-parametric search engine, data export and indexing menus are provided for a user-accessible browsing experience. The main aim of this work was the development of a data repository linking clinical information and molecular differential expression in several CVD-related traits from multi-omics studies (genomics, transcriptomics, proteomics and metabolomics). As an example case of how to query and identify data sets within the database framework and concomitantly demonstrate the database utility, we queried CAD-associated studies and performed a systems-level integrative analysis. URL: www.padb.org/cvd
Collapse
Affiliation(s)
- Marco Fernandes
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Alisha Patel
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Holger Husi
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
- Division of Biomedical Sciences, Centre for Health Science, University of the Highlands and Islands, Inverness, United Kingdom
- * E-mail:
| |
Collapse
|
543
|
Abstract
Cardiovascular disease is one of the leading causes of death and disability in the world. Atherosclerosis, characterized by lipid accumulation and chronic inflammation in the vessel wall, is the main feature of cardiovascular disease. Although the amounts of fruits and vegetables present in the diets vary by country, diets, worldwide, contain large amounts of spices; this may have positive or negative effects on the initiation and development of atherosclerosis. In this review, we focused on the potential protective effects of specific nutrients from spices, such as pepper, ginger, garlic, onion, cinnamon and chili, in atherosclerosis and atherosclerotic cardiovascular disease. The mechanisms, epidemiological analysis, and clinical studies focusing on a variety of spices are covered in this review. Based on the integrated information, we aimed to raise specific recommendations for people with different dietary styles for the prevention of atherosclerotic cardiovascular disease through dietary habit adjustments.
Collapse
|
544
|
|
545
|
Jin P, Bian Y, Wang K, Cong G, Yan R, Sha Y, Ma X, Zhou J, Yuan Z, Jia S. Homocysteine accelerates atherosclerosis via inhibiting LXRα-mediated ABCA1/ABCG1-dependent cholesterol efflux from macrophages. Life Sci 2018; 214:41-50. [PMID: 30393020 DOI: 10.1016/j.lfs.2018.10.060] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2018] [Revised: 10/15/2018] [Accepted: 10/26/2018] [Indexed: 11/19/2022]
Abstract
AIMS Macrophage-derived foam-cell formation plays a crucial role in the development of atherosclerosis, and liver X receptor alpha (LXRα) is a key regulator of lipid metabolism in macrophages. Homocysteine (Hcy) is an independent risk factor of atherosclerosis; however, the regulation of lipid metabolism and role of LXRα induced by Hcy in macrophages is still unknown. The present study aimed to investigate the potential role of Hcy in disordered lipid metabolism and atherosclerotic lesions, especially the effects of Hcy on cholesterol efflux in macrophages and the possible mechanisms. MAIN METHODS In vitro, lipid accumulation and cholesterol efflux were evaluated in THP-1 macrophages with Hcy intervention. Real-time quantitative PCR and western blot analyses were used to assess mRNA and protein levels. In vivo, atherosclerotic lesions and lipid profiles were evaluated by methionine diet-induced hyperhomocysteinemia (HHcy) in ApoE-/- mice. The LXRα agonist T0901317 was used to verify the role of LXRα in HHcy-accelerated atherosclerosis. KEY FINDINGS Hcy promoted lipid accumulation and inhibited cholesterol efflux in THP-1 macrophages. HHcy mice showed increased lesion area and lipid accumulation in plaque. Both studies in vitro and in vivo showed decreased expression of ATP binding cassette transporter A1 (ABCA1) and G1 (ABCG1). T0901317 treatment increased ABCA1 and ABCG1 levels; reversed macrophage-derived foam-cell formation in THP-1 macrophages and reduced atherosclerotic lesions in ApoE-/- mice. SIGNIFICANCE Inhibition of LXRα-mediated ABCA1/ABCG1-dependent cholesterol efflux from macrophages is a novel mechanism in Hcy-accelerated atherosclerosis.
Collapse
Affiliation(s)
- Ping Jin
- Department of Clinical Medicine, Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Yitong Bian
- Department of Radiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Kai Wang
- Heart Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Guangzhi Cong
- Heart Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Ru Yan
- Institute of Cardiovascular Diseases, General Hospital of Ningxia Medical University Yinchuan, Ningxia 750001, China
| | - Yong Sha
- Heart Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Xueping Ma
- Heart Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China
| | - Juan Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of the Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zuyi Yuan
- Department of Cardiovascular Medicine, The First Affiliated Hospital of the Medical School, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Shaobin Jia
- Heart Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750001, China; Institute of Cardiovascular Diseases, General Hospital of Ningxia Medical University Yinchuan, Ningxia 750001, China.
