1
|
Pi H, Wang G, Wang Y, Zhang M, He Q, Zheng X, Yin K, Zhao G, Jiang T. Immunological perspectives on atherosclerotic plaque formation and progression. Front Immunol 2024; 15:1437821. [PMID: 39399488 PMCID: PMC11466832 DOI: 10.3389/fimmu.2024.1437821] [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: 05/24/2024] [Accepted: 09/09/2024] [Indexed: 10/15/2024] Open
Abstract
Atherosclerosis serves as the primary catalyst for numerous cardiovascular diseases. Growing evidence suggests that the immune response is involved in every stage of atherosclerotic plaque evolution. Rapid, but not specific, innate immune arms, including neutrophils, monocytes/macrophages, dendritic cells (DCs) and other innate immune cells, as well as pattern-recognition receptors and various inflammatory mediators, contribute to atherogenesis. The specific adaptive immune response, governed by T cells and B cells, antibodies, and immunomodulatory cytokines potently regulates disease activity and progression. In the inflammatory microenvironment, the heterogeneity of leukocyte subpopulations plays a very important regulatory role in plaque evolution. With advances in experimental techniques, the fine mechanisms of immune system involvement in atherosclerotic plaque evolution are becoming known. In this review, we examine the critical immune responses involved in atherosclerotic plaque evolution, in particular, looking at atherosclerosis from the perspective of evolutionary immunobiology. A comprehensive understanding of the interplay between plaque evolution and plaque immunity provides clues for strategically combating atherosclerosis.
Collapse
Affiliation(s)
- Hui Pi
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Guangliang Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Yu Wang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ming Zhang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Qin He
- Department of Microbiology and Immunology, Dali University, Dali, Yunnan, China
| | - Xilong Zheng
- Departments of Biochemistry and Molecular Biology and Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kai Yin
- Department of General Practice, The Fifth Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, China
| | - Guojun Zhao
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| | - Ting Jiang
- Affiliated Qingyuan Hospital, Guangzhou Medical University (Qingyuan People’s Hospital), Qingyuan, Guangdong, China
| |
Collapse
|
2
|
Straeten FA, Strecker JK, Börsch AL, Maus B, Hoppen M, Schmeddes B, Härtel L, Fleck AK, van Zyl S, Straeten T, Beuker C, Koecke M, Mueller-Miny L, Faber C, Meyer zu Hörste G, Klotz L, Minnerup J, Schmidt-Pogoda A. A dietary intervention with conjugated linoleic acid enhances microstructural white matter reorganization in experimental stroke. Front Neurol 2024; 15:1341958. [PMID: 39372701 PMCID: PMC11449868 DOI: 10.3389/fneur.2024.1341958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 08/19/2024] [Indexed: 10/08/2024] Open
Abstract
Background A dietary supplementation with conjugated linoleic acid (CLA) was shown to attenuate inflammation and increase the proportions of circulating regulatory T cells (Tregs) and M2-type macrophages in disease models such as autoimmune encephalitis and arteriosclerosis. Since Tregs and anti-inflammatory (M2-type) macrophages were found to enhance stroke recovery, we hypothesized that CLA-supplementation might improve stroke recovery via immune modulatory effects. Methods Functional assessment was performed over 90 days after induction of experimental photothrombotic stroke in wild type mice (n = 37, sham n = 10). Subsequently, immunological characterization of different immunological compartments (n = 16), ex vivo magnetic resonance (MR, n = 12) imaging and immunohistochemical staining (n = 8) was performed. Additionally, we tested the effect of CLA in vitro on peripheral blood mononuclear cells from human stroke patients and healthy controls (n = 12). Results MR diffusion tensor imaging (DTI) demonstrated enhanced microstructural reorganization of interhemispheric white matter tracts, dependent on lesion size. Functional recovery over 90 days remained unaffected. Detailed immunological analyses across various compartments revealed no significant long-term immunological alterations due to CLA. However, analyses of human blood samples post-stroke showed reduced levels of pro-inflammatory interferon-γ (IFN-γ) and tumor necrosis factor alpha (TNF-α) release by T-lymphocytes following in vitro treatment with CLA. Conclusion We aimed to explore the efficacy of a dietary intervention with minimal known side effects that could be accessible to human stroke patients, regardless of the degree of disability, and without the risks associated with aggressive immunomodulatory therapies. Our main findings include improved microstructural reorganization in small infarcts and a reduced inflammatory response of human T cells in vitro.
Collapse
Affiliation(s)
- Frederike A. Straeten
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Jan-Kolja Strecker
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Anna-Lena Börsch
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Bastian Maus
- Translational Research Imaging Center, University of Münster, Münster, Germany
- Clinic of Radiology, University Hospital Münster, University of Münster, Münster, Germany
| | - Maike Hoppen
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Birgit Schmeddes
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Lucia Härtel
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Ann-Katrin Fleck
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Stephanie van Zyl
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Tabea Straeten
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Carolin Beuker
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Mailin Koecke
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Louisa Mueller-Miny
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Cornelius Faber
- Translational Research Imaging Center, University of Münster, Münster, Germany
- Clinic of Radiology, University Hospital Münster, University of Münster, Münster, Germany
| | - Gerd Meyer zu Hörste
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Luisa Klotz
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Jens Minnerup
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| | - Antje Schmidt-Pogoda
- Department of Neurology, University Hospital Münster, University of Münster, Münster, Germany
| |
Collapse
|
3
|
Xu T, Chen G, Li J, Zhang Y. Exploring causal correlations between inflammatory cytokines and intervertebral disc degeneration: A Mendelian randomization. JOR Spine 2024; 7:e1349. [PMID: 38993524 PMCID: PMC11237178 DOI: 10.1002/jsp2.1349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 07/13/2024] Open
Abstract
Background Inflammatory cytokines have been reported to be related to intervertebral disc degeneration (IVDD) in several previous studies. However, it remains unclear about the causal relationship between inflammatory cytokines and IVDD. This study employs Mendelian randomization (MR) to analyze the causal link between inflammatory cytokines and the risk of IVDD. Method We used genetic variants associated with inflammatory cytokines from a meta-analysis of genome-wide association study (GWAS) in 8293 Finns as instrumental variables and IVDD data were sourced from the FinnGen consortium. The main analytical approach utilized Inverse-Variance Weighting (IVW) with random effects to assess the causal relationship. Additionally, complementary methods such as MR-Egger, weighted median, simple mode, weighted mode, and MR pleiotropy residual sum and outlier were employed to enhance the robustness of the final results. Result We found interferon-gamma (IFN-γ, p = 2.14 × 10-6, OR = 0.870, 95% CI = 0.821-0.921), interleukin-1 beta (IL-1b, p = 0.012, OR = 0.951, 95% CI = 0.914-0.989), interleukin-4 (IL-4, p = 0.034, OR = 0.946, 95% CI = 0.899-0.996), interleukin-18 (IL-18, p = 0.028, OR = 0.964, 95% CI = 0.934-0.996), granulocyte colony-stimulating factor (GCSF, p = 0.010, OR = 0.919, 95% CI = 0.861-0.980), and Stromal cell-derived factor 1a (SDF1a, p = 0.014, OR = 1.072, 95% CI = 1.014-1.134) were causally associated with risk of IVDD. Conclusion Our MR analyses found a potential causal relationship between six inflammation cytokines (IFN-γ, IL-1b, IL-4, IL-18, SDF1a, and GCSF) and altered IVDD risk.
Collapse
Affiliation(s)
- Tao Xu
- Department of Orthopedics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Guangzi Chen
- Department of Orthopedics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| | - Jian Li
- Department of OrthopaedicsThird Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalTaiyuanPeople's Republic of China
| | - Yingchi Zhang
- Department of Orthopedics, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Department of Traumatology, Tongji Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
| |
Collapse
|
4
|
La Chica Lhoëst MT, Martinez A, Claudi L, Garcia E, Benitez-Amaro A, Polishchuk A, Piñero J, Vilades D, Guerra JM, Sanz F, Rotllan N, Escolà-Gil JC, Llorente-Cortés V. Mechanisms modulating foam cell formation in the arterial intima: exploring new therapeutic opportunities in atherosclerosis. Front Cardiovasc Med 2024; 11:1381520. [PMID: 38952543 PMCID: PMC11215187 DOI: 10.3389/fcvm.2024.1381520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 05/28/2024] [Indexed: 07/03/2024] Open
Abstract
In recent years, the role of macrophages as the primary cell type contributing to foam cell formation and atheroma plaque development has been widely acknowledged. However, it has been long recognized that diffuse intimal thickening (DIM), which precedes the formation of early fatty streaks in humans, primarily consists of lipid-loaded smooth muscle cells (SMCs) and their secreted proteoglycans. Recent studies have further supported the notion that SMCs constitute the majority of foam cells in advanced atherosclerotic plaques. Given that SMCs are a major component of the vascular wall, they serve as a significant source of microvesicles and exosomes, which have the potential to regulate the physiology of other vascular cells. Notably, more than half of the foam cells present in atherosclerotic lesions are of SMC origin. In this review, we describe several mechanisms underlying the formation of intimal foam-like cells in atherosclerotic plaques. Based on these mechanisms, we discuss novel therapeutic approaches that have been developed to regulate the generation of intimal foam-like cells. These innovative strategies hold promise for improving the management of atherosclerosis in the near future.
Collapse
Affiliation(s)
- M. T. La Chica Lhoëst
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - A. Martinez
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - L. Claudi
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - E. Garcia
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - A. Benitez-Amaro
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - A. Polishchuk
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
| | - J. Piñero
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - D. Vilades
- Department of Cardiology, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB-SANTPAU), Universitat Autonoma de Barcelona, Barcelona, Spain
- Department of Cardiovascular, CIBERCV, Institute of Health Carlos III, Madrid, Spain
| | - J. M. Guerra
- Department of Cardiology, Hospital de la Santa Creu I Sant Pau, Biomedical Research Institute Sant Pau (IIB-SANTPAU), Universitat Autonoma de Barcelona, Barcelona, Spain
- Department of Cardiovascular, CIBERCV, Institute of Health Carlos III, Madrid, Spain
| | - F. Sanz
- Research Programme on Biomedical Informatics (GRIB), Department of Experimental and Health Sciences (DCEXS), Hospital del Mar Medical Research Institute (IMIM), Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - N. Rotllan
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Department of Cardiovascular, CIBERDEM, Institute of Health Carlos III, Madrid, Spain
| | - J. C. Escolà-Gil
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Department of Cardiovascular, CIBERDEM, Institute of Health Carlos III, Madrid, Spain
| | - V. Llorente-Cortés
- Department of Experimental Pathology, Institute of Biomedical Research of Barcelona (IIBB)-Spanish National Research Council (CSIC), Barcelona, Spain
- Department of Cardiovascular, Institut de Recerca Sant Pau (IR SANT PAU), Barcelona, Spain
- Department of Cardiovascular, CIBERCV, Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
5
|
Xia J, Chen J, Xing X, Meng J, Song X, Lou D. Dendrobine regulates STAT3 to attenuate mitochondrial dysfunction and senescence in vascular endothelial cells triggered by oxidized low-density lipoprotein. Drug Dev Res 2024; 85:e22152. [PMID: 38349255 DOI: 10.1002/ddr.22152] [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: 07/11/2023] [Revised: 01/05/2024] [Accepted: 01/10/2024] [Indexed: 02/15/2024]
Abstract
Our previous studies have highlighted the potential therapeutic efficacy of dendrobine, an alkaloid, in atherosclerosis (AS), nevertheless, the underlying mechanism remains unclear. This study employs a combination of network pharmacology and in vitro experiments to explore the regulatory pathways involved. Through network pharmacology, the biological function for intersection targets between dendrobine and AS were identified. Molecular docking was conducted to investigate the interaction between the dominant target and dendrobine. Human umbilical vein endothelial cells (HUVECs) were treated with oxidized low-density lipoprotein (ox-LDL) to mimic AS, and the effects of dendrobine on cell viability, lipid deposition, mitochondrial function, and cellular senescence were evaluated. Subsequently, cells were treated with the mitophagy inhibitor Mdivi-1 and the STAT3 agonist colivelin to assess the role of mitophagy and STAT3 signaling in dendrobine regulation. Intersection targets were associated with biological processes, including reactive oxygen species production. Dendrobine attenuated the effects of ox-LDL treatment on HUVECs, mitigating changes in cell activity, lipid deposition, mitochondrial function, and cellular senescence. Both Mdivi-1 and colivelin treatments resulted in decreased cell viability and increased cellular senescence, with colivelin suppressing mitophagy. Cotreatment with Mdivi-1 and colivelin further aggravated cellular senescence and inhibited FoxO signaling. Together, this study indicated that dendrobine regulated the STAT3/FoxO signaling pathway, alleviating mitochondrial dysfunction and cellular senescence. This study contributes valuable insights to the potential clinical application of dendrobine.
Collapse
Affiliation(s)
- Jia Xia
- Department of Rheumatology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jingyi Chen
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinyue Xing
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jing Meng
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaoying Song
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Danfei Lou
- Department of Emergency, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
6
|
Jing J, Guo J, Dai R, Zhu C, Zhang Z. Targeting gut microbiota and immune crosstalk: potential mechanisms of natural products in the treatment of atherosclerosis. Front Pharmacol 2023; 14:1252907. [PMID: 37719851 PMCID: PMC10504665 DOI: 10.3389/fphar.2023.1252907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 08/21/2023] [Indexed: 09/19/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory reaction that primarily affects large and medium-sized arteries. It is a major cause of cardiovascular disease and peripheral arterial occlusive disease. The pathogenesis of AS involves specific structural and functional alterations in various populations of vascular cells at different stages of the disease. The immune response is involved throughout the entire developmental stage of AS, and targeting immune cells presents a promising avenue for its treatment. Over the past 2 decades, studies have shown that gut microbiota (GM) and its metabolites, such as trimethylamine-N-oxide, have a significant impact on the progression of AS. Interestingly, it has also been reported that there are complex mechanisms of action between GM and their metabolites, immune responses, and natural products that can have an impact on AS. GM and its metabolites regulate the functional expression of immune cells and have potential impacts on AS. Natural products have a wide range of health properties, and researchers are increasingly focusing on their role in AS. Now, there is compelling evidence that natural products provide an alternative approach to improving immune function in the AS microenvironment by modulating the GM. Natural product metabolites such as resveratrol, berberine, curcumin, and quercetin may improve the intestinal microenvironment by modulating the relative abundance of GM, which in turn influences the accumulation of GM metabolites. Natural products can delay the progression of AS by regulating the metabolism of GM, inhibiting the migration of monocytes and macrophages, promoting the polarization of the M2 phenotype of macrophages, down-regulating the level of inflammatory factors, regulating the balance of Treg/Th17, and inhibiting the formation of foam cells. Based on the above, we describe recent advances in the use of natural products that target GM and immune cells crosstalk to treat AS, which may bring some insights to guide the treatment of AS.
Collapse
Affiliation(s)
- Jinpeng Jing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Jing Guo
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Dai
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Chaojun Zhu
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- Institute of TCM Ulcers, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Surgical Department of Traditional Chinese Medicine, Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Ge P, Li H, Ya X, Xu Y, Ma L, He Q, Wang R, Liu Z, Zhang Q, Zhang Y, Wang W, Zhang D, Zhao J. Single-cell atlas reveals different immune environments between stable and vulnerable atherosclerotic plaques. Front Immunol 2023; 13:1085468. [PMID: 36741406 PMCID: PMC9889979 DOI: 10.3389/fimmu.2022.1085468] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 12/20/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction Regardless of the degree of stenosis, vulnerable plaque is an important cause of ischemic stroke and thrombotic complications. The changes of the immune microenvironment within plaques seem to be an important factor affecting the characteristics of the plaque. However, the differences of immune microenvironment between stable and vulnerable plaques were remained unknown. Methods In this study, RNA-sequencing was performed on superficial temporal arteries from 5 traumatic patients and plaques from 3 atherosclerotic patients to preliminary identify the key immune response processes in plaques. Mass cytometry (CyTOF) technology was used to explore differences in immune composition between 9 vulnerable plaques and 12 stable plaques. Finally, immunofluorescence technique was used to validate our findings in the previous analysis. Results Our results showed that more CD86+CD68+ M1 pro-inflammatory macrophages were found in vulnerable plaques, while CD4+T memory cells were mainly found in stable plaques. In addition, a CD11c+ subset of CD4+T cells with higher IFN-r secretion was found within the vulnerable plaque. In two subsets of B cells, CD19+CD20-B cells in vulnerable plaques secreted more TNF-a and IL-6, while CD19-CD20+B cells expressed more PD-1 molecules. Conclusion In conclusion, our study suggested that M1-like macrophages are the major cell subset affecting plaque stability, while functional B cells may also contribute to plaque stability.
Collapse
Affiliation(s)
- Peicong Ge
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Hao Li
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Xiaolong Ya
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yiqiao Xu
- Capital Medical University, Beijing, China
| | - Long Ma
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Qiheng He
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Rong Wang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Zechen Liu
- Department of Biostatistics, Harvard School of Public Health, Huntington Avenue, Boston, MA, United States
| | - Qian Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yan Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wenjing Wang
- Beijing Institute of Hepatology, Beijing YouAn Hospital, Capital Medical University, Beijing, China,*Correspondence: Wenjing Wang, ; Dong Zhang, ; Jizong Zhao,
| | - Dong Zhang
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,Department of Neurosurgery, Beijing Hospital, Beijing, China,*Correspondence: Wenjing Wang, ; Dong Zhang, ; Jizong Zhao,
| | - Jizong Zhao
- Department of Neurosurgery, Beijing Tiantan Hospital, Beijing, China,China National Clinical Research Center for Neurological Diseases, Beijing, China,*Correspondence: Wenjing Wang, ; Dong Zhang, ; Jizong Zhao,
| |
Collapse
|
8
|
Anti-Inflammatory Potential of Fucoidan for Atherosclerosis: In Silico and In Vitro Studies in THP-1 Cells. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27103197. [PMID: 35630678 PMCID: PMC9146328 DOI: 10.3390/molecules27103197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 01/13/2023]
Abstract
Several diseases, including atherosclerosis, are characterized by inflammation, which is initiated by leukocyte migration to the inflamed lesion. Hence, genes implicated in the early stages of inflammation are potential therapeutic targets to effectively reduce atherogenesis. Algal-derived polysaccharides are one of the most promising sources for pharmaceutical application, although their mechanism of action is still poorly understood. The present study uses a computational method to anticipate the effect of fucoidan and alginate on interactions with adhesion molecules and chemokine, followed by an assessment of the cytotoxicity of the best-predicted bioactive compound for human monocytic THP-1 macrophages by lactate dehydrogenase and crystal violet assay. Moreover, an in vitro pharmacodynamics evaluation was performed. Molecular docking results indicate that fucoidan has a greater affinity for L-and E-selectin, monocyte chemoattractant protein 1 (MCP-1), and intercellular adhesion molecule-1 (ICAM-1) as compared to alginate. Interestingly, there was no fucoidan cytotoxicity on THP-1 macrophages, even at 200 µg/mL for 24 h. The strong interaction between fucoidan and L-selectin in silico explained its ability to inhibit the THP-1 monocytes migration in vitro. MCP-1 and ICAM-1 expression levels in THP-1 macrophages treated with 50 µg/mL fucoidan for 24 h, followed by induction by IFN-γ, were shown to be significantly suppressed as eight- and four-fold changes, respectively, relative to cells treated only with IFN-γ. These results indicate that the electrostatic interaction of fucoidan improves its binding affinity to inflammatory markers in silico and reduces their expression in THP-1 cells in vitro, thus making fucoidan a good candidate to prevent inflammation.
Collapse
|
9
|
Liu L, Shi Z, Ji X, Zhang W, Luan J, Zahr T, Qiang L. Adipokines, adiposity, and atherosclerosis. Cell Mol Life Sci 2022; 79:272. [PMID: 35503385 PMCID: PMC11073100 DOI: 10.1007/s00018-022-04286-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/11/2022] [Accepted: 04/03/2022] [Indexed: 12/12/2022]
Abstract
Characterized by a surplus of whole-body adiposity, obesity is strongly associated with the prognosis of atherosclerosis, a hallmark of coronary artery disease (CAD) and the major contributor to cardiovascular disease (CVD) mortality. Adipose tissue serves a primary role as a lipid-storage organ, secreting cytokines known as adipokines that affect whole-body metabolism, inflammation, and endocrine functions. Emerging evidence suggests that adipokines can play important roles in atherosclerosis development, progression, as well as regression. Here, we review the versatile functions of various adipokines in atherosclerosis and divide these respective functions into three major groups: protective, deteriorative, and undefined. The protective adipokines represented here are adiponectin, fibroblast growth factor 21 (FGF-21), C1q tumor necrosis factor-related protein 9 (CTRP9), and progranulin, while the deteriorative adipokines listed include leptin, chemerin, resistin, Interleukin- 6 (IL-6), and more, with additional adipokines that have unclear roles denoted as undefined adipokines. Comprehensively categorizing adipokines in the context of atherosclerosis can help elucidate the various pathways involved and potentially pave novel therapeutic approaches to treat CVDs.
Collapse
Affiliation(s)
- Longhua Liu
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China.
| | - Zunhan Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Xiaohui Ji
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Wenqian Zhang
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Jinwen Luan
- School of Kinesiology, Shanghai University of Sport, Shanghai, People's Republic of China
| | - Tarik Zahr
- Department of Pharmacology, Columbia University, New York, NY, USA
| | - Li Qiang
- Department of Pathology and Cellular Biology and Naomi Berrie Diabetes Center, Columbia University, New York, NY, USA.
| |
Collapse
|
10
|
Moschetti L, Piantoni S, Vizzardi E, Sciatti E, Riccardi M, Franceschini F, Cavazzana I. Endothelial Dysfunction in Systemic Lupus Erythematosus and Systemic Sclerosis: A Common Trigger for Different Microvascular Diseases. Front Med (Lausanne) 2022; 9:849086. [PMID: 35462989 PMCID: PMC9023861 DOI: 10.3389/fmed.2022.849086] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
This review describes the complex interplay between inflammation, vasculopathy and fibrosis that involve the heart and peripheral small vessels, leading to endothelial stiffness, vascular damage, and early aging in patients with systemic lupus erythematosus and systemic sclerosis, which represents two different models of vascular dysfunction among systemic autoimmune diseases. In fact, despite the fact that diagnostic methods and therapies have been significantly improved in the last years, affected patients show an excess of cardiovascular mortality if compared with the general population. In addition, we provide a complete overview on the new techniques which are used for the evaluation of endothelial dysfunction in a preclinical phase, which could represent a new approach in the assessment of cardiovascular risk in these patients.
Collapse
Affiliation(s)
- Liala Moschetti
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Silvia Piantoni
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
- *Correspondence: Silvia Piantoni,
| | - Enrico Vizzardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | | | - Mauro Riccardi
- Cardiology Unit, ASST Spedali Civili of Brescia, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Franco Franceschini
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Ilaria Cavazzana
- Rheumatology and Clinical immunology Unit, ASST Spedali Civili of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
11
|
Abstract
Statins are 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors used worldwide to manage dyslipidaemia and thus limit the development of atherosclerotic disease and its complications. These atheroprotective drugs are now known to exert pleiotropic actions outside of their cholesterol-lowering activity, including altering immune cell function. Macrophages are phagocytic leukocytes that play critical functional roles in the pathogenesis of atherosclerosis and are directly targeted by statins. Early studies documented the anti-inflammatory effects of statins on macrophages, but emerging evidence suggests that these drugs can also enhance pro-inflammatory macrophage responses, creating an unresolved paradox. This review comprehensively examines the in vitro, in vivo, and clinical literature to document the statin-induced changes in macrophage polarization and immunomodulatory functions, explore the underlying mechanisms involved, and offer potential explanations for this paradox. A better understanding of the immunomodulatory actions of statins on macrophages should pave the way for the development of novel therapeutic approaches to manage atherosclerosis and other chronic diseases and conditions characterised by unresolved inflammation.
Collapse
|
12
|
Huwait EA, Saddeek SY, Al-Massabi RF, Almowallad SJ, Pushparaj PN, Kalamegam G. Antiatherogenic Effects of Quercetin in the THP-1 Macrophage Model In Vitro, With Insights Into Its Signaling Mechanisms Using In Silico Analysis. Front Pharmacol 2021; 12:698138. [PMID: 34385920 PMCID: PMC8353397 DOI: 10.3389/fphar.2021.698138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/25/2021] [Indexed: 01/08/2023] Open
Abstract
Background: Atherosclerosis (AS), a major risk factor for stroke and brain tissue destruction, is an inflammatory disease of the blood vessels, and the underlying pathology is inflammation mediated by various chemokines and cytokines. Quercetin, a natural flavonol, is reported to have both anti-inflammatory and antioxidant properties. As such, in the present study, we evaluated the antiatherogenic effects of quercetin in a human THP-1 cell line in vitro and also the signaling mechanisms using in silico analysis. Materials and Methods: THP-1 macrophages exposed to different concentrations of quercetin (5–100 μM for 24 h) were tested for cytotoxicity. Real-time gene expression assay for intercellular adhesion molecule-1 (ICAM-1) and monocyte chemoattractant protein-1 (MCP-1) was carried out following treatment with quercetin at 15 and 30 μM for 24 h either in the absence or presence of interferon (IFN-γ) for 3 h to induce inflammation. Monocyte migration and cholesterol efflux were also assessed. Results: Quercetin did not exert any cytotoxic effects on THP-1 cells at the various concentrations tested. The gene expression assay showed a significant decrease in ICAM-1 (by 3.05 and 2.70) and MCP-1 (by 22.71 and 27.03), respectively. Quercetin at 15 µM decreased THP-1 monocyte migration by 33% compared to the MCP-1-treated cells. It also increased cholesterol efflux significantly by1.64-fold and 1.60-fold either alone or in combination with IFN-γ, respectively. Ingenuity Pathway Analysis of the molecular interactions of quercetin identified canonical pathways directly related to lipid uptake and cholesterol efflux. Furthermore, CD36, SR-A, and LXR-α also demonstrated significant increases by 72.16-, 149.10-, and 29.68-fold, respectively. Conclusion: Our results from both in vitro and in silico studies identified that quercetin inhibited the THP-1 monocyte migration, MCP-1, and ICAM-1 and increased cholesterol efflux probably mediated via the LXR/RXR signaling pathway. Therefore, quercetin will help prevent cell infiltration in atherosclerotic plaques and reduce the risk of stroke or brain destruction.
Collapse
Affiliation(s)
- Etimad A Huwait
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Cell Culture Unit and Experimental Biochemistry Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Salma Y Saddeek
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Cell Culture Unit and Experimental Biochemistry Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.,Chemistry Department, Faculty of Sciences, University of Hafr Al Batin, Hafr Al Batin, Saudi Arabia
| | - Rehab F Al-Massabi
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Cell Culture Unit and Experimental Biochemistry Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.,Biochemistry Department, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Sanaa J Almowallad
- Biochemistry Department, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Cell Culture Unit and Experimental Biochemistry Unit, King Fahad Medical Research Centre, King Abdulaziz University, Jeddah, Saudi Arabia.,Biochemistry Department, Faculty of Sciences, University of Tabuk, Tabuk, Saudi Arabia
| | - Peter Natesan Pushparaj
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Gauthaman Kalamegam
- Center of Excellence in Genomic Medicine Research, King Abdulaziz University, Jeddah, Saudi Arabia.,Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia.,Pharmaceutical Division, Nibblen Life Sciences Private Limited, Chennai, India
| |
Collapse
|
13
|
Cai D, Liu H, Wang J, Hou Y, Pang T, Lin H, He C. Balasubramide derivative 3C attenuates atherosclerosis in apolipoprotein E-deficient mice: role of AMPK-STAT1-STING signaling pathway. Aging (Albany NY) 2021; 13:12160-12178. [PMID: 33901014 PMCID: PMC8109080 DOI: 10.18632/aging.202929] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Accepted: 11/30/2020] [Indexed: 12/12/2022]
Abstract
We previously reported the neuroprotective effects of (+)-balasubramide derived compound 3C, but its action on atherosclerosis in vivo remains unknown. The study was designed to investigate the potential effects of 3C on atherogenesis and explore the possible underlying mechanisms. 3C ameliorated high-fat diet-induced body weight gain, hyperlipidemia, and atherosclerotic plaque burden in apolipoprotein E-deficient (ApoE-/-) mice after 10 weeks of treatment. 3C suppressed the expression of genes involved in triglyceride synthesis in liver. 3C prevented aortic inflammation as evidenced by reduction of adhesive molecule levels and macrophage infiltration. Mechanistic studies revealed that activation of AMP-activated protein kinase (AMPK) is central to the athero-protective effects of 3C. Increased AMPK activity by 3C resulted in suppressing interferon-γ (IFN-γ) induced activation of signal transducer and activator of transcription-1 (STAT1) and stimulator of interferon genes (STING) signaling pathways and downstream pro-inflammatory markers. Moreover, 3C inhibited ox-LDL triggered lipid accumulation and IFN-γ induced phenotypic switch toward M1 macrophage in RAW 264.7 cells. Our present data suggest that 3C prevents atherosclerosis via pleiotropic effects, including amelioration of lipid profiles, vascular inflammation and macrophage pro-inflammatory phenotype. 3C has the potential to be developed as a promising drug for atherosclerosis and related cardiovascular disease.
Collapse
Affiliation(s)
- Dongcheng Cai
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Hongxia Liu
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Jing Wang
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Yuanlong Hou
- Jiangsu Province Key Laboratory of Drug Metabolism, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| | - Tao Pang
- Jiangsu Key Laboratory of Drug Screening, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
| | - Hansen Lin
- College of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chaoyong He
- State Key Laboratory of Natural Medicines, Department of Pharmacology, China Pharmaceutical University, Nanjing 210009, Jiangsu, China
| |
Collapse
|
14
|
Falasca K, Lanuti P, Ucciferri C, Pieragostino D, Cufaro MC, Bologna G, Federici L, Miscia S, Pontolillo M, Auricchio A, Del Boccio P, Marchisio M, Vecchiet J. Circulating extracellular vesicles as new inflammation marker in HIV infection. AIDS 2021; 35:595-604. [PMID: 33306552 DOI: 10.1097/qad.0000000000002794] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Extracellular vesicles, released by cell pullulation, are surrounded by a phospholipid bilayer and carry proteins as well and genetic material. It has been shown that extracellular vesicles mediate intercellular communication in several conditions, such as inflammation, immunodeficiency, tumor growth, and viral infections. Here, we analyzed circulating levels of extracellular vesicles in order to clarify their role in chronic inflammation mechanisms characterizing HIV patients. METHODS We analyzed and subtyped circulating levels of extracellular vesicles, through a recently developed flow cytometry method. In detail, endothelial-derived extracellular vesicles (CD31+/CD41a-/CD45-, EMVs), extracellular vesicles stemming from leukocytes (CD45+, LMVs) and platelets (CD41a+/CD31+) were identified and enumerated. Moreover, we analyzed the extracellular vesicle protein cargo with proteomic analysis. RESULTS Circulating levels of total extracellular vesicles, EMVs and LMVs were significantly lower in the HIV+ patients than in healthy subjects, whereas platelet-derived extracellular vesicles resulted higher in patients than in the healthy population. Proteomic analysis showed the upregulation of gammaIFN and IL1α, and down-regulation of OSM, NF-kB, LIF, and RXRA signaling resulted activated in this patients. CONCLUSION These data demonstrate, for the first time that HIV infection induces the production of extracellular vesicles containing mediators that possibly feed the chronic inflammation and the viral replication. These two effects are connected as the inflammation itself induces the viral replication. We, therefore, hypothesize that HIV infection inhibits the production of extracellular vesicles that carry anti-inflammatory molecules.
Collapse
Affiliation(s)
- Katia Falasca
- Clinic of Infectious Diseases, Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara, Chieti
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara
- Center for Advanced Studies and Technology (CAST)
| | - Claudio Ucciferri
- Clinic of Infectious Diseases, Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara, Chieti
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST)
- Department of Medical, Oral and Biotechnological Sciences
| | - Maria Concetta Cufaro
- Center for Advanced Studies and Technology (CAST)
- Department of Pharmacy, University 'G. d'Annunzio' of Chieti-Pescara, Chieti, Italy
| | - Giuseppina Bologna
- Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara
- Center for Advanced Studies and Technology (CAST)
| | - Luca Federici
- Center for Advanced Studies and Technology (CAST)
- Department of Medical, Oral and Biotechnological Sciences
| | - Sebastiano Miscia
- Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara
- Center for Advanced Studies and Technology (CAST)
| | - Michela Pontolillo
- Clinic of Infectious Diseases, Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara, Chieti
| | - Antonio Auricchio
- Clinic of Infectious Diseases, Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara, Chieti
| | - Piero Del Boccio
- Center for Advanced Studies and Technology (CAST)
- Department of Pharmacy, University 'G. d'Annunzio' of Chieti-Pescara, Chieti, Italy
| | - Marco Marchisio
- Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara
- Center for Advanced Studies and Technology (CAST)
| | - Jacopo Vecchiet
- Clinic of Infectious Diseases, Department of Medicine and Aging Sciences, University 'G. d'Annunzio' of Chieti-Pescara, Chieti
| |
Collapse
|
15
|
Zhang Y, Wang Z, Shi B, Li Y, Wang R, Sun J, Hu Y, Yuan C, Xu Q. Effect of gingival mesenchymal stem cell-derived exosomes on inflammatory macrophages in a high-lipid microenvironment. Int Immunopharmacol 2021; 94:107455. [PMID: 33582592 DOI: 10.1016/j.intimp.2021.107455] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/27/2021] [Accepted: 01/28/2021] [Indexed: 02/09/2023]
Abstract
OBJECTIVE The aim of this study was to examine the effect of gingival mesenchymal stem cells derived exosomes (GMSC-Exos) on lipopolysaccharide/interferon-gamma (LPS/INF-γ)-induced inflammatory macrophages in a high-lipid microenvironment. MATERIALS AND METHODS Exosomes were obtained by culturing gingival mesenchymal stem cells (GMSCs) in alpha-MEM with exosome-free fetal bovine serum for 48 h. The control group was produced in vitro by inducing human acute monocytic leukemia cells (THP-1 cells) into naïve macrophages (M0). Inflammatory macrophages (M1) were made by activating M0 macrophages with LPS/IFN-γ. These M1 macrophages were treated with oxidized low-density lipoprotein (ox-LDL) to create the high-lipid group, of which some macrophages were further treated with GMSC-Exos for 24 h to form the GMSC-Exos group. Supernatants were collected, and total RNA were extracted for downstream analysis. The expression of surface markers in macrophages were analyzed by flow cytometry. The lipid accumulation level was assessed by oil red O staining. RESULTS Exosomes were successfully isolated from GMSC medium. The GMSC-Exos group showed lower Tumor Necrosis Factor-α (TNF-α), Interleukin-6 (IL-6), Interleukin-1β (IL-1β), and cluster of differentiation 86 (CD86) expression levels than the high-lipid group, and the highest levels of Interleukin-10 (IL-10) among all groups. The GMSC-Exos group showed significant reductions in TNF-α levels than the high-lipid group, and significant escalations in IL-10 levels than the other two groups. Oil red o Staining showed that lipid accumulation in macrophages was inhibited in the GMSC-Exos group. CONCLUSIONS GMSC-Exos reduce the release level and expression of inflammatory factors, inhibit lipid accumulation, and promote the polarization of pro-inflammatory macrophages into anti-inflammatory phenotype in a high-lipid microenvironment.
Collapse
Affiliation(s)
- Yalong Zhang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, Shandong, China; School of Stomatology of Qingdao University, Qingdao 266003, China
| | - Zhiguo Wang
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Bohong Shi
- Department of Stomatology, People's Hospital of Rizhao, 126 Taian Road, Rizhao 276826, Shandong, China
| | - Yan Li
- Department of Stomatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, Shandong, China; School of Stomatology of Qingdao University, Qingdao 266003, China
| | - Ru Wang
- Department of Stomatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, Shandong, China; School of Stomatology of Qingdao University, Qingdao 266003, China
| | - Jiayao Sun
- Department of Stomatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, Shandong, China; School of Stomatology of Qingdao University, Qingdao 266003, China
| | - Yingzhe Hu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, Shandong, China; School of Stomatology of Qingdao University, Qingdao 266003, China
| | - Changqing Yuan
- Department of Stomatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, Shandong, China.
| | - Quanchen Xu
- Department of Stomatology, The Affiliated Hospital of Qingdao University, No. