1
|
Gallo A, Le Goff W, Santos RD, Fichtner I, Carugo S, Corsini A, Sirtori C, Ruscica M. Hypercholesterolemia and inflammation-Cooperative cardiovascular risk factors. Eur J Clin Invest 2025; 55:e14326. [PMID: 39370572 PMCID: PMC11628670 DOI: 10.1111/eci.14326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/02/2024] [Indexed: 10/08/2024]
Abstract
BACKGROUND Maintaining low concentrations of plasma low-density lipoprotein cholesterol (LDLc) over time decreases the number of LDL particles trapped within the artery wall, slows the progression of atherosclerosis and delays the age at which mature atherosclerotic plaques develop. This substantially reduces the lifetime risk of atherosclerotic cardiovascular disease (ASCVD) events. In this context, plaque development and vulnerability result not only from lipid accumulation but also from inflammation. RESULTS Changes in the composition of immune cells, including macrophages, dendritic cells, T cells, B cells, mast cells and neutrophils, along with altered cytokine and chemokine release, disrupt the equilibrium between inflammation and anti-inflammatory mechanisms at plaque sites. Considering that it is not a competition between LDLc and inflammation, but instead that they are partners in crime, the present narrative review aims to give an overview of the main inflammatory molecular pathways linked to raised LDLc concentrations and to describe the impact of lipid-lowering approaches on the inflammatory and lipid burden. Although remarkable changes in LDLc are driven by the most recent lipid lowering combinations, the relative reduction in plasma C-reactive protein appears to be independent of the magnitude of LDLc lowering. CONCLUSION Identifying clinical biomarkers of inflammation (e.g. interleukin-6) and possible targets for therapy holds promise for monitoring and reducing the ASCVD burden in suitable patients.
Collapse
Affiliation(s)
- Antonio Gallo
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Wilfried Le Goff
- Lipidology and Cardiovascular Prevention Unit, Department of Nutrition, APHP, Hôpital Pitié‐SalpètriêreSorbonne Université, INSERM UMR1166ParisFrance
| | - Raul D. Santos
- Academic Research Organization Hospital Israelita Albert Einstein and Lipid Clinic Heart Institute (InCor)University of Sao Paulo Medical School HospitalSao PauloBrazil
| | - Isabella Fichtner
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Stefano Carugo
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
- Department of Clinical Sciences and Community HealthUniversità degli Studi di MilanoMilanItaly
| | - Alberto Corsini
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Cesare Sirtori
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
| | - Massimiliano Ruscica
- Department of Pharmacological and Biomolecular Sciences “Rodolfo Paoletti”Università degli Studi di MilanoMilanItaly
- Department of Cardio‐Thoracic‐Vascular DiseasesFoundation IRCCS Cà Granda Ospedale Maggiore PoliclinicoMilanItaly
| |
Collapse
|
2
|
Yılmaz C, Karaduman A, Tiryaki MM, Güvendi Şengör B, Unkun T, Kültürsay B, Zehir R. Predictive value of the Naples prognostic score for no-reflow phenomenon after saphenous vein graft stenting. Biomark Med 2024:1-10. [PMID: 39711087 DOI: 10.1080/17520363.2024.2443383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND No-reflow is a critical adverse event associated with percutaneous coronary intervention (PCI), particularly during saphenous vein graft (SVG) procedures. The Naples Prognostic Score (NPS) reflects inflammatory status, but its relationship with no-reflow remains unclear. Therefore, we aimed to evaluate the relationship between NPS and no-reflow occurrence following SVG PCI. METHODS We retrospectively analyzed 286 patients who underwent SVG PCI from January 2020 to January 2024, with a median age of 65 years and 85.7% male. Participants were categorized into low NPS (0-2, 48.6%) and high NPS (3-4, 51.4%) groups. Two nested models were developed by adding NPS (continuous and categorical, respectively) to the base model. RESULTS Higher no-reflow rates were noted in the high-NPS group (48.5% vs. 9.5%, p < 0.05). Multivariable regression revealed that a higher NPS significantly increased no-reflow risk, with odds ratios of 5.966 (95% CI: 3.066-11.611) for continuous NPS and 10.110 (95% CI: 3.194-32.002) for categorical NPS. Adding NPS to the base model significantly improved predictive performance (likelihood-ratio test p < 0.001). Model 1 demonstrated the best performance (X2 : 84.857, R2 : 0.468) and discriminative ability (AUC: 0.888). CONCLUSION Our findings suggest that NPS is a strong predictor of no-reflow following SVG PCI.
Collapse
Affiliation(s)
- Cemalettin Yılmaz
- Department of Cardiology, Malazgirt State Hospital, Malazgirt, Mus, Turkey
| | - Ahmet Karaduman
- Department of Cardiology, Kartal Kosuyolu Research and Education Hospital, Kartal, Istanbul, Turkey
| | | | - Büşra Güvendi Şengör
- Department of Cardiology, Kartal Kosuyolu Research and Education Hospital, Kartal, Istanbul, Turkey
| | - Tuba Unkun
- Department of Cardiology, Kartal Kosuyolu Research and Education Hospital, Kartal, Istanbul, Turkey
| | - Barkın Kültürsay
- Department of Cardiology, Tunceli State Hospital, Tunceli, Turkey
| | - Regayip Zehir
- Department of Cardiology, Kartal Kosuyolu Research and Education Hospital, Kartal, Istanbul, Turkey
| |
Collapse
|
3
|
He H, Jiang T, Ding M, Zhu Y, Xu X, Huang Y, Yu W, Ou H. Nox1/PAK1 is required for angiotensin II-induced vascular inflammation and abdominal aortic aneurysm formation. Redox Biol 2024; 79:103477. [PMID: 39721498 DOI: 10.1016/j.redox.2024.103477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Revised: 10/27/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024] Open
Abstract
NADPH oxidase 1 (Nox1) is a major isoform of Nox in vascular smooth muscle cells (VSMCs). VSMC activation and extracellular matrix (ECM) remodelling induce abdominal aortic aneurysm (AAA). In this study, we aim to determine the role of Nox1 in the progression of AAA and explore the underling mechanism. ApoE-/-Nox1SMCko mice in which the Nox1 gene was smooth muscle cell (SMC)-specifically deleted in ApoE-/- background, were infused with angiotensin II (Ang II) for 28 days. We found the Nox1 deficiency reduced AAA formation and increased survival compared with ApoE-/-Nox1y/flox mice. Abdominal aortic ROS and monocyts/macrophages were reduced in the ApoE-/-Nox1SMCko mice after Ang II-infusion. The SMC-specific Nox1 deletion caused less elastin fragments and lower matrix metalloproteinase (MMP) activities in the abdominal aorta. Further, we found the Nox1 protein interacted with p21-activated kinase 1 (PAK1) in Ang II-stimulated VSMCs. The PAK1, controlled by Nox1/ROS, promoted VSMC proliferation, migration and differentiation; this is associated with increased activities of vimentin and cofilin, and cytoskeleton modulation. Moreover, we found that the Nox1/PAK1 activated the downstream MAPKs (ERK1/2, p38 and JNKs) and NF-κB, and upregulated Sp1-mediated MMP2 expression upon Ang II-stimulation. Finally, overexpression of PAK1 in the ApoE-/-Nox1SMCko mice increased vascular elastic fibre degradation, pro-inflammatory cytokine expression and AAA incidence. Therefore, we conclude that Nox1, together with PAK1, facilitates Ang II-induced VSMC activation, vascular inflammation and ECM remodelling, and thus potentiates the AAA formation.
Collapse
Affiliation(s)
- Hui He
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Tianyu Jiang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Meng Ding
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Yuan Zhu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Xiaoting Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Yashuang Huang
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Wenfeng Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China
| | - Hailong Ou
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Guizhou Medical University, Gui'an, 561113, Guizhou, PR China.
| |
Collapse
|
4
|
Xue Q, Zhang L, Wang R, Xu J, Wang C, Gao S, Fang X, Meng C, Lu R, Guo L. Hexavalent chromium reduces testosterone levels by impairing lipophagy and disrupting lipid metabolism homeostasis: Based on a metabolomic analysis. Toxicology 2024; 508:153908. [PMID: 39121936 DOI: 10.1016/j.tox.2024.153908] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/24/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Hexavalent chromium (Cr(VI)) causes testicular damage and reduces testosterone secretion. Testosterone synthesis relies on cholesterol as a raw material, and its availability can be affected by lipophagy. However, the role of lipophagy in Cr(VI)-induced testicular damage and reduced testosterone secretion remains unclear. In this study, we investigated the effect of Cr(VI) on lipid metabolism and lipophagy in the testes of ICR mice. Forty mice were randomly divided into four groups and exposed to different doses of Cr(VI) (0, 75, 100, 125 mg/kg) for thirty days. Cr(VI) increased the rate of sperm abnormalities, decreased testosterone level, and decreased the levels of testosterone synthesis-related proteins, namely steroidogenic acute regulatory (StAR) and 3β-hydroxysteroid dehydrogenase (3β-HSD) proteins. Through metabolomic analysis, Oil Red O staining, and biochemical indicator (triglyceride and total cholesterol) analysis, Cr(VI) was found to disrupt testicular lipid metabolism. Further investigation revealed that Cr(VI) inhibited the AMP-activated protein kinase (AMPK)/sterol regulatory element-binding protein 1 (SREBP1) pathway, elevated levels of the autophagy-related proteins microtubule-associated protein 1 light chain 3B (LC3B) and sequestosome 1 (SQSTM1)/P62 and lipophagy-related proteins Rab7 and Rab10, while increasing colocalization of LC3B and Perilipin2. These findings suggest that Cr(VI) exposure leads to abnormal lipid metabolism in the testes by suppressing the AMPK/SREBP1 pathway and disrupting lipophagy, ultimately reducing testosterone level and inducing testicular damage.
Collapse
Affiliation(s)
- Qian Xue
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Le Zhang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Rui Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Jiayunzhu Xu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Chaofan Wang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Shidi Gao
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Xin Fang
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Chunyang Meng
- Department of Spine Surgery, China-Japan Union Hospital of Jilin University, Changchun, China.
| | - Rifeng Lu
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| | - Li Guo
- Department of Toxicology, School of Public Health, Jilin University, Changchun, China.
| |
Collapse
|
5
|
Zhang W, Wang R, Shi F. Impact of serum apolipoproteins on the prognosis of acute ischemic stroke after thrombolysis. J Stroke Cerebrovasc Dis 2024; 33:107944. [PMID: 39153582 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024] Open
Abstract
OBJECTIVE The purpose of this study is to the relationship between peripheral apolipoproteins and the prognosis of patients with acute ischemic stroke (AIS) after thrombolysis. METHODS A total of 231 AIS patients with thrombolysis was enrolled. Serum apolipoproteins were measured on admission after thrombolysis. All patients enrolled were followed up for 90 days. Their functional outcomes were assessed by the modified Rankin Scale (mRS). Good functional outcome was considered as mRS < 3. Logistic regression was applied to assess the association between serum apolipoproteins and the mRS at 90 days. RESULTS In multivariate analysis,1) ApoB (OR=0.099, 95%CI=0.017∼0.575, p=0.010) and ApoB/ApoA-1(OR=0.113, 95%CI=0.015∼0.868, p=0.036) were independent risk factors of good functional outcome at 90 days. 2). there were significant differences in the mRS score distribution at 90 days in groups according to the ROC cutoff values of ApoB (0.85g/L) and ApoB/ApoA-1 ratio (0.61) (all p<0.05). CONCLUSION Our findings demonstrated ApoB and ApoB/apoA-1 ratio were independent risk factors for good functional outcome at 90 days, and the ApoB level below 0.85g/L and ApoB/ApoA-1 ratio below 0.61 could be associated with a better functional outcome in this study population.
Collapse
Affiliation(s)
- Wenhua Zhang
- Department of Neurology, Hangzhou Traditional Chinese Medicine Hospital affiliated to Zhejiang Chinese Medical University, 453# Tiyuchang Road, Hangzhou 310007, China.
| | - Ruiming Wang
- Department of Neurology, Hangzhou Traditional Chinese Medicine Hospital affiliated to Zhejiang Chinese Medical University, 453# Tiyuchang Road, Hangzhou 310007, China
| | - Fangying Shi
- Department of Neurology, Hangzhou Traditional Chinese Medicine Hospital affiliated to Zhejiang Chinese Medical University, 453# Tiyuchang Road, Hangzhou 310007, China
| |
Collapse
|
6
|
Wang H, Kim SJ, Lei Y, Wang S, Wang H, Huang H, Zhang H, Tsung A. Neutrophil extracellular traps in homeostasis and disease. Signal Transduct Target Ther 2024; 9:235. [PMID: 39300084 PMCID: PMC11415080 DOI: 10.1038/s41392-024-01933-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 06/25/2024] [Accepted: 07/16/2024] [Indexed: 09/22/2024] Open
Abstract
Neutrophil extracellular traps (NETs), crucial in immune defense mechanisms, are renowned for their propensity to expel decondensed chromatin embedded with inflammatory proteins. Our comprehension of NETs in pathogen clearance, immune regulation and disease pathogenesis, has grown significantly in recent years. NETs are not only pivotal in the context of infections but also exhibit significant involvement in sterile inflammation. Evidence suggests that excessive accumulation of NETs can result in vessel occlusion, tissue damage, and prolonged inflammatory responses, thereby contributing to the progression and exacerbation of various pathological states. Nevertheless, NETs exhibit dual functionalities in certain pathological contexts. While NETs may act as autoantigens, aggregated NET complexes can function as inflammatory mediators by degrading proinflammatory cytokines and chemokines. The delineation of molecules and signaling pathways governing NET formation aids in refining our appreciation of NETs' role in immune homeostasis, inflammation, autoimmune diseases, metabolic dysregulation, and cancer. In this comprehensive review, we delve into the multifaceted roles of NETs in both homeostasis and disease, whilst discussing their potential as therapeutic targets. Our aim is to enhance the understanding of the intricate functions of NETs across the spectrum from physiology to pathology.
Collapse
Affiliation(s)
- Han Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Susan J Kim
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Yu Lei
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shuhui Wang
- Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hui Wang
- Department of Medical Genetics, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hai Huang
- Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Hongji Zhang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| | - Allan Tsung
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
7
|
Lindsay S, Li Y. Coarse-grained modeling of annexin A2-induced microdomain formation on a vesicle. Biophys J 2024; 123:2431-2442. [PMID: 38859585 PMCID: PMC11365106 DOI: 10.1016/j.bpj.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 05/04/2024] [Accepted: 06/06/2024] [Indexed: 06/12/2024] Open
Abstract
Annexin A2 (A2)-induced microdomain formation is a key step in biological processes such as Ca2+-mediated exocytosis in neuroendocrine cells. In this work, a total of 15 coarse-grained molecular dynamics simulations were performed on vesicle models having a diameter of approximately 250 Å for 15 μs each using the Martini2 force field. Five simulations were performed in the presence of 10 A2, 5 in the presence of A2 but absence of PIP2, and 5 simulations in the absence of A2 but presence of PIP2. Consistent results were generated among the simulations. A2-induced PIP2 microdomain formation was observed and shown to occur in three phases: A2-vesicle association, localized A2-induced PIP2 clustering, and A2 aggregation driving PIP2 microdomain formation. The relationship between A2 aggregation and PIP2 microdomain formation was quantitatively described using a novel method which calculated the variance among protein and lipid positions via the Fréchet mean. A large reduction in PIP2 variance was observed in the presence of A2 but not in its absence. This reduction in PIP2 variance was proportional to the reduction observed in A2 variance and demonstrates that the observed PIP2 microdomain formation is dependent upon A2 aggregation. The three-phase model of A2-induced microdomain formation generated in this work will serve as a valuable guide for further experimental studies and the development of novel A2 inhibitors. No microdomain formation was observed in the absence of A2 and minimal A2-membrane interaction was observed in the absence of PIP2.
Collapse
Affiliation(s)
- S Lindsay
- Department of Chemistry, East Carolina University, Greenville, North Carolina
| | - Y Li
- Department of Chemistry, East Carolina University, Greenville, North Carolina.
| |
Collapse
|
8
|
Shang J, Ma Y, Liu X, Sun S, Pang X, Zhou R, Huan S, He Y, Xiong B, Zhang XB. Single-particle rotational microrheology enables pathological staging of macrophage foaming and antiatherosclerotic studies. Proc Natl Acad Sci U S A 2024; 121:e2403740121. [PMID: 39102540 PMCID: PMC11331104 DOI: 10.1073/pnas.2403740121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 07/01/2024] [Indexed: 08/07/2024] Open
Abstract
The formation of macrophage-derived foam cells has been recognized as the pathological hallmark of atherosclerotic diseases. However, the pathological evolution dynamics and underlying regulatory mechanisms remain largely unknown. Herein, we introduce a single-particle rotational microrheology method for pathological staging of macrophage foaming and antiatherosclerotic explorations by probing the dynamic changes of lysosomal viscous feature over the pathological evolution progression. The principle of this method involves continuous monitoring of out-of-plane rotation-caused scattering brightness fluctuations of the gold nanorod (AuNR) probe-based microrheometer and subsequent determination of rotational relaxation time to analyze the viscous feature in macrophage lysosomes. With this method, we demonstrated the lysosomal viscous feature as a robust pathological reporter and uncovered three distinct pathological stages underlying the evolution dynamics, which are highly correlated with a pathological stage-dependent activation of the NLRP3 inflammasome-involved positive feedback loop. We also validated the potential of this positive feedback loop as a promising therapeutic target and revealed the time window-dependent efficacy of NLRP3 inflammasome-targeted drugs against atherosclerotic diseases. To our knowledge, the pathological staging of macrophage foaming and the pathological stage-dependent activation of the NLRP3 inflammasome-involved positive feedback mechanism have not yet been reported. These findings provide insights into in-depth understanding of evolutionary features and regulatory mechanisms of macrophage foaming, which can benefit the analysis of effective therapeutical drugs as well as the time window of drug treatment against atherosclerotic diseases in preclinical studies.
Collapse
Affiliation(s)
- Jinhui Shang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Yuan Ma
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Xixuan Liu
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Shijie Sun
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Xiayun Pang
- State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang712083, China
| | - Rui Zhou
- State Key Laboratory of Research and Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi University of Chinese Medicine, Xianyang712083, China
| | - Shuangyan Huan
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Yan He
- Department of Chemistry, Tsinghua University, Beijing100084, China
| | - Bin Xiong
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| | - Xiao-Bing Zhang
- State Key Laboratory for Chemo/Bio-Sensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha410082, China
| |
Collapse
|
9
|
Koynarev V, Borgos KKA, Kohlbrecher J, Porcar L, Nielsen JE, Lund R. Antimicrobial Peptides Increase Line Tension in Raft-Forming Lipid Membranes. J Am Chem Soc 2024; 146:20891-20903. [PMID: 39018511 PMCID: PMC11295182 DOI: 10.1021/jacs.4c05377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/19/2024]
Abstract
The formation of phase separated membrane domains is believed to be essential for the function of the cell. The precise composition and physical properties of lipid bilayer domains play crucial roles in regulating protein activity and governing cellular processes. Perturbation of the domain structure in human cells can be related to neurodegenerative diseases and cancer. Lipid rafts are also believed to be essential in bacteria, potentially serving as targets for antibiotics. An important question is how the membrane domain structure is affected by bioactive and therapeutic molecules, such as surface-active peptides, which target cellular membranes. Here we focus on antimicrobial peptides (AMPs), crucial components of the innate immune system, to gain insights into their interaction with model lipid membranes containing domains. Using small-angle neutron/X-ray scattering (SANS/SAXS), we show that the addition of several natural AMPs (indolicidin, LL-37, magainin II, and aurein 2.2) causes substantial growth and restructuring of the domains, which corresponds to increased line tension. Contrast variation SANS and SAXS results demonstrate that the peptide inserts evenly in both phases, and the increased line tension can be related to preferential and concentration dependent thinning of the unsaturated membrane phase. We speculate that the lateral restructuring caused by the AMPs may have important consequences in affecting physiological functions of real cells. This work thus shines important light onto the complex interactions and lateral (re)organization in lipid membranes, which is relevant for a molecular understanding of diseases and the action of antibiotics.
Collapse
Affiliation(s)
| | | | - Joachim Kohlbrecher
- Laboratory
for Neutron Scattering and Imaging, Paul
Scherrer Institut, Villigen 5232, Switzerland
| | - Lionel Porcar
- Institut
Laue-Langevin, 71 Av. des Martyrs, 38000 Grenoble, France
| | | | - Reidar Lund
- Department
of Chemistry, University of Oslo, Postboks 1033 Blindern, 0315 Oslo, Norway
- Hylleraas
Centre for Quantum Molecular Sciences, University
of Oslo, Postboks 1033 Blindern, 0315 Oslo, Norway
| |
Collapse
|
10
|
Zhang Y, Ding X, Yuan C, Yang Y, Zhang Q, Yao J, Zhang Y, Wang J, Dai Y. Anti-Inflammatory Responses Produced with Nippostrongylus brasiliensis-Derived Uridine via the Mitochondrial ATP-Sensitive Potassium Channel and Its Anti-Atherosclerosis Effect in an Apolipoprotein E Gene Knockout Mouse Model. Biomolecules 2024; 14:672. [PMID: 38927075 PMCID: PMC11201709 DOI: 10.3390/biom14060672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/06/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis (AS) has become the leading cause of cardiovascular disease worldwide. Our previous study had observed that Nippostrongylus brasiliensis (Nb) infection or its derived products could inhibit AS development by inducing an anti-inflammatory response. We performed a metabolic analysis to screen Nb-derived metabolites with anti-inflammation activity and evaluated the AS-prevention effect. We observed that the metabolite uridine had higher expression levels in mice infected with the Nb and ES (excretory-secretory) products and could be selected as a key metabolite. ES and uridine interventions could reduce the pro-inflammatory responses and increase the anti-inflammatory responses in vitro and in vivo. The apolipoprotein E gene knockout (ApoE-/-) mice were fed with a high-fat diet for the AS modeling. Following the in vivo intervention, ES products or uridine significantly reduced serum and liver lipid levels, alleviated the formation of atherosclerosis, and reduced the pro-inflammatory responses in serum or plaques, while the anti-inflammatory responses showed opposite trends. After blocking with 5-HD (5-hydroxydecanoate sodium) in vitro, the mRNA levels of M2 markers were significantly reduced. When blocked with 5-HD in vivo, the degree of atherosclerosis was worsened, the pro-inflammatory responses were increased compared to the uridine group, while the anti-inflammatory responses decreased accordingly. Uridine, a key metabolite from Nippostrongylus brasiliensis, showed anti-inflammatory and anti-atherosclerotic effects in vitro and in vivo, which depend on the activation of the mitochondrial ATP-sensitive potassium channel.
Collapse
Affiliation(s)
- Yingshu Zhang
- School of Public Health, Nanjing Medical University, Longmian Avenue 101, Nanjing 211166, China
| | - Xin Ding
- Jiangsu Provincial Medical Key Laboratory, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Institute of Parasitic Diseases, Yang Xiang 117, Wuxi 214064, China
| | - Caiyi Yuan
- School of Public Health, Nanjing Medical University, Longmian Avenue 101, Nanjing 211166, China
| | - Yougui Yang
- School of Public Health, Nanjing Medical University, Longmian Avenue 101, Nanjing 211166, China
- Jiangsu Provincial Medical Key Laboratory, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Institute of Parasitic Diseases, Yang Xiang 117, Wuxi 214064, China
- Department of Parasitic Disease Control, Lishui District Center for Disease Control and Prevention, Middle Street 17, Nanjing 211200, China
| | - Qiang Zhang
- Jiangsu Provincial Medical Key Laboratory, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Institute of Parasitic Diseases, Yang Xiang 117, Wuxi 214064, China
| | - Jiakai Yao
- Jiangsu Provincial Medical Key Laboratory, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Institute of Parasitic Diseases, Yang Xiang 117, Wuxi 214064, China
| | - Ying Zhang
- Jiangsu Provincial Medical Key Laboratory, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Institute of Parasitic Diseases, Yang Xiang 117, Wuxi 214064, China
| | - Junhong Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Guangzhou Road 300, Nanjing 210029, China
| | - Yang Dai
- School of Public Health, Nanjing Medical University, Longmian Avenue 101, Nanjing 211166, China
- Jiangsu Provincial Medical Key Laboratory, Jiangsu Provincial Key Laboratory on Parasite and Vector Control Technology, National Health Commission Key Laboratory of Parasitic Disease Control and Prevention, Jiangsu Institute of Parasitic Diseases, Yang Xiang 117, Wuxi 214064, China
| |
Collapse
|
11
|
Chen Z, Kong X, Ma Q, Chen J, Zeng Y, Liu H, Wang X, Lu S. The impact of Mycobacterium tuberculosis on the macrophage cholesterol metabolism pathway. Front Immunol 2024; 15:1402024. [PMID: 38873598 PMCID: PMC11169584 DOI: 10.3389/fimmu.2024.1402024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 05/14/2024] [Indexed: 06/15/2024] Open
Abstract
Mycobacterium tuberculosis (Mtb) is an intracellular pathogen capable of adapting and surviving within macrophages, utilizing host nutrients for its growth and replication. Cholesterol is the main carbon source during the infection process of Mtb. Cholesterol metabolism in macrophages is tightly associated with cell functions such as phagocytosis of pathogens, antigen presentation, inflammatory responses, and tissue repair. Research has shown that Mtb infection increases the uptake of low-density lipoprotein (LDL) and cholesterol by macrophages, and enhances de novo cholesterol synthesis in macrophages. Excessive cholesterol is converted into cholesterol esters, while the degradation of cholesterol esters in macrophages is inhibited by Mtb. Furthermore, Mtb infection suppresses the expression of ATP-binding cassette (ABC) transporters in macrophages, impeding cholesterol efflux. These alterations result in the massive accumulation of cholesterol in macrophages, promoting the formation of lipid droplets and foam cells, which ultimately facilitates the persistent survival of Mtb and the progression of tuberculosis (TB), including granuloma formation, tissue cavitation, and systemic dissemination. Mtb infection may also promote the conversion of cholesterol into oxidized cholesterol within macrophages, with the oxidized cholesterol exhibiting anti-Mtb activity. Recent drug development has discovered that reducing cholesterol levels in macrophages can inhibit the invasion of Mtb into macrophages and increase the permeability of anti-tuberculosis drugs. The development of drugs targeting cholesterol metabolic pathways in macrophages, as well as the modification of existing drugs, holds promise for the development of more efficient anti-tuberculosis medications.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiaomin Wang
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| | - Shuihua Lu
- National Clinical Research Center for Infectious Diseases, Shenzhen Third People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
12
|
Govatati S, Kumar R, Boro M, Traylor JG, Orr AW, Lusis AJ, Rao GN. TRIM13 reduces cholesterol efflux and increases oxidized LDL uptake leading to foam cell formation and atherosclerosis. J Biol Chem 2024; 300:107224. [PMID: 38537695 PMCID: PMC11053335 DOI: 10.1016/j.jbc.2024.107224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 03/11/2024] [Accepted: 03/13/2024] [Indexed: 04/25/2024] Open
Abstract
Impaired cholesterol efflux and/or uptake can influence arterial lipid accumulation leading to atherosclerosis. Here, we report that tripartite motif-containing protein 13 (TRIM13), a RING-type E3 ubiquitin ligase, plays a role in arterial lipid accumulation leading to atherosclerosis. Using molecular approaches and KO mouse model, we found that TRIM13 expression was induced both in the aorta and peritoneal macrophages (pMφ) of ApoE-/- mice in response to Western diet (WD) in vivo. Furthermore, proatherogenic cytokine interleukin-1β also induced TRIM13 expression both in pMφ and vascular smooth muscle cells. Furthermore, we found that TRIM13 via ubiquitination and degradation of liver X receptor (LXR)α/β downregulates the expression of their target genes ABCA1/G1 and thereby inhibits cholesterol efflux. In addition, TRIM13 by ubiquitinating and degrading suppressor of cytokine signaling 1/3 (SOCS1/3) mediates signal transducer and activator of transcription 1 (STAT1) activation, CD36 expression, and foam cell formation. In line with these observations, genetic deletion of TRIM13 by rescuing cholesterol efflux and inhibiting foam cell formation protects against diet-induced atherosclerosis. We also found that while TRIM13 and CD36 levels were increased, LXRα/β, ABCA1/G1, and SOCS3 levels were decreased both in Mφ and smooth muscle cells of stenotic human coronary arteries as compared to nonstenotic arteries. More intriguingly, the expression levels of TRIM13 and its downstream signaling molecules were correlated with the severity of stenotic lesions. Together, these observations reveal for the first time that TRIM13 plays a crucial role in diet-induced atherosclerosis, and that it could be a potential drug target against this vascular lesion.
Collapse
Affiliation(s)
- Suresh Govatati
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Raj Kumar
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Monoranjan Boro
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - James G Traylor
- Department of Pathology, Louisiana State University Health Science Center, Shreveport, Louisiana, USA
| | - A Wayne Orr
- Department of Pathology, Louisiana State University Health Science Center, Shreveport, Louisiana, USA
| | - Aldons J Lusis
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, California, USA
| | - Gadiparthi N Rao
- Department of Physiology, University of Tennessee Health Science Center, Memphis, Tennessee, USA.
| |
Collapse
|
13
|
Bernard C, Carotenuto AR, Pugno NM, Fraldi M, Deseri L. Modelling lipid rafts formation through chemo-mechanical interplay triggered by receptor-ligand binding. Biomech Model Mechanobiol 2024; 23:485-505. [PMID: 38060155 PMCID: PMC10963483 DOI: 10.1007/s10237-023-01787-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Accepted: 10/21/2023] [Indexed: 12/08/2023]
Abstract
Cell membranes, mediator of many biological mechanisms from adhesion and metabolism up to mutation and infection, are highly dynamic and heterogeneous environments exhibiting a strong coupling between biochemical events and structural re-organisation. This involves conformational changes induced, at lower scales, by lipid order transitions and by the micro-mechanical interplay of lipids with transmembrane proteins and molecular diffusion. Particular attention is focused on lipid rafts, ordered lipid microdomains rich of signalling proteins, that co-localise to enhance substance trafficking and activate different intracellular biochemical pathways. In this framework, the theoretical modelling of the dynamic clustering of lipid rafts implies a full multiphysics coupling between the kinetics of phase changes and the mechanical work performed by transmembrane proteins on lipids, involving the bilayer elasticity. This mechanism produces complex interspecific dynamics in which membrane stresses and chemical potentials do compete by determining different morphological arrangements, alteration in diffusive walkways and coalescence phenomena, with a consequent influence on both signalling potential and intracellular processes. Therefore, after identifying the leading chemo-mechanical interactions, the present work investigates from a modelling perspective the spatio-temporal evolution of raft domains to theoretically explain co-localisation and synergy between proteins' activation and raft formation, by coupling diffusive and mechanical phenomena to observe different morphological patterns and clustering of ordered lipids. This could help to gain new insights into the remodelling of cell membranes and could potentially suggest mechanically based strategies to control their selectivity, by orienting intracellular functions and mechanotransduction.
Collapse
Affiliation(s)
- Chiara Bernard
- Department of Civil, Environmental and Mechanical Engineering, University of Trento, Trento, Italy
| | - Angelo Rosario Carotenuto
- Department of Structures for Engineering and Architecture, University of Naples "Federico II", Naples, Italy
- Laboratory of Integrated Mechanics and Imaging for Testing and Simulation (LIMITS), University of Naples "Federico II", Naples, Italy
| | - Nicola Maria Pugno
- Department of Civil, Environmental and Mechanical Engineering, University of Trento, Trento, Italy
- Laboratory for Bioinspired, Bionic, Nano, Meta Materials and Mechanics, University of Trento, Trento, Italy
- School of Engineering and Materials Science, Queen Mary University of London, London, UK
| | - Massimiliano Fraldi
- Department of Structures for Engineering and Architecture, University of Naples "Federico II", Naples, Italy
- Laboratory of Integrated Mechanics and Imaging for Testing and Simulation (LIMITS), University of Naples "Federico II", Naples, Italy
- Département de Physique, LPENS, École Normale Supérieure-PSL, Paris, France
| | - Luca Deseri
- Department of Civil, Environmental and Mechanical Engineering, University of Trento, Trento, Italy.
- Department of Mechanical Engineering and Material Sciences, MEMS-SSoE, University of Pittsburgh, Pittsburgh, USA.
- Department of Civil and Environmental Engineering, Carnegie Mellon University, Pittsburgh, USA.
- Department of Mechanical Engineering, Carnegie Mellon University, Pittsburgh, USA.
| |
Collapse
|
14
|
Yang S, Min X, Hu L, Zheng M, Lu S, Zhao M, Jia S. RFX1 regulates foam cell formation and atherosclerosis by mediating CD36 expression. Int Immunopharmacol 2024; 130:111751. [PMID: 38402833 DOI: 10.1016/j.intimp.2024.111751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 02/06/2024] [Accepted: 02/21/2024] [Indexed: 02/27/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis (AS) is a continuously low-grade inflammatory disease, and monocyte-derived macrophages play a vital role in AS pathogenesis. Regulatory factor X1 (RFX1) has been reported to participate in differentiation of various cells. Our previous report showed that RFX1 expression in CD14+ monocytes from AS patients was decreased and closely related to AS development. Macrophages mostly derive from monocytes and play an important role in AS plaque formation and stability. However, the functions of RFX1 in the formation of macrophage-derived foam cells and consequent AS development are unclear. METHODS We explored the effects of RFX1 on oxidation low lipoprotein (ox-LDL)-stimulated foam cell formation and CD36 expression by increasing or silencing Rfx1 expression in mouse peritoneal macrophages (PMAs). The ApoE-/-Rfx1f/f or ApoE-/-Rfx1f/f Lyz2-Cre mice fed a high-fat diet for 24 weeks were used to further examine the effect of RFX1 on AS pathogenesis. We then performed dual luciferase reporter assays to study the regulation of RFX1 for CD36 transcription. RESULTS Our results demonstrate that RFX1 expression was significantly reduced in ox-LDL induced foam cells and negatively correlated with lipid uptake in macrophages. Besides, Rfx1 deficiency in myeloid cells aggravated atherosclerotic lesions in ApoE-/- mice. Mechanistically, RFX1 inhibited CD36 expression by directly regulating CD36 transcription in macrophages. CONCLUSIONS The reduction of RFX1 expression in macrophages is a vital determinant for foam cell formation and the initiation of AS, proving a potential novel approach for the treatment of AS disease.
Collapse
Affiliation(s)
- Shuang Yang
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Xiaoli Min
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Longyuan Hu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Meiling Zheng
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Shuang Lu
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China
| | - Ming Zhao
- Department of Dermatology, Second Xiangya Hospital, Central South University, Hunan Key Laboratory of Medical Epigenomics, Changsha 410011, China; Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China; Key Laboratory of Basic and Translational Research on Immune-Mediated Skin Diseases, Chinese Academy of Medical Sciences, Nanjing, China
| | - Sujie Jia
- Department of Pharmacy, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing 210042, China.
| |
Collapse
|
15
|
Chen X, Song C, Ma X, Tao J, Hu L, Xu Y, Yi Y, Yang X, Jiang L. High lipoprotein(a) concentration is associated with moyamoya disease. Lipids Health Dis 2024; 23:21. [PMID: 38254149 PMCID: PMC10802057 DOI: 10.1186/s12944-024-02015-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Accepted: 01/10/2024] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Moyamoya disease (MMD) has attracted the attention of scholars because of its rarity and unknown etiology. METHODS Data for this study were sourced from the Second Affiliated Hospital of Nanchang University. Regression analyses were conducted to examine the association in Lipoprotein [Lp(a)] and MMD. R and IBM SPSS were conducted. RESULTS A cohort comprising 1012 MMD patients and 2024 controls was established through the propensity score matching method. Compared with controls, MMD patients showed higher median Lp(a) concentrations [18.5 (9.6-37.8) mg/dL vs. 14.9 (7.8-30.5) mg/dL, P < 0.001]. The odds ratios and 95% confidence intervals for Lp(a) were calculated in three models: unadjusted model, model 1 (adjusted for body mass index and systolic blood pressure), and model 2 (adjusted for model 1 plus triglyceride, C-reactive protein, homocysteine, and low-density lipoprotein cholesterol). Results were [1.613 (1.299-2.002), P < 0.001], [1.598 (1.286-1.986), P < 0.001], and [1.661 (1.330-2.074), P < 0.001], respectively. Furthermore, age, sex, or hypertension status had nothing to do with this relationship. CONCLUSIONS Positive relationship exists between Lp(a) and MMD.
Collapse
Affiliation(s)
- Xinyue Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Chenxin Song
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Xianrun Ma
- School of Basic Medical Sciences, Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Junjie Tao
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, 330006, China
| | - Lijuan Hu
- Department of Nursing, Nanchang Medical College, Nanchang, Jiangxi, China
| | - Yuan Xu
- Department of Medical Big Data Center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yingping Yi
- Department of Medical Big Data Center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Xinlei Yang
- Biobank center, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| | - Long Jiang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China.
| |
Collapse
|
16
|
Sviridov D, Bukrinsky M. Neuro-HIV-New insights into pathogenesis and emerging therapeutic targets. FASEB J 2023; 37:e23301. [PMID: 37942865 PMCID: PMC11032165 DOI: 10.1096/fj.202301239rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/10/2023]
Abstract
HIV-associated neurocognitive disorders (HAND) is a term describing a complex set of cognitive impairments accompanying HIV infection. Successful antiretroviral therapy (ART) reduces the most severe forms of HAND, but milder forms affect over 50% of people living with HIV (PLWH). Pathogenesis of HAND in the ART era remains unknown. A variety of pathogenic factors, such as persistent HIV replication in the brain reservoir, HIV proteins released from infected brain cells, HIV-induced neuroinflammation, and some components of ART, have been implicated in driving HAND pathogenesis in ART-treated individuals. Here, we propose another factor-impairment of cholesterol homeostasis and lipid rafts by HIV-1 protein Nef-as a possible contributor to HAND pathogenesis. These effects of Nef on cholesterol may also underlie the effects of other pathogenic factors that constitute the multifactorial nature of HAND pathogenesis. The proposed Nef- and cholesterol-focused mechanism may provide a long-sought unified explanation of HAND pathogenesis that takes into account all contributing factors. Evidence for the impairment by Nef of cellular cholesterol balance, potential effects of this impairment on brain cells, and opportunities to therapeutically target this element of HAND pathogenesis are discussed.
Collapse
Affiliation(s)
- Dmitri Sviridov
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Michael Bukrinsky
- The George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
17
|
Hou J, Liu J, Huang Z, Wang Y, Yao H, Hu Z, Shi C, Xu J, Wang Q. Structure and function of the membrane microdomains in osteoclasts. Bone Res 2023; 11:61. [PMID: 37989999 PMCID: PMC10663511 DOI: 10.1038/s41413-023-00294-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 09/07/2023] [Accepted: 09/18/2023] [Indexed: 11/23/2023] Open
Abstract
The cell membrane structure is closely related to the occurrence and progression of many metabolic bone diseases observed in the clinic and is an important target to the development of therapeutic strategies for these diseases. Strong experimental evidence supports the existence of membrane microdomains in osteoclasts (OCs). However, the potential membrane microdomains and the crucial mechanisms underlying their roles in OCs have not been fully characterized. Membrane microdomain components, such as scaffolding proteins and the actin cytoskeleton, as well as the roles of individual membrane proteins, need to be elucidated. In this review, we discuss the compositions and critical functions of membrane microdomains that determine the biological behavior of OCs through the three main stages of the OC life cycle.
Collapse
Affiliation(s)
- Jialong Hou
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Liu
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhixian Huang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yining Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hanbing Yao
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhenxin Hu
- Department of Spine Surgery, Peking University Fourth School of Clinical Medicine, Beijing, China
| | - Chengge Shi
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiake Xu
- School of Biomedical Sciences, The University of Western Australia, Perth, WA, Australia.
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China.
| | - Qingqing Wang
- Department of Orthopaedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China.
- Department of Orthopaedics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| |
Collapse
|
18
|
Wang K, Zhou W, Hu G, Wang L, Cai R, Tian T. TFEB SUMOylation in macrophages accelerates atherosclerosis by promoting the formation of foam cells through inhibiting lysosomal activity. Cell Mol Life Sci 2023; 80:358. [PMID: 37950772 PMCID: PMC11071895 DOI: 10.1007/s00018-023-04981-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 11/13/2023]
Abstract
Atherosclerosis (AS) is a serious cardiovascular disease. One of its hallmarks is hyperlipidemia. Inhibiting the formation of macrophage foam cells is critical for alleviating AS. Transcription factor EB (TFEB) can limit the formation of macrophage foam cells by upregulating lysosomal activity. We examined whether TFEB SUMOylation is involved in this progress during AS. In this study, we investigated the role of TFEB SUMOylation in macrophages in AS using TFEB SUMOylation deficiency Ldlr-/- (TFEB-KR: Ldlr-/-) transgenic mice and TFEB-KR bone marrow-derived macrophages. We observed that TFEB-KR: Ldlr-/- atherosclerotic mice had thinner plaques and macrophages with higher lysosomal activity when compared to WT: Ldlr-/- mice. TFEB SUMOylation in macrophages decreased after oxidized low-density lipoprotein (OxLDL) treatment in vitro. Compared with wild type macrophages, TFEB-KR macrophages exhibited less lipid deposition after OxLDL treatment. Our study demonstrated that in AS, deSUMOylation of TFEB could inhibit the formation of macrophage foam cells through enhancing lysosomal biogenesis and autophagy, further reducing the accumulation of lipids in macrophages, and ultimately alleviating the development of AS. Thus, TFEB SUMOylation can be a switch to modulate macrophage foam cells formation and used as a potential target for AS therapy.
Collapse
Affiliation(s)
- Kezhou Wang
- Department of Pathology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Zhou
- Department of Urology, Renji Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Gaolei Hu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lifeng Wang
- Department of Ophthalmology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Rd., Shanghai, China
| | - Rong Cai
- Department of Biochemistry and Molecular Cell Biology, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Tian Tian
- Department of Ophthalmology, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University School of Medicine, No. 1665, Kongjiang Rd., Shanghai, China.
| |
Collapse
|
19
|
Teng D, Chen H, Jia W, Ren Q, Ding X, Zhang L, Gong L, Wang H, Zhong L, Yang J. Identification and validation of hub genes involved in foam cell formation and atherosclerosis development via bioinformatics. PeerJ 2023; 11:e16122. [PMID: 37810795 PMCID: PMC10557941 DOI: 10.7717/peerj.16122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/27/2023] [Indexed: 10/10/2023] Open
Abstract
Background Foam cells play crucial roles in all phases of atherosclerosis. However, until now, the specific mechanisms by which these foam cells contribute to atherosclerosis remain unclear. We aimed to identify novel foam cell biomarkers and interventional targets for atherosclerosis, characterizing their potential mechanisms in the progression of atherosclerosis. Methods Microarray data of atherosclerosis and foam cells were downloaded from the Gene Expression Omnibus (GEO) database. Differentially expression genes (DEGs) were screened using the "LIMMA" package in R software. The Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis and Gene Ontology (GO) annotation were both carried out. Hub genes were found in Cytoscape after a protein-protein interaction (PPI) enrichment analysis was carried out. Validation of important genes in the GSE41571 dataset, cellular assays, and tissue samples. Results A total of 407 DEGs in atherosclerosis and 219 DEGs in foam cells were identified, and the DEGs in atherosclerosis were mainly involved in cell proliferation and differentiation. CSF1R and PLAUR were identified as common hub genes and validated in GSE41571. In addition, we also found that the expression of CSF1R and PLAUR gradually increased with the accumulation of lipids and disease progression in cell and tissue experiments. Conclusion CSF1R and PLAUR are key hub genes of foam cells and may play an important role in the biological process of atherosclerosis. These results advance our understanding of the mechanism behind atherosclerosis and potential therapeutic targets for future development.
Collapse
Affiliation(s)
- Da Teng
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
- Qingdao University, Qingdao, China
| | - Hongping Chen
- Department of Cardiology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
| | - Wenjuan Jia
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
- Qingdao University, Qingdao, China
| | - Qingmiao Ren
- The Precision Medicine Laboratory, The First Hospital of Lanzhou University, Lanzhou, China
| | - Xiaoning Ding
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Lihui Zhang
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
- Qingdao University, Qingdao, China
| | - Lei Gong
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Hua Wang
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Lin Zhong
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
| | - Jun Yang
- Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China
- Qingdao University, Qingdao, China
| |
Collapse
|
20
|
Marques-da-Silva D, Lagoa R. Rafting on the Evidence for Lipid Raft-like Domains as Hubs Triggering Environmental Toxicants' Cellular Effects. Molecules 2023; 28:6598. [PMID: 37764374 PMCID: PMC10536579 DOI: 10.3390/molecules28186598] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/07/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
The plasma membrane lipid rafts are cholesterol- and sphingolipid-enriched domains that allow regularly distributed, sub-micro-sized structures englobing proteins to compartmentalize cellular processes. These membrane domains can be highly heterogeneous and dynamic, functioning as signal transduction platforms that amplify the local concentrations and signaling of individual components. Moreover, they participate in cell signaling routes that are known to be important targets of environmental toxicants affecting cell redox status and calcium homeostasis, immune regulation, and hormonal functions. In this work, the evidence that plasma membrane raft-like domains operate as hubs for toxicants' cellular actions is discussed, and suggestions for future research are provided. Several studies address the insertion of pesticides and other organic pollutants into membranes, their accumulation in lipid rafts, or lipid rafts' disruption by polychlorinated biphenyls (PCBs), benzo[a]pyrene (B[a]P), and even metals/metalloids. In hepatocytes, macrophages, or neurons, B[a]P, airborne particulate matter, and other toxicants caused rafts' protein and lipid remodeling, oxidative changes, or amyloidogenesis. Different studies investigated the role of the invaginated lipid rafts present in endothelial cells in mediating the vascular inflammatory effects of PCBs. Furthermore, in vitro and in vivo data strongly implicate raft-localized NADPH oxidases, the aryl hydrocarbon receptor, caveolin-1, and protein kinases in the toxic mechanisms of occupational and environmental chemicals.
Collapse
Affiliation(s)
- Dorinda Marques-da-Silva
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| | - Ricardo Lagoa
- LSRE—Laboratory of Separation and Reaction Engineering and LCM—Laboratory of Catalysis and Materials, School of Management and Technology, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal;
- ALiCE—Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal
- School of Technology and Management, Polytechnic Institute of Leiria, Morro do Lena-Alto do Vieiro, 2411-901 Leiria, Portugal
| |
Collapse
|
21
|
Deng Q, Li H, Yue X, Guo C, Sun Y, Ma C, Gao J, Wu Y, Du B, Yang J, Zhang C, Zhang W. Smooth muscle liver kinase B1 inhibits foam cell formation and atherosclerosis via direct phosphorylation and activation of SIRT6. Cell Death Dis 2023; 14:542. [PMID: 37607939 PMCID: PMC10444762 DOI: 10.1038/s41419-023-06054-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 08/01/2023] [Accepted: 08/08/2023] [Indexed: 08/24/2023]
Abstract
Foam cell formation is a hallmark of the early phase of atherosclerosis. Growing evidence has demonstrated that vascular smooth muscle cells (VSMCs) comprise a considerable proportion of foam cells. Liver kinase B1 (LKB1) plays a crucial part in cardiovascular diseases. However, the role of LKB1 in VSMC-derived foam cell formation and atherosclerosis remains unclear. To explore the effects of LKB1 on VSMC-derived foam cell formation and atherosclerosis, we generated smooth muscle-specific LKB1 knockout (LKB1SMKO) mice by crossbreeding LKB1flox/flox mice with SM22α-CreERT2 mice. LKB1 expression decreased in plaque-loaded aortas and oxidized low-density lipoprotein (oxLDL)-treated VSMCs. Compared with controls, atherosclerosis development was exacerbated in LKB1SMKO mice via the promotion of VSMC-derived foam cell formation. Conversely, LKB1 overexpression inhibited lipid uptake and foam cell formation in VSMCs. Mechanistically, LKB1 binds to SIRT6 and directly phosphorylates and activates it, thereby reducing lectin-like oxLDL receptor-1 (LOX-1) via SIRT6-dependent histone deacetylation. Finally, adeno-associated virus (AAV)-mediated LOX-1 deficiency in smooth muscle ameliorated atherosclerosis in LKB1SMKO mice. Our findings suggest that LKB1 may modulate VSMC-derived foam cell formation and atherosclerosis via the phosphorylation and activation of SIRT6.
Collapse
Affiliation(s)
- Qiming Deng
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Hongxuan Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Xiaolin Yue
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chenghu Guo
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Yuanyuan Sun
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chang Ma
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Jiangang Gao
- School of Life Science and Key Laboratory of the Ministry of Education for Experimental Teratology, Shandong University, Jinan, China
| | - Yue Wu
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Bin Du
- Department of Cardiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Jianmin Yang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Cheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| | - Wencheng Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
22
|
Mohanta O, Ray A, Jena S, Sahoo A, Panda SS, Das PK, Nayak S, Panda PC. Mesosphaerum suaveolens Essential Oil Attenuates Inflammatory Response and Oxidative Stress in LPS-Stimulated RAW 264.7 Macrophages by Regulating NF-κB Signaling Pathway. Molecules 2023; 28:5817. [PMID: 37570786 PMCID: PMC10420984 DOI: 10.3390/molecules28155817] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/21/2023] [Accepted: 07/28/2023] [Indexed: 08/13/2023] Open
Abstract
Mesosphaerum suaveolens (L.) Kuntze (Syn. Hyptis suaveolens (L.) Poit.) is a wild essential-oil-bearing plant having multiple uses in traditional medicine, perfumery, food, agriculture, and pharmaceutical industries. The present paper is the first report on the in vitro anti-inflammatory effects of the leaf essential oil of M. suaveolens (MSLEO) and unravels its molecular mechanism in LPS-stimulated RAW 264.7 macrophage cells. GC-MS analysis of the essential oil (EO) isolated from the leaves by hydro-distillation led to the identification of 48 constituents, accounting for 90.55% of the total oil, and β-caryophyllene (16.17%), phyllocladene (11.85%), abietatriene (11.46%), and spathulenol (7.89%) were found to be the major components. MSLEO treatment had no effect on the viability of RAW 264.7 cells up to a concentration of 100 μg/mL, and the EO was responsible for a reduction in proinflammatory cytokines like IL-6, IL-1β, and TNF-α, a decrease in intracellular ROS production, and the restoration of oxidative damage by elevating the levels of endogenous antioxidative enzymes like CAT, SOD, GPx, and GSH. RT-qPCR analysis indicated that MSLEO reduced the mRNA expression levels of iNOS and COX-2 as compared to the LPS-induced group. In addition, a confocal microscopy analysis showed that MSLEO inhibited the translocation of NF-κB from the cytosol to the nucleus. The results of this experiment demonstrate that MSLEO possesses significant anti-inflammatory potential by preventing the activation of NF-κB, which, in turn, inhibits the downstream expression of other inflammatory mediators associated with the activation of the NF-κB pathway in LPS-induced RAW 264.7 cells. Thus, the leaf essential oil of M. suaveolens may prove to be a promising therapeutic agent for the treatment of inflammation, and targeting the NF-κB signaling pathway may be considered as an attractive approach for anti-inflammatory therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Pratap Chandra Panda
- Centre for Biotechnology, Siksha ‘O’ Anusandhan (Deemed to be University), Kalinga Nagar, Bhubaneswar 751003, India
| |
Collapse
|
23
|
Chong SY, Wang X, van Bloois L, Huang C, Syeda NS, Zhang S, Ting HJ, Nair V, Lin Y, Lou CKL, Benetti AA, Yu X, Lim NJY, Tan MS, Lim HY, Lim SY, Thiam CH, Looi WD, Zharkova O, Chew NWS, Ng CH, Bonney GK, Muthiah M, Chen X, Pastorin G, Richards AM, Angeli V, Storm G, Wang JW. Injectable liposomal docosahexaenoic acid alleviates atherosclerosis progression and enhances plaque stability. J Control Release 2023; 360:344-364. [PMID: 37406819 DOI: 10.1016/j.jconrel.2023.06.035] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 06/12/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory vascular disease that is characterized by the accumulation of lipids and immune cells in plaques built up inside artery walls. Docosahexaenoic acid (DHA, 22:6n-3), an omega-3 polyunsaturated fatty acid (PUFA), which exerts anti-inflammatory and antioxidant properties, has long been purported to be of therapeutic benefit to atherosclerosis patients. However, large clinical trials have yielded inconsistent data, likely due to variations in the formulation, dosage, and bioavailability of DHA following oral intake. To fully exploit its potential therapeutic effects, we have developed an injectable liposomal DHA formulation intended for intravenous administration as a plaque-targeted nanomedicine. The liposomal formulation protects DHA against chemical degradation and increases its local concentration within atherosclerotic lesions. Mechanistically, DHA liposomes are readily phagocytosed by activated macrophages, exert potent anti-inflammatory and antioxidant effects, and inhibit foam cell formation. Upon intravenous administration, DHA liposomes accumulate preferentially in atherosclerotic lesional macrophages and promote polarization of macrophages towards an anti-inflammatory M2 phenotype, resulting in attenuation of atherosclerosis progression in both ApoE-/- and Ldlr-/- experimental models. Plaque composition analysis demonstrates that liposomal DHA inhibits macrophage infiltration, reduces lipid deposition, and increases collagen content, thus improving the stability of atherosclerotic plaques against rupture. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) further reveals that DHA liposomes can partly restore the complex lipid profile of the plaques to that of early-stage plaques. In conclusion, DHA liposomes offer a promising approach for applying DHA to stabilize atherosclerotic plaques and attenuate atherosclerosis progression, thereby preventing atherosclerosis-related cardiovascular events.
Collapse
Affiliation(s)
- Suet Yen Chong
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Xiaoyuan Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Louis van Bloois
- Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands
| | - Chenyuan Huang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nilofer Sayed Syeda
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Sitong Zhang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hui Jun Ting
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Vaarsha Nair
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Yuanzhe Lin
- Department of Biomedical Engineering, National University of Singapore, 117583 Singapore, Singapore
| | - Charles Kang Liang Lou
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Ayca Altay Benetti
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - Xiaodong Yu
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Nicole Jia Ying Lim
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Michelle Siying Tan
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore
| | - Hwee Ying Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Sheau Yng Lim
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Chung Hwee Thiam
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Wen Donq Looi
- Bruker Daltonics, Bruker Singapore Pte. Ltd., 138671 Singapore, Singapore
| | - Olga Zharkova
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Nicholas W S Chew
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Department of Cardiology, National University Heart Centre, National University Hospital, 119074 Singapore, Singapore
| | - Cheng Han Ng
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Glenn Kunnath Bonney
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, National University Hospital, 119074 Singapore, Singapore
| | - Mark Muthiah
- Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Division of Gastroenterology and Hepatology, Department of Medicine, National University Hospital, 119074 Singapore, Singapore; National University Centre for Organ Transplantation, National University Health System, 119074 Singapore, Singapore
| | - Xiaoyuan Chen
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 119074 Singapore, Singapore; Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, Faculty of Engineering, National University of Singapore, 117575 Singapore, Singapore; Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Giorgia Pastorin
- Department of Pharmacy, Faculty of Science, National University of Singapore, 117543 Singapore, Singapore
| | - A Mark Richards
- Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore
| | - Veronique Angeli
- Immunology Translational Research Programme, Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, 117456 Singapore, Singapore; Immunology Programme, Life Sciences Institute, National University of Singapore, 117456 Singapore, Singapore
| | - Gert Storm
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Pharmaceutics, Faculty of Science, Utrecht University, 3584 CG Utrecht, the Netherlands; Department of Biomaterials, Science and Technology, Faculty of Science and Technology, University of Twente, 7522 NB Enschede, the Netherlands.
| | - Jiong-Wei Wang
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228 Singapore, Singapore; Cardiovascular Research Institute, Yong Loo Lin School of Medicine, National University of Singapore, 117599 Singapore, Singapore; Nanomedicine Translational Research Programme, Centre for NanoMedicine, Yong Loo Lin School of Medicine, National University of Singapore, 117609 Singapore, Singapore; Department of Physiology, National University of Singapore, 117593 Singapore, Singapore.
| |
Collapse
|
24
|
Shao B, Afshinnia F, Mathew AV, Ronsein GE, Thornock C, Irwin AD, Kansal M, Rao PS, Dobre M, Al-Kindi S, Weir MR, Go A, He J, Chen J, Feldman H, Bornfeldt KE, Pennathur S. Low concentrations of medium-sized HDL particles predict incident CVD in chronic kidney disease patients. J Lipid Res 2023; 64:100381. [PMID: 37100172 PMCID: PMC10323925 DOI: 10.1016/j.jlr.2023.100381] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 04/18/2023] [Accepted: 04/19/2023] [Indexed: 04/28/2023] Open
Abstract
Patients with chronic kidney disease (CKD) are at high risk for CVD. However, traditional CVD risk factors cannot completely explain the increased risk. Altered HDL proteome is linked with incident CVD in CKD patients, but it is unclear whether other HDL metrics are associated with incident CVD in this population. In the current study, we analyzed samples from two independent prospective case-control cohorts of CKD patients, the Clinical Phenotyping and Resource Biobank Core (CPROBE) and the Chronic Renal Insufficiency Cohort (CRIC). We measured HDL particle sizes and concentrations (HDL-P) by calibrated ion mobility analysis and HDL cholesterol efflux capacity (CEC) by cAMP-stimulated J774 macrophages in 92 subjects from the CPROBE cohort (46 CVD and 46 controls) and in 91 subjects from the CRIC cohort (34 CVD and 57 controls). We tested associations of HDL metrics with incident CVD using logistic regression analysis. No significant associations were found for HDL-C or HDL-CEC in either cohort. Total HDL-P was only negatively associated with incident CVD in the CRIC cohort in unadjusted analysis. Among the six sized HDL subspecies, only medium-sized HDL-P was significantly and negatively associated with incident CVD in both cohorts after adjusting for clinical confounders and lipid risk factors with odds ratios (per 1-SD) of 0.45 (0.22-0.93, P = 0.032) and 0.42 (0.20-0.87, P = 0.019) for CPROBE and CRIC cohorts, respectively. Our observations indicate that medium-sized HDL-P-but not other-sized HDL-P or total HDL-P, HDL-C, or HDL-CEC-may be a prognostic cardiovascular risk marker in CKD.
Collapse
Affiliation(s)
- Baohai Shao
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA.
| | - Farsad Afshinnia
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Anna V Mathew
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Graziella E Ronsein
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Carissa Thornock
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Angela D Irwin
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Mayank Kansal
- Department of Cardiology, University of Illinois at Chicago, Chicago, IL, USA
| | - Panduranga S Rao
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Mirela Dobre
- Division of Nephrology and Hypertension, Case Western Reserve University, Cleveland, OH, USA
| | - Sadeer Al-Kindi
- Division of Nephrology and Hypertension, Case Western Reserve University, Cleveland, OH, USA
| | - Matthew R Weir
- Division of Nephrology, Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Alan Go
- Department of Health System Science, Kaiser Permanente Bernard J. Tyson School of Medicine, Pasadena, CA, USA
| | - Jiang He
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Jing Chen
- Department of Epidemiology, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA; Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Harold Feldman
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Karin E Bornfeldt
- Department of Medicine, UW Medicine Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Subramaniam Pennathur
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA; Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
25
|
Size matters: HDL particle populations and the risk of infection. Nat Rev Cardiol 2023; 20:279-280. [PMID: 36792718 DOI: 10.1038/s41569-023-00844-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2023]
|
26
|
iPSC-Derived Macrophages: The Differentiation Protocol Affects Cell Immune Characteristics and Differentiation Trajectories. Int J Mol Sci 2022; 23:ijms232416087. [PMID: 36555728 PMCID: PMC9781144 DOI: 10.3390/ijms232416087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
The generation of human macrophages from induced pluripotent stem cells (iMacs) is a rapidly developing approach used to create disease models, screen drugs, study macrophage-pathogen interactions and develop macrophage-based cell therapy. To generate iMacs, different types of protocols have been suggested, all thought to result in the generation of similar iMac populations. However, direct comparison of iMacs generated using different protocols has not been performed. We have compared the productivity, the differentiation trajectories and the characteristics of iMacs generated using two widely used protocols: one based on the formation of embryoid bodies and the induction of myeloid differentiation by only two cytokines, interleukin-3 and macrophage colony-stimulating factor, and the other utilizing multiple exogenous factors for iMac generation. We report inter-protocol differences in the following: (i) protocol productivity; (ii) dynamic changes in the expression of genes related to inflammation and lipid homeostasis following iMac differentiation and (iii) the transcriptomic profiles of terminally differentiated iMacs, including the expression of genes involved in inflammatory response, antigen presentation and lipid homeostasis. The results document the dependence of fine iMac characteristics on the type of differentiation protocol, which is important for further development of the field, including the development of iMac-based cell therapy.
Collapse
|
27
|
There is urgent need to treat atherosclerotic cardiovascular disease risk earlier, more intensively, and with greater precision: A review of current practice and recommendations for improved effectiveness. Am J Prev Cardiol 2022; 12:100371. [PMID: 36124049 PMCID: PMC9482082 DOI: 10.1016/j.ajpc.2022.100371] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/10/2022] [Accepted: 08/05/2022] [Indexed: 12/12/2022] Open
Abstract
Atherosclerotic cardiovascular disease (ASCVD) is epidemic throughout the world and is etiologic for such acute cardiovascular events as myocardial infarction, ischemic stroke, unstable angina, and death. ASCVD also impacts risk for dementia, chronic kidney disease peripheral arterial disease and mobility, impaired sexual response, and a host of other visceral impairments that adversely impact the quality and rate of progression of aging. The relationship between low-density lipoprotein cholesterol (LDL-C) and risk for ASCVD is one of the most highly established and investigated issues in the entirety of modern medicine. Elevated LDL-C is a necessary condition for atherogenesis induction. Basic scientific investigation, prospective longitudinal cohorts, and randomized clinical trials have all validated this association. Yet despite the enormous number of clinical trials which support the need for reducing the burden of atherogenic lipoprotein in blood, the percentage of high and very high-risk patients who achieve risk stratified LDL-C target reductions is low and has remained low for the last thirty years. Atherosclerosis is a preventable disease. As clinicians, the time has come for us to take primordial and primary prevention more serously. Despite a plethora of therapeutic approaches, the large majority of patients at risk for ASCVD are poorly or inadequately treated, leaving them vulnerable to disease progression, acute cardiovascular events, and poor aging due to loss of function in multiple visceral organs. Herein we discuss the need to greatly intensify efforts to reduce risk, decrease disease burden, and provide more comprehensive and earlier risk assessment to optimally prevent ASCVD and its complications. Evidence is presented to support that treatment should aim for far lower goals in cholesterol management, should take into account many more factors than commonly employed today and should begin significantly earlier in life.
Collapse
|
28
|
Schieffer E, Schieffer B. The rationale for the treatment of long-Covid symptoms – A cardiologist's view. Front Cardiovasc Med 2022; 9:992686. [PMID: 36186977 PMCID: PMC9520195 DOI: 10.3389/fcvm.2022.992686] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 08/24/2022] [Indexed: 11/13/2022] Open
Abstract
The ongoing coronavirus disease 2019 pandemic left us with thousands of patients suffering from neurological, cardiovascular, and psychiatric disorders named post-acute sequelae of COVID-19 or just long-Covid. In parallel, the vaccination campaigns against SARS-CoV-2 spike protein saved millions of lives worldwide but long-Covid symptoms also appeared rarely following vaccination with a strong overlap to the “canonical” long-Covid symptoms. A therapeutic strategy targeting both, post-VAC and post-SARS-CoV-2 long-Covid symptoms is warranted since exposure to the S-protein either by vaccination or SARS-CoV-2 infection may trigger identical immuno-inflammatory cascades resulting in long-Covid symptoms.
Collapse
|
29
|
Krüger K, Tirekoglou P, Weyh C. Immunological mechanisms of exercise therapy in dyslipidemia. Front Physiol 2022; 13:903713. [PMID: 36003652 PMCID: PMC9393246 DOI: 10.3389/fphys.2022.903713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/07/2022] [Indexed: 12/05/2022] Open
Abstract
Numerous studies demonstrated the strong link between dyslipidemia and the cardiovascular risk. Physical activity and exercise represent effective prevention and therapy strategies for dyslipidemia and at the same time counteract numerous comorbidities that often accompany the disease. The physiological mechanisms are manifold, and primary mechanisms might be an increased energy consumption and associated adaptations of the substrate metabolism. Recent studies showed that there are bidirectional interactions between dyslipidemia and the immune system. Thus, abnormal blood lipids may favor pro-inflammatory processes, and at the same time inflammatory processes may also promote dyslipidemia. Physical activity has been shown to affect numerous immunological processes and has primarily anti-inflammatory effects. These are manifested by altered leukocyte subtypes, cytokine patterns, stress protein expression, and by reducing hallmarks of immunosenescence. The aim of this review is to describe the effects of exercise on the treatment dyslipidemia and to discuss possible immunological mechanisms against the background of the current literature.
Collapse
Affiliation(s)
- Karsten Krüger
- Department of Exercise Physiology and Sports Therapy, Institute of Sport Science, Justus-Liebig-University Giessen, Giessen, Germany
| | | | | |
Collapse
|
30
|
Sviridov D, Miller YI, Bukrinsky MI. Trained Immunity and HIV Infection. Front Immunol 2022; 13:903884. [PMID: 35874772 PMCID: PMC9304701 DOI: 10.3389/fimmu.2022.903884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
Findings that certain infections induce immunity not only against the causing agent, but also against an unrelated pathogen have intrigued investigators for many years. Recently, underlying mechanisms of this phenomenon have started to come to light. It was found that the key cells responsible for heterologous protection are innate immune cells such as natural killer cells (NKs), dendritic cells, and monocytes/macrophages. These cells are 'primed' by initial infection, allowing them to provide enhanced response to subsequent infection by the same or unrelated agent. This phenomenon of innate immune memory was termed 'trained immunity'. The proposed mechanism for trained immunity involves activation by the first stimulus of metabolic pathways that lead to epigenetic changes, which maintain the cell in a "trained" state, allowing enhanced responses to a subsequent stimulus. Innate immune memory can lead either to enhanced responses or to suppression of subsequent responses ('tolerance'), depending on the strength and length of the initial stimulation of the immune cells. In the context of HIV infection, innate memory induced by infection is not well understood. In this Hypothesis and Theory article, we discuss evidence for HIV-induced trained immunity in human monocytes, its possible mechanisms, and implications for HIV-associated co-morbidities.
Collapse
Affiliation(s)
- Dmitri Sviridov
- Laboratory of Lipoproteins and Atherosclerosis, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC, Australia
| | - Yury I. Miller
- Department of Medicine, University of California, San Diego, San Diego, CA, United States
| | - Michael I. Bukrinsky
- Department of Microbiology, Immunology and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, United States
| |
Collapse
|
31
|
Busnelli M, Manzini S, Colombo A, Franchi E, Bonacina F, Chiara M, Arnaboldi F, Donetti E, Ambrogi F, Oleari R, Lettieri A, Horner D, Scanziani E, Norata GD, Chiesa G. Lack of ApoA-I in ApoEKO Mice Causes Skin Xanthomas, Worsening of Inflammation, and Increased Coronary Atherosclerosis in the Absence of Hyperlipidemia. Arterioscler Thromb Vasc Biol 2022; 42:839-856. [PMID: 35587694 PMCID: PMC9205301 DOI: 10.1161/atvbaha.122.317790] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Background: HDL (high-density lipoprotein) and its major protein component, apoA-I (apolipoprotein A-I), play a unique role in cholesterol homeostasis and immunity. ApoA-I deficiency in hyperlipidemic, atheroprone mice was shown to drive cholesterol accumulation and inflammatory cell activation/proliferation. The present study was aimed at investigating the impact of apoA-I deficiency on lipid deposition and local/systemic inflammation in normolipidemic conditions. Methods: ApoE deficient mice, apoE/apoA-I double deficient (DKO) mice, DKO mice overexpressing human apoA-I, and C57Bl/6J control mice were fed normal laboratory diet until 30 weeks of age. Plasma lipids were quantified, atherosclerosis development at the aortic sinus and coronary arteries was measured, skin ultrastructure was evaluated by electron microscopy. Blood and lymphoid organs were characterized through histological, immunocytofluorimetric, and whole transcriptome analyses. Results: DKO were characterized by almost complete HDL deficiency and by plasma total cholesterol levels comparable to control mice. Only DKO showed xanthoma formation and severe inflammation in the skin-draining lymph nodes, whose transcriptome analysis revealed a dramatic impairment in energy metabolism and fatty acid oxidation pathways. An increased presence of CD4+ T effector memory cells was detected in blood, spleen, and skin-draining lymph nodes of DKO. A worsening of atherosclerosis at the aortic sinus and coronary arteries was also observed in DKO versus apoE deficient. Human apoA-I overexpression in the DKO background was able to rescue the skin phenotype and halt atherosclerosis development. Conclusions: HDL deficiency, in the absence of hyperlipidemia, is associated with severe alterations of skin morphology, aortic and coronary atherosclerosis, local and systemic inflammation.
Collapse
Affiliation(s)
- Marco Busnelli
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Stefano Manzini
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Alice Colombo
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Elsa Franchi
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Matteo Chiara
- Department of Biosciences (M.C., D.H.), Università degli Studi di Milano, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy (M.C., D.H.)
| | - Francesca Arnaboldi
- Department of Biomedical Sciences for Health (F. Arnaboldi, E.D.), Università degli Studi di Milano, Italy
| | - Elena Donetti
- Department of Biomedical Sciences for Health (F. Arnaboldi, E.D.), Università degli Studi di Milano, Italy
| | - Federico Ambrogi
- Department of Clinical Sciences and Community Health (F. Ambrogi), Università degli Studi di Milano, Italy
| | - Roberto Oleari
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - Antonella Lettieri
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| | - David Horner
- Department of Biosciences (M.C., D.H.), Università degli Studi di Milano, Italy.,Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies, National Research Council, Bari, Italy (M.C., D.H.)
| | - Eugenio Scanziani
- Department of Veterinary Medicine (E.S.), Università degli Studi di Milano, Italy.,Mouse and Animal Pathology Laboratory (MAPLab), Fondazione UniMi, Milan, Italy (E.S.)
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy.,Centro per lo Studio dell'Aterosclerosi, Bassini Hospital, Cinisello B, Milan, Italy (G.D.N.)
| | - Giulia Chiesa
- Department of Pharmacological and Biomolecular Sciences (M.B., S.M., A.C., E.F., F.B., R.O., A.L., G.D.N., G.C.), Università degli Studi di Milano, Italy
| |
Collapse
|
32
|
Roncato R, Angelini J, Pani A, Talotta R. Lipid rafts as viral entry routes and immune platforms: A double-edged sword in SARS-CoV-2 infection? Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159140. [PMID: 35248801 PMCID: PMC8894694 DOI: 10.1016/j.bbalip.2022.159140] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/13/2022] [Accepted: 02/25/2022] [Indexed: 12/15/2022]
Abstract
Lipid rafts are nanoscopic compartments of cell membranes that serve a variety of biological functions. They play a crucial role in viral infections, as enveloped viruses such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) can exploit rafts to enter or quit target cells. On the other hand, lipid rafts contribute to the formation of immune synapses and their proper functioning is a prerequisite for adequate immune response and viral clearance. In this narrative review we dissect the panorama focusing on this singular aspect of cell biology in the context of SARS-CoV-2 infection and therapy. A lipid raft-mediated mechanism can be hypothesized for many drugs recommended or considered for the treatment of SARS-CoV-2 infection, such as glucocorticoids, antimalarials, immunosuppressants and antiviral agents. Furthermore, the additional use of lipid-lowering agents, like statins, may affect the lipid composition of membrane rafts and thus influence the processes occurring in these compartments. The combination of drugs acting on lipid rafts may be successful in the treatment of more severe forms of the disease and should be reserved for further investigation.
Collapse
Affiliation(s)
- Rossana Roncato
- Experimental and Clinical Pharmacology Unit, Centro di Riferimento Oncologico di Aviano (CRO), Istituto di Ricovero e Cura a carattere Scientifico (IRCCS), via Gallini, 33081 Aviano (PN), Italy
| | - Jacopo Angelini
- Clinical Pharmacology Institute, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), via Pozzuolo, 33100 Udine, Italy
| | - Arianna Pani
- Toxicology Department of Oncology and Hemato-Oncology, University of Milan, via Vanvitelli, 20133 Milan, Italy
| | - Rossella Talotta
- Department of Clinical and Experimental Medicine, Rheumatology Unit, AOU "Gaetano Martino", University of Messina, 98100 Messina, Italy
| |
Collapse
|
33
|
Kanal Y, Şeyda Kanal HE, Yakut İ, Özen Y, Özbay MB, Gülcihan Balci K, Yayla C. CRP Albumin Ratio May Predict No Reflow in Patients Undergoing Percutaneous Coronary Intervention for Saphenous Vein Graft Stenosis. Angiology 2022:33197221098277. [PMID: 35500071 DOI: 10.1177/00033197221098277] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Many hypotheses have been proposed to explain no-reflow (NR). Some of these hypotheses, state that NR may be caused by damage to the vascular endothelium and an inflammatory process. In a recent study that did not include patients with coronary artery bypass graft (CABG), the ratio of C-reactive protein (CRP) to albumin (CAR) was found to be associated with NR. Our study aims to evaluate the relationship between CAR and NR in patients who underwent percutaneous coronary intervention (PCI) for saphenous vein graft (SVG). In this retrospective study, among the patients with CABG who underwent primary or elective coronary angiography, 242 patients who underwent PCI to the SVG were selected. The incidence of NR was 19.8% (n = 48). Diabetes mellitus, left ventricular ejection fraction (LVEF), stent length, and CAR were found as independent predictors of NR in multivariate logistic regression analysis (P < .05). Using a cut-off level of .930, the CAR predicted NR with a sensitivity of 75% and a specificity of 73% (AUC: .814, 95% CI: .749-.879, P < .001). The CAR was a better predictor than both stent length and LVEF. CAR was found to be the strongest predictor of NR in our study.
Collapse
Affiliation(s)
- Yücel Kanal
- Department of Cardiology, 233154Tokat State Hospital, Tokat, Turkey
| | | | - İdris Yakut
- Department of Cardiology, Ankara Gazi Mustafa Kemal Vocatıonal and Envıronmental Dıseases Hospıtal, Ankara, Turkey
| | - Yasin Özen
- Department of Cardiology, Sivas Sample Hospital, Sivas, Turkey
| | | | - Kevser Gülcihan Balci
- Department of Cardiology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| | - Cagri Yayla
- Department of Cardiology, University of Health Sciences, Ankara City Hospital, Ankara, Turkey
| |
Collapse
|
34
|
Hafiane A, Gianopoulos I, Sorci-Thomas MG, Daskalopoulou SS. Current models of apolipoprotein A-I lipidation by adenosine triphosphate binding cassette transporter A1. Curr Opin Lipidol 2022; 33:139-145. [PMID: 34581311 DOI: 10.1097/mol.0000000000000786] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The primary cardioprotective function of high-density lipoprotein (HDL) is to remove excess cellular free cholesterol (FC) from peripheral tissues and deliver it to the liver. Here, we summarize recent research that examines apolipoprotein A-I (apoA-I) lipidation models by adenosine triphosphate binding cassette transporter A1 (ABCA1) and discuss its relevance in atherosclerotic cardiovascular disease (ASCVD). RECENT FINDINGS The first step in HDL formation involves the interaction between apoA-I and ABCA1, where ABCA1 mediates the removal of FC and phospholipids from lipid-laden macrophages to form discoidal nascent HDL (nHDL). However, there are currently no clear-cut systematic models that characterize HDL formation. A number of recent studies have investigated the importance of apoA-I C- and N-terminal domains required for optimal cholesterol efflux and nHDL production. Furthermore, functional ABCA1 is required for direct or indirect binding to apoA-I where ABCA1 dimer-monomer interconversion facilitates apoA-I lipidation from plasma membrane microdomains. Microparticles are also another lipid source for apoA-I solubilization into nHDL. SUMMARY ApoA-I and ABCA1 are key factors in macrophage-mediated cholesterol efflux and nHDL production. Understanding of the key steps in HDL formation may unlock the therapeutic potential of HDL and improve clinical management of ASCVD.
Collapse
Affiliation(s)
- Anouar Hafiane
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Ioanna Gianopoulos
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
| | - Mary G Sorci-Thomas
- Division of Endocrinology, Metabolism and Clinical Nutrition, Department of Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada
- Division of Internal Medicine, Department of Medicine, Faculty of Medicine, McGill University Health Centre, McGill University Montreal, Montreal, Canada
| |
Collapse
|
35
|
Woollett LA, Catov JM, Jones HN. Roles of maternal HDL during pregnancy. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159106. [PMID: 34995789 DOI: 10.1016/j.bbalip.2021.159106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 11/11/2021] [Accepted: 12/05/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND High density lipoproteins (HDL) were first linked to cardiovascular disease (CVD) over 30 years ago when an inverse relationship was shown between CVD and HDL-cholesterol levels. It is now apparent that HDL composition and function, not cholesterol levels, are the pertinent measurements describing HDL's role in various disease processes, especially those with subclinical or overt inflammation. SCOPE OF REVIEW Pregnancy is also an inflammatory state. When inflammation becomes excessive during pregnancy, there is an increased risk for adverse outcomes that affect the health of the mother and fetus, including preterm birth and preeclampsia. Though studies on HDL during pregnancy are limited, recent evidence demonstrates that HDL composition and function change during pregnancy and in women with adverse outcomes. GENERAL SIGNIFICANCE In this review, we will discuss how HDL may play a role in maintaining a healthy pregnancy and how impairments in function could lead to pregnancies with adverse outcomes.
Collapse
Affiliation(s)
- Laura A Woollett
- Department of Pathology and Laboratory Medicine, University of Cincinnati Medical School, Cincinnati, OH, United States of America.
| | - Janet M Catov
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Pittsburgh School of Medicine and Magee Women's Research Institute, Pittsburgh, PA, United States of America.
| | - Helen N Jones
- Center for Research in Perinatal Outcomes, Department of Physiology and Functional Genomics, University of Florida, Gainesville, FL, United States of America; Center for Research in Perinatal Outcomes, Department of Obstetrics and Gynecology, University of Florida, Gainesville, FL, United States of America.
| |
Collapse
|
36
|
Lee-Rueckert M, Lappalainen J, Kovanen PT, Escola-Gil JC. Lipid-Laden Macrophages and Inflammation in Atherosclerosis and Cancer: An Integrative View. Front Cardiovasc Med 2022; 9:777822. [PMID: 35237673 PMCID: PMC8882850 DOI: 10.3389/fcvm.2022.777822] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 01/18/2022] [Indexed: 12/12/2022] Open
Abstract
Atherosclerotic arterial plaques and malignant solid tumors contain macrophages, which participate in anaerobic metabolism, acidosis, and inflammatory processes inherent in the development of either disease. The tissue-resident macrophage populations originate from precursor cells derived from the yolk sac and from circulating bone marrow-derived monocytes. In the tissues, they differentiate into varying functional phenotypes in response to local microenvironmental stimulation. Broadly categorized, the macrophages are activated to polarize into proinflammatory M1 and anti-inflammatory M2 phenotypes; yet, noticeable plasticity allows them to dynamically shift between several distinct functional subtypes. In atherosclerosis, low-density lipoprotein (LDL)-derived cholesterol accumulates within macrophages as cytoplasmic lipid droplets thereby generating macrophage foam cells, which are involved in all steps of atherosclerosis. The conversion of macrophages into foam cells may suppress the expression of given proinflammatory genes and thereby initiate their transcriptional reprogramming toward an anti-inflammatory phenotype. In this particular sense, foam cell formation can be considered anti-atherogenic. The tumor-associated macrophages (TAMs) may become polarized into anti-tumoral M1 and pro-tumoral M2 phenotypes. Mechanistically, the TAMs can regulate the survival and proliferation of the surrounding cancer cells and participate in various aspects of tumor formation, progression, and metastasis. The TAMs may accumulate lipids, but their type and their specific roles in tumorigenesis are still poorly understood. Here, we discuss how the phenotypic and functional plasticity of macrophages allows their multifunctional response to the distinct microenvironments in developing atherosclerotic lesions and in developing malignant tumors. We also discuss how the inflammatory reactions of the macrophages may influence the development of atherosclerotic plaques and malignant tumors, and highlight the potential therapeutic effects of targeting lipid-laden macrophages in either disease.
Collapse
Affiliation(s)
| | | | - Petri T. Kovanen
- Wihuri Research Institute, Helsinki, Finland
- *Correspondence: Petri T. Kovanen
| | - Joan Carles Escola-Gil
- Institut d'Investigacions Biomèdiques (IIB) Sant Pau and CIBER de Diabetes y Enfermedades Metabólicas Asociadas, Barcelona, Spain
- Joan Carles Escola-Gil
| |
Collapse
|
37
|
Skorupski WJ, Grygier M, Lesiak M, Kałużna-Oleksy M. Coronary Stent Thrombosis in COVID-19 Patients: A Systematic Review of Cases Reported Worldwide. Viruses 2022; 14:v14020260. [PMID: 35215853 PMCID: PMC8876054 DOI: 10.3390/v14020260] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/19/2022] [Accepted: 01/26/2022] [Indexed: 12/12/2022] Open
Abstract
Approximately 5 million percutaneous coronary interventions are performed worldwide annually. Therefore, stent-related complications pose a serious public health concern. Stent thrombosis, although rare, is usually catastrophic, often associated with extensive myocardial infarction or death. Because little progress has been made in outcomes following stent thrombosis, ongoing research is focusing on further understanding the predictors as well as frequency and timing in various patient subgroups. Coronavirus disease-2019 (COVID-19), a viral illness caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2), activates inflammatory mechanisms that potentially create a prothrombotic environment and increases the risk of local micro thromboembolism and all types of stent thrombosis. In-stent thrombosis occurrence increased during the COVID-19 pandemic, however, there is still lack of comprehensive studies describing this population. This review and worldwide analysis of coronary stent thrombosis cases related to COVID-19 summarizes all available data.
Collapse
|
38
|
HDL and Lipid Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1377:49-61. [DOI: 10.1007/978-981-19-1592-5_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
39
|
Vachon L, Smaani A, Tessier N, Jean G, Demers A, Milasan A, Ardo N, Jarry S, Villeneuve L, Alikashani A, Finherty V, Ruiz M, Sorci-Thomas MG, Mayer G, Martel C. Downregulation of low-density lipoprotein receptor mRNA in lymphatic endothelial cells impairs lymphatic function through changes in intracellular lipids. Theranostics 2022; 12:1440-1458. [PMID: 35154499 PMCID: PMC8771568 DOI: 10.7150/thno.58780] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 12/20/2021] [Indexed: 11/18/2022] Open
Abstract
Rationale: Impairment in lymphatic transport is associated with the onset and progression of atherosclerosis in animal models. The downregulation of low-density-lipoprotein receptor (LDLR) expression, rather than increased circulating cholesterol level per se, is involved in early atherosclerosis-related lymphatic dysfunction. Enhancing lymphatic function in Ldlr-/- mice with a mutant form of VEGF-C (VEGF-C 152s), a selective VEGFR-3 agonist, successfully delayed atherosclerotic plaque onset when mice were subsequently fed a high-fat diet. However, the specific mechanisms by which LDLR protects against lymphatic function impairment is unknown. Methods and results: We have thus injected wild-type and Pcsk9-/- mice with an adeno-associated virus type 1 expressing a shRNA for silencing Ldlr in vivo. We herein report that lymphatic contractility is reduced upon Ldlr dowregulation in wild-type mice only. Our in vitro experiments reveal that a decrease in LDLR expression at the mRNA level reduces the chromosome duplication phase and the protein expression of VEGFR-3, a membrane-bound key lymphatic marker. Furthermore, it also significantly reduced the levels of 18 lipid subclasses, including key constituents of lipid rafts as well as the transcription of several genes involved in cholesterol biosynthesis and cellular and metabolic processes. Exogenous PCSK9 only reduces lymphatic endothelial-LDLR at the protein level and does not affect lymphatic endothelial cell integrity. This puts forward that PCSK9 may act upon lymphatic muscle cells to mediate its effect on lymphatic contraction capacity in vivo. Conclusion: Our results suggest that treatments that specifically palliate the down regulation of LDLR mRNA in lymphatic endothelial cells preserve the integrity of the lymphatic endothelium and sustain lymphatic function, a prerequisite player in atherosclerosis.
Collapse
Affiliation(s)
- Laurent Vachon
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Ali Smaani
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Nolwenn Tessier
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Gabriel Jean
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Annie Demers
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Andreea Milasan
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Nadine Ardo
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
- Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Stéphanie Jarry
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Louis Villeneuve
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | | | - Vincent Finherty
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| | - Matthieu Ruiz
- Department of Nutrition, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Metabolomics platform, Montreal, Quebec, Canada
| | | | - Gaétan Mayer
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
- Faculty of Pharmacy, Université de Montréal, Montreal, QC, Canada
| | - Catherine Martel
- Department of Medicine, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
- Montreal Heart Institute, Research Center, Montreal, Quebec, Canada
| |
Collapse
|
40
|
Essential Oils from Zingiber striolatum Diels Attenuate Inflammatory Response and Oxidative Stress through Regulation of MAPK and NF-κB Signaling Pathways. Antioxidants (Basel) 2021; 10:antiox10122019. [PMID: 34943122 PMCID: PMC8698606 DOI: 10.3390/antiox10122019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/02/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
Zingiber striolatum Diels (Z. striolatum), a widely popular vegetable in China, is famous for its medicinal and nutritional values. However, the anti-inflammatory effects of essential oil from Z. striolatum (EOZS) remain unclear. In this study, EOZS from seven regions in China were extracted and analyzed by GC–MS. LPS-induced RAW264.7 cells and 12-O-Tetradecanoylphorbol 13-acetate (TPA)-stimulated mice were used to evaluate the anti-inflammatory effects of EOZS. Results show that 116 compounds were identified in EOZS from seven locations. Samples 2, 4 and 5 showed the best capability on DPPH radical scavenging and NO inhibition. They also significantly reduced the production of ROS, pro-inflammatory cytokines, macrophage morphological changes, migration and phagocytic capability. Transcriptomics revealed MAPK and NF-κB signaling pathways may be involved in the anti-inflammatory mechanism, and the predictions were proven by Western blotting. In TPA-induced mice, EOZS reduced the degree of ear swelling and local immune cell infiltration by blocking the activation of MAPK and NF-κB signaling pathways, which was consistent with the in vitro experimental results. Our research unveils the antioxidant capability and potential molecular mechanism of EOZS in regulating inflammatory response, and suggests the application of EOZS as a natural antioxidant and anti-inflammatory agent in the pharmaceutical and functional food industries.
Collapse
|
41
|
Rotllan N, Camacho M, Tondo M, Diarte-Añazco EMG, Canyelles M, Méndez-Lara KA, Benitez S, Alonso N, Mauricio D, Escolà-Gil JC, Blanco-Vaca F, Julve J. Therapeutic Potential of Emerging NAD+-Increasing Strategies for Cardiovascular Diseases. Antioxidants (Basel) 2021; 10:1939. [PMID: 34943043 PMCID: PMC8750485 DOI: 10.3390/antiox10121939] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide. Aging and/or metabolic stress directly impact the cardiovascular system. Over the last few years, the contributions of altered nicotinamide adenine dinucleotide (NAD+) metabolism to aging and other pathological conditions closely related to cardiovascular diseases have been intensively investigated. NAD+ bioavailability decreases with age and cardiometabolic conditions in several mammalian tissues. Compelling data suggest that declining tissue NAD+ is commonly related to mitochondrial dysfunction and might be considered as a therapeutic target. Thus, NAD+ replenishment by either genetic or natural dietary NAD+-increasing strategies has been recently demonstrated to be effective for improving the pathophysiology of cardiac and vascular health in different experimental models, as well as human health, to a lesser extent. Here, we review and discuss recent experimental evidence illustrating that increasing NAD+ bioavailability, particularly by the use of natural NAD+ precursors, may offer hope for new therapeutic strategies to prevent and treat cardiovascular diseases.
Collapse
Affiliation(s)
- Noemi Rotllan
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
| | - Mercedes Camacho
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
- CIBER de Enfermedades Cardiovasculares, CIBERCV, 28029 Madrid, Spain
| | - Mireia Tondo
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
- Department of Biochemistry, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain
| | - Elena M. G. Diarte-Añazco
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
| | - Marina Canyelles
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
| | - Karen Alejandra Méndez-Lara
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
| | - Sonia Benitez
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
| | - Núria Alonso
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
- Department of Endocrinology & Nutrition, Hospital Universitari Germans Trias i Pujol, 08916 Barcelona, Spain
| | - Didac Mauricio
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
- Department of Endocrinology & Nutrition, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain
| | - Joan Carles Escolà-Gil
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
| | - Francisco Blanco-Vaca
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
- Department of Biochemistry, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain
| | - Josep Julve
- Institut de Recerca i d’Investigació Biomèdica de l’Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, 08041 Barcelona, Spain; (N.R.); (M.C.); (E.M.G.D.-A.); (M.C.); (K.A.M.-L.); (S.B.)
- Department of Biochemistry and Molecular Biology, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas, CIBERDEM, 28029 Madrid, Spain; (N.A.); (D.M.)
| |
Collapse
|
42
|
Roberts LB, Kapoor P, Howard JK, Shah AM, Lord GM. An update on the roles of immune system-derived microRNAs in cardiovascular diseases. Cardiovasc Res 2021; 117:2434-2449. [PMID: 33483751 PMCID: PMC8562329 DOI: 10.1093/cvr/cvab007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 01/08/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVD) are a leading cause of human death worldwide. Over the past two decades, the emerging field of cardioimmunology has demonstrated how cells of the immune system play vital roles in the pathogenesis of CVD. MicroRNAs (miRNAs) are critical regulators of cellular identity and function. Cell-intrinsic, as well as cell-extrinsic, roles of immune and inflammatory cell-derived miRNAs have been, and continue to be, extensively studied. Several 'immuno-miRNAs' appear to be specifically expressed or demonstrate greatly enriched expression within leucocytes. Identification of miRNAs as critical regulators of immune system signalling pathways has posed the question of whether and how targeting these molecules therapeutically, may afford opportunities for disease treatment and/or management. As the field of cardioimmunology rapidly continues to advance, this review discusses findings from recent human and murine studies which contribute to our understanding of how leucocytes of innate and adaptive immunity are regulated-and may also regulate other cell types, via the actions of the miRNAs they express, in the context of CVD. Finally, we focus on available information regarding miRNA regulation of regulatory T cells and argue that targeted manipulation of miRNA regulated pathways in these cells may hold therapeutic promise for the treatment of CVD and associated risk factors.
Collapse
Affiliation(s)
- Luke B Roberts
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Puja Kapoor
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Jane K Howard
- School of Life Course Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
| | - Ajay M Shah
- School of Cardiovascular Medicine and Sciences, King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Graham M Lord
- School of Immunology and Microbial Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK
- Faculty of Biology, Medicine and Health, University of Manchester, 46 Grafton Street, Manchester M13 9NT, UK
| |
Collapse
|
43
|
Anti-Oxidative and Anti-Inflammatory Activities of Astragalus membranaceus Fermented by Lactiplantibacillus plantarum on LPS-Induced RAW 264.7 Cells. FERMENTATION 2021. [DOI: 10.3390/fermentation7040252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Astragalus membranaceus (AM) has been used for anti-oxidative, anti-inflammatory, anti-cancer, and immunomodulatory activities. In this study, we confirmed that the anti-oxidative and anti-inflammatory effects of AM were enhanced after it was fermented by Lactiplantibacillus plantarum. The anti-oxidative effect was measured by 2,2-Diphenyl-1-picrylhydrazyl (DPPH) radical levels, total phenolic contents (TPC), reducing power, and H2O2 levels. AM-LP MG5145 and MG5276 showed higher free radical scavenging activity than AM-NF (51.22%). In addition, AM-LP MG5145 and MG5276 showed higher TPC (49.11 mg GAE/mL), reducing power (OD700 = 0.37), and H2O2 (1.71 µM) than AM-NF. The calycosin contents determined to AM-NF (17.24 ng/mL), AM-LP MG5145 (139.94 ng/mL), and MG5276 (351.01 ng/mL) using UPLC-ESI-MS/MS. Anti-inflammatory effects were analyzed by investigating the inhibitory effects of fermented AM on cytotoxicity, NO production, and mRNA expression of COX-2, iNOS, NF-κB, and TNF-α in LPS-induced RAW 264.7 cells. AM-LP MG5145 and MG5276 showed no cytotoxicity. AM-LP MG5145 (50.86%) and MG5276 (51.66%) inhibited NO production in LPS-induced RAW 264.7 cells. Moreover, AM-LP MG5145 and MG5276 downregulated macrophage iNOS, COX2, TNF-α, and NF-κB expression. In conclusion, A. membranaceus fermented by L. plantarum MG5145 and MG5276 can be used in cosmetics and health foods as natural antioxidant compounds.
Collapse
|
44
|
Ververs FA, Engelen SE, Nuboer R, Vastert B, van der Ent CK, Van't Land B, Garssen J, Monaco C, Boes M, Schipper HS. Immunometabolic factors in adolescent chronic disease are associated with Th1 skewing of invariant Natural Killer T cells. Sci Rep 2021; 11:20082. [PMID: 34635725 PMCID: PMC8505552 DOI: 10.1038/s41598-021-99580-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Accepted: 09/20/2021] [Indexed: 11/17/2022] Open
Abstract
Invariant Natural Killer T (iNKT) cells respond to the ligation of lipid antigen-CD1d complexes via their T-cell receptor and are implicated in various immunometabolic diseases. We considered that immunometabolic factors might affect iNKT cell function. To this end, we investigated iNKT cell phenotype and function in a cohort of adolescents with chronic disease and immunometabolic abnormalities. We analyzed peripheral blood iNKT cells of adolescents with cystic fibrosis (CF, n = 24), corrected coarctation of the aorta (CoA, n = 25), juvenile idiopathic arthritis (JIA, n = 20), obesity (OB, n = 20), and corrected atrial septal defect (ASD, n = 25) as controls. To study transcriptional differences, we performed RNA sequencing on a subset of obese patients and controls. Finally, we performed standardized co-culture experiments using patient plasma, to investigate the effect of plasma factors on iNKT cell function. We found comparable iNKT cell numbers across patient groups, except for reduced iNKT cell numbers in JIA patients. Upon ex-vivo activation, we observed enhanced IFN-γ/IL-4 cytokine ratios in iNKT cells of obese adolescents versus controls. The Th1-skewed iNKT cell cytokine profile of obese adolescents was not explained by a distinct transcriptional profile of the iNKT cells. Co-culture experiments with patient plasma revealed that across all patient groups, obesity-associated plasma factors including LDL-cholesterol, leptin, and fatty-acid binding protein 4 (FABP4) coincided with higher IFN-γ production, whereas high HDL-cholesterol and insulin sensitivity (QUICKI) coincided with higher IL-4 production. LDL and HDL supplementation in co-culture studies confirmed the effects of lipoproteins on iNKT cell cytokine production. These results suggest that circulating immunometabolic factors such as lipoproteins may be involved in Th1 skewing of the iNKT cell cytokine response in immunometabolic disease.
Collapse
Affiliation(s)
- Francesca A Ververs
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | - Roos Nuboer
- Department of Pediatrics, Meander Medical Center Amersfoort, Amersfoort, The Netherlands
| | - Bas Vastert
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Cornelis K van der Ent
- Department of Pediatric Pulmonology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Belinda Van't Land
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Center of Excellence Immunology, Danone Nutricia Research, Utrecht, The Netherlands
| | - Johan Garssen
- Center of Excellence Immunology, Danone Nutricia Research, Utrecht, The Netherlands
- Division Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Beta Faculty, Utrecht University, Utrecht, The Netherlands
| | - Claudia Monaco
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Marianne Boes
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatric Rheumatology and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Henk S Schipper
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands.
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Department of Pediatric Cardiology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
45
|
Altered HDL Proteome Predicts Incident CVD in Chronic Kidney Disease Patients. J Lipid Res 2021; 62:100135. [PMID: 34634315 PMCID: PMC8566900 DOI: 10.1016/j.jlr.2021.100135] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 10/04/2021] [Accepted: 10/05/2021] [Indexed: 12/31/2022] Open
Abstract
Patients with chronic kidney disease (CKD) are at high risk for CVD. However, traditional lipid risk factors, including low HDL levels, cannot completely explain the increased risk. Altered HDL proteome is linked with both CVD and CKD, but the role of HDL proteins in incident CVD events in patients with CKD is unknown. In this prospective case-control study, we used targeted proteomics to quantify 31 HDL proteins in 92 subjects (46 incident new CVD and 46 one-to-one matched controls) at various stages of CKD. We tested associations of HDL proteins with incident CVD using matched logistic regression analysis. In the model fully adjusted for clinical confounders, lipid levels, C-reactive protein, and proteinuria, no significant associations were found for HDL-C, but we observed inverse associations between levels of HDL proteins paraoxonase/arylesterase 1 (PON1), paraoxonase/arylesterase 3 (PON3), and LCAT and incident CVD. Odds ratios (per 1 SD) were 0.38 (0.18–0.97, P = 0.042), 0.42 (0.20–0.92, P = 0.031), and 0.30 (0.11–0.83, P = 0.020) for PON1, PON3, and LCAT, respectively. Apolipoprotein A-IV remained associated with incident CVD in CKD patients in models adjusted for clinical confounders and lipid levels but lost significance with the addition of C-reactive protein and proteinuria to the model. In conclusion, levels of four HDL proteins, PON1, PON3, LCAT, and apolipoprotein A-IV, were found to be inversely associated with incident CVD events in CKD patients. Our observations indicate that HDLs' protein cargo, but not HDL-C levels, can serve as a marker—and perhaps mediator—for elevated CVD risk in CKD patients.
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW This study reviews the mechanisms of HDL cholesterol immunomodulation in the context of the mechanisms of chronic inflammation and immunosuppression causing persistent inflammation, immunosuppression and catabolism syndrome (PICS) and describes potential therapies and gaps in current research. RECENT FINDINGS Low HDL cholesterol is predictive of acute sepsis severity and outcome. Recent research has indicated apolipoprotein is a prognostic indicator of long-term outcomes. The pathobiologic mechanisms of PICS have been elucidated in the past several years. Recent research of the interaction of HDL pathways in related chronic inflammatory diseases may provide insights into further mechanisms and therapeutic targets. SUMMARY HDL significantly influences innate and adaptive immune pathways relating to chronic disease and inflammation. Further research is needed to better characterize these interactions in the setting of PICS.
Collapse
Affiliation(s)
- Grant Barker
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Julia R Winer
- University of Florida College of Medicine, Gainesville, Florida
| | - Faheem W Guirgis
- Department of Emergency Medicine, University of Florida College of Medicine, Jacksonville
| | - Srinivasa Reddy
- Department of Molecular and Medical Pharmacology, University of California, Los Angeles, California, USA
| |
Collapse
|
47
|
Fan Y, Liu J, Miao J, Zhang X, Yan Y, Bai L, Chang J, Wang Y, Wang L, Bian Y, Zhou H. Anti-inflammatory activity of the Tongmai Yangxin pill in the treatment of coronary heart disease is associated with estrogen receptor and NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2021; 276:114106. [PMID: 33838287 DOI: 10.1016/j.jep.2021.114106] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Tongmai Yangxin Pill (TMYX) is a patented traditional Chinese medicine originating from two classic prescriptions, Zhigancao Decoction and Shenmai Yin, which composed of 11 Chinese medicinal herbs: Rehmannia glutinosa (Gaertn.) DC., Spatholobus suberectus Dunn, Ophiopogon japonicus (Thunb.) Ker Gawl., Glycyrrhiza uralensis Fisch., Polygonum multiflorum Thunb., Equus asinus L., Schisandra chinensis (Turcz.) Baill., Codonopsis pilosula (Franch.) Nannf., Chinemys reevesii (Gray), Ziziphus jujuba Mill. and Cinnamomum cassia (L.) J.Presl (Committee of the Pharmacopoeia of PR China, 2015). TMYX has marketed in China for the treatment of chest pain, palpitation, angina, irregular heartbeat and coronary heart disease (CHD) for several decades. Previous studies have confirmed that TMYX can treat CHD by reducing inflammation, but the underlying pharmacological mechanism remains unclear. AIM OF THE STUDY This study aimed to declare the underlying pharmacological mechanism of anti-inflammatory activity of TMYX in the treatment of CHD via clinical trial, microarray study, bioinformatics analysis and the vitro assays. MATERIALS AND METHODS Eight CHD patients' serum biochemical indices including coagulation function, lipid metabolism, endothelial injury, metalloprotease, adhesion molecule, inflammatory mediator and homocysteine were measured to investigate the reduction of CHD risk by TMYX oral administration (40 pills/time, 2 times/day) for eight weeks. The expression profile chips and Ingenuity Pathway Analysis (IPA) were assessed to reveal the global transcriptional response and predict related functions, diseases and canonical pathways. The in vitro anti-inflammatory actions of TMYX were evaluated using oxidized low-density lipoprotein (100 μg/mL) induced murine RAW264.7 macrophage with an ethanol extract from TMYX (EETMYX) (25-100 μg/mL). RESULTS TMYX treatment showed reduced levels of apolipoprotein B, endothelin 1, nuclear factor κB (NF-κB) and homocysteine in CHD patients. In contrast, the treatment increased the ratio of apolipoprotein A/apolipoprotein B. EETMYX restored cell morphology and suppressed the lipid deposition of the induced foam cells. EETMYX exerted anti-inflammatory effects by raising the mRNA and protein expression of Estrogen receptor 1 (ESR1), blocking the reduction of IκBa level and the phosphorylation of IKKα/β, IκBα and NF-κB p65, accompanied by inhibiting MCP-1, TNF-α and IL-6 production, which were consistent with bioinformatics predictions. CONCLUSION TMYX treatment improved the biochemical indices in CHD patients. EETMYX effectively attenuated macrophage foam cell formation and exhibited anti-inflammatory activity is associated with regulating ESR1 and NF-κB signaling pathway activity.
Collapse
Affiliation(s)
- Yadong Fan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jianwei Liu
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jing Miao
- Tianjin Second People's Hospital, Tianjin, 300192, China
| | - Xiaoyu Zhang
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Yiqi Yan
- Research Institute of Traditional Chinese Medicine, Tianjin University of Chinese Medicine, Tianjin, 301617, China
| | - Liding Bai
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Jun Chang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Ying Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Li Wang
- Tianjin Second People's Hospital, Tianjin, 300192, China
| | - Yuhong Bian
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| | - Huifang Zhou
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
48
|
Circulating Biomarkers Reflecting Destabilization Mechanisms of Coronary Artery Plaques: Are We Looking for the Impossible? Biomolecules 2021; 11:biom11060881. [PMID: 34198543 PMCID: PMC8231770 DOI: 10.3390/biom11060881] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Revised: 06/11/2021] [Accepted: 06/12/2021] [Indexed: 12/12/2022] Open
Abstract
Despite significant strides to mitigate the complications of acute coronary syndrome (ACS), this clinical entity still represents a major global health burden. It has so far been well-established that most of the plaques leading to ACS are not a result of gradual narrowing of the vessel lumen, but rather a result of sudden disruption of vulnerable atherosclerotic plaques. As most of the developed imaging modalities for vulnerable plaque detection are invasive, multiple biomarkers were proposed to identify their presence. Owing to the pivotal role of lipids and inflammation in the pathophysiology of atherosclerosis, most of the biomarkers originated from one of those processes, whereas recent advancements in molecular sciences shed light on the use of microRNAs. Yet, at present there are no clinically implemented biomarkers or any other method for that matter that could non-invasively, yet reliably, diagnose the vulnerable plaque. Hence, in this review we summarized the available knowledge regarding the pathophysiology of plaque instability, the current evidence on potential biomarkers associated with plaque destabilization and finally, we discussed if search for biomarkers could one day bring us to non-invasive, cost-effective, yet valid way of diagnosing the vulnerable, rupture-prone coronary artery plaques.
Collapse
|
49
|
Piao L, Li Y, Narisawa M, Shen X, Cheng XW. Role of Dipeptidyl Peptidase-4 in Atherosclerotic Cardiovascular Disease in Humans and Animals with Chronic Stress. Int Heart J 2021; 62:470-478. [PMID: 33994495 DOI: 10.1536/ihj.20-181] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Exposure to psychosocial stress is a risk factor for cardiovascular disease, including vascular atherosclerosis-based cardiovascular disease (ACVD). Dipeptidyl peptidase-4 (DPP-4) is a complex enzyme that acts as a membrane-anchored cell surface exopeptidase. DPP-4 is upregulated in metabolic and inflammatory cardiovascular disorders. DPP-4 exhibits many physiological and pharmacological functions by regulating its extremely abundant substrates, such as glucagon-like peptide-1 (GLP-1). Over the last 10 years, emerging data have demonstrated unexpected roles of DPP-4 in extracellular and intracellular signaling, immune activation, inflammation, oxidative stress production, cell apoptosis, insulin resistance, and lipid metabolism. This mini-review focuses on recent novel findings in this field, highlighting a DPP-4-mediated regulation of GLP-1-dependent and -independent signaling pathways as a potential therapeutic molecular target in treatments of chronic psychological stress-related ACVD in humans and animals.
Collapse
Affiliation(s)
- Limei Piao
- Department of Cardiology, Yanbian University Hospital
| | - Yanglong Li
- Department of Cardiology, Yanbian University Hospital
| | - Megumi Narisawa
- Department of Cardiology, Nagoya University Graduate School of Medicine
| | - Xionghu Shen
- Department of Oncology, Yanbian University Hospital
| | - Xian Wu Cheng
- Department of Cardiology, Yanbian University Hospital
| |
Collapse
|
50
|
Bonacina F, Pirillo A, Catapano AL, Norata GD. HDL in Immune-Inflammatory Responses: Implications beyond Cardiovascular Diseases. Cells 2021; 10:cells10051061. [PMID: 33947039 PMCID: PMC8146776 DOI: 10.3390/cells10051061] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/27/2021] [Accepted: 04/27/2021] [Indexed: 12/15/2022] Open
Abstract
High density lipoproteins (HDL) are heterogeneous particles composed by a vast array of proteins and lipids, mostly recognized for their cardiovascular (CV) protective effects. However, evidences from basic to clinical research have contributed to depict a role of HDL in the modulation of immune-inflammatory response thus paving the road to investigate their involvement in other diseases beyond those related to the CV system. HDL-C levels and HDL composition are indeed altered in patients with autoimmune diseases and usually associated to disease severity. At molecular levels, HDL have been shown to modulate the anti-inflammatory potential of endothelial cells and, by controlling the amount of cellular cholesterol, to interfere with the signaling through plasma membrane lipid rafts in immune cells. These findings, coupled to observations acquired from subjects carrying mutations in genes related to HDL system, have helped to elucidate the contribution of HDL beyond cholesterol efflux thus posing HDL-based therapies as a compelling interventional approach to limit the inflammatory burden of immune-inflammatory diseases.
Collapse
Affiliation(s)
- Fabrizia Bonacina
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
| | - Angela Pirillo
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, 20092 Milan, Italy;
- IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
| | - Alberico L. Catapano
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
- IRCCS MultiMedica, Sesto S. Giovanni, 20099 Milan, Italy
- Correspondence: (A.L.C.); (G.D.N.)
| | - Giuseppe D. Norata
- Department of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, 20133 Milan, Italy;
- Center for the Study of Atherosclerosis, E. Bassini Hospital, Cinisello Balsamo, 20092 Milan, Italy;
- Correspondence: (A.L.C.); (G.D.N.)
| |
Collapse
|