| |
Collapse
|
546
|
Abstract
PURPOSE OF REVIEW Atherosclerosis is a chronic inflammatory disease in which subendothelial infiltration of lipoproteins leads to inflamed lesions in arteries. Despite improvements in secondary prevention, most cardiovascular events cannot be avoided with current therapies. This review focuses on novel mechanistic insights on lipid-driven immune activation, which could pave the way for new anti-inflammatory treatments for atherosclerosis. RECENT FINDINGS Immunometabolic interactions can shape the immune response. Within atherosclerotic plaques, macrophages and T cells are the dominant immune cell populations. Using multiple mechanisms, lipoprotein-derived components activate both the innate and adaptive immune systems. Cholesterol crystals and apolipoprotein B-peptides have been shown to activate macrophages and T cells, respectively. Lipoproteins are also important modulators of regulatory T cells that can hamper vascular inflammation. In the liver, T cells can influence hepatic inflammation and lipoprotein metabolism. Hence, there is an intricate crosstalk between the immune system and lipoprotein metabolism. SUMMARY Novel treatments are needed to prevent clinical manifestations of atherosclerosis. Improved understanding of lipid-driven immunometabolic responses is likely to reveal new therapeutic targets.
Collapse
Affiliation(s)
- Anton Gisterå
- Cardiovascular Medicine Unit, Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | |
Collapse
|
547
|
Hypermethylation of the Micro-RNA 145 Promoter Is the Key Regulator for NLRP3 Inflammasome-Induced Activation and Plaque Formation. JACC Basic Transl Sci 2018; 3:604-624. [PMID: 30456333 PMCID: PMC6234615 DOI: 10.1016/j.jacbts.2018.06.004] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 05/14/2018] [Accepted: 06/19/2018] [Indexed: 01/17/2023]
Abstract
miR-145 in vessels decreases with plaque progression. DNMT1 and TET2 dynamic imbalance leads to miR-145 promoter hypermethylation. Reduction of miR-145 activates NLRP3 inflammasome through CD137/NFATc1 signaling. DNMT1 and TET2 could be promising therapeutic candidates for atherosclerosis in the future.
Two major issues are involved in clinical atherosclerosis treatment. First, there are no significant clinical markers for early diagnosis of atherosclerosis. Second, the plaque will not regress once it initiates even if the risk factors are removed. In this paper, the research shows that the hypermethylation level of the microRNA 145 (miR-145) promoter is related to a DNMT1 and TET2 dynamic imbalance. The reduction of miR-145 causes NLRP3 (nucleotide-binding oligomerization domain-like receptor protein 3) inflammasome activation through CD137/NFATc1 signaling. These findings could be a potential target for plaque regression in the future.
Collapse
|
548
|
Wang H, Yang Y, Sun X, Tian F, Guo S, Wang W, Tian Z, Jin H, Zhang Z, Tian Y. Sonodynamic therapy-induced foam cells apoptosis activates the phagocytic PPARγ-LXRα-ABCA1/ABCG1 pathway and promotes cholesterol efflux in advanced plaque. Am J Cancer Res 2018; 8:4969-4984. [PMID: 30429880 PMCID: PMC6217053 DOI: 10.7150/thno.26193] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 09/10/2018] [Indexed: 01/09/2023] Open
Abstract
In advanced atherosclerotic plaques, defective efferocytosis of apoptotic foam cells and decreased cholesterol efflux contribute to lesion progression. In our previous study, we demonstrated that 5-aminolevulinic acid (ALA)-mediated sonodynamic therapy (SDT) could induce foam cells apoptosis via the mitochondrial-caspase pathway. In the current research, we sought to explore ALA-SDT-induced apoptosis of phagocytes and the effects of cholesterol efflux and efferocytosis in advanced apoE-/- mice plaque. Methods: apoE-/- mice fed western diet were treated with ALA-SDT and sacrificed at day 1, day 3, day 7 and day 28 post treatment. THP-1 macrophage-derived foam cells were treated with ALA-SDT. 5 hours later, the supernatant was collected and added to fresh foam cells (phagocytes). Then, the lipid area, efferocytosis, cholesterol efflux, anti-inflammatory reactions and PPARγ-LXRα-ABCA1/ABCG1 pathway were detected in plaque in vivo and in phagocytes in vitro. Results: We found that ALA-SDT induced foam cells apoptosis coupled with efferocytosis and upregulation of Mer tyrosine kinase (MerTK) both in vivo and in vitro. The lipid content in plaque decreased as early as 1 day after ALA-SDT and this tendency persisted until 28 days. The enhancement of phagocytes cholesterol efflux was accompanied by an approximately 40% decrease in free cholesterol and a 24% decrease in total cholesterol in vitro. More importantly, anti-inflammatory factors such as TGFβ and IL-10 were upregulated by ALA-SDT treatment. Finally, we found that PPARγ-LXRα-ABCA1/ABCG1 pathway was activated both in vivo and in vitro by ALA-SDT, which could be blocked by PPARγ siRNA. Conclusions: Activation of PPARγ-LXRα-ABCA1/ABCG1 pathway induced by ALA-SDT treatment engages a virtuous cycle that enhances efferocytosis, cholesterol efflux and anti-inflammatory reactions in advanced plaque in vivo and in phagocytes in vitro.
Collapse
|
549
|
Cai J, Xu M, Zhang X, Li H. Innate Immune Signaling in Nonalcoholic Fatty Liver Disease and Cardiovascular Diseases. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2018; 14:153-184. [PMID: 30230967 DOI: 10.1146/annurev-pathmechdis-012418-013003] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The physiological significance of innate immune signaling lies primarily in its role in host defense against invading pathogens. It is becoming increasingly clear that innate immune signaling also modulates the development of metabolic diseases, especially nonalcoholic fatty liver disease and cardiovascular diseases, which are characterized by chronic, low-grade inflammation due to a disarrangement of innate immune signaling. Notably, recent studies indicate that in addition to regulating canonical innate immune-mediated inflammatory responses (or immune-dependent signaling-induced responses), molecules of the innate immune system regulate pathophysiological responses in multiple organs during metabolic disturbances (termed immune-independent signaling-induced responses), including the disruption of metabolic homeostasis, tissue repair, and cell survival. In addition, emerging evidence from the study of immunometabolism indicates that the systemic metabolic status may have profound effects on cellular immune function and phenotypes through the alteration of cell-intrinsic metabolism. We summarize how the innate immune system interacts with metabolic disturbances to trigger immune-dependent and immune-independent pathogenesis in the context of nonalcoholic fatty liver disease, as representative of metabolic diseases, and cardiovascular diseases.
Collapse
Affiliation(s)
- Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China.,Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha 410013, China
| | - Meng Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China
| | - Xiaojing Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan 430060, China; .,Institute of Model Animals of Wuhan University, Wuhan 430072, China.,Basic Medical School, Wuhan University, Wuhan 430071, China
| |
Collapse
|
550
|
Orsatti CL, Sobreira ML, Sandrim VC, Nahas-Neto J, Orsatti FL, Nahas EAP. Autophagy-related 16-like 1gene polymorphism, risk factors for cardiovascular disease and associated carotid intima-media thickness in postmenopausal women. Clin Biochem 2018; 61:12-17. [PMID: 30236831 DOI: 10.1016/j.clinbiochem.2018.09.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 09/13/2018] [Accepted: 09/16/2018] [Indexed: 12/12/2022]
Abstract
BACKGROUND Early identification of asymptomatic postmenopausal women (PW), who are more predisposed to developing cardiovascular disease (CVD), is an important preventive strategy. Autophagy-related 16-like 1 (ATG16L1) is an autophagy gene known to control host immune responses and is associated with a variety of diseases, including CVD. OBJECTIVE The aim of the study was to associate the ATG16L1 polymorphism variant with subclinical carotid atherosclerosis in asymptomatic PW. STUDY DESIGN This cross-sectional study included 210 Brazilian postmenopausal women (age ≥ 45 years with amenorrhea ≥12 months). Clinical, anthropometric and biochemical assessments were performed to evaluate the cardiovascular risk factors. DNA was extracted from buccal cells and the ATG16L1 (T300A) polymorphism was determined by the polymerase chain reaction (PCR). The carotid intima-media thickness and/or the presence of plaques were evaluated by carotid duplex ultrasound. For statistical analysis, the t-test, logistic regression and analysis of covariance (ANCOVA) were used. RESULTS The presence of the polymorphic allele forATG16L1 (T300A) was found in 77.47% (A/G = 49.87%, G/G = 27.60%). The ATG16L1 (T300A) polymorphism is significantly associated with increased carotid intima-media thickness (IMT) after adjustments of the confounding variables (P < .037). No significant associations were observed between the polymorphism with other risk factors for CVD in PW. CONCLUSION In postmenopausal women, the ATG16L1 (T300A) polymorphism is significantly associated with increased carotid IMT (marker of atherosclerotic disease) after adjustments of the confounding variables (P < .037). Thus, identifying the ATG16L1 polymorphism is an important strategy for screening asymptomatic PW who are more predisposed to developing CVD.
Collapse
Affiliation(s)
- Claudio Lera Orsatti
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil.
| | - Marcone Lima Sobreira
- Department of Surgery, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Valéria Cristina Sandrim
- Department of Pharmacology, Institute of Biosciences of Botucatu, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Jorge Nahas-Neto
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| | - Fabio Lera Orsatti
- Department of Sport Sciences, Federal University of Triângulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Eliana Aguiar Petri Nahas
- Department of Gynecology and Obstetrics, Botucatu Medical School, Sao Paulo State University-UNESP, Botucatu, São Paulo, Brazil
| |
Collapse
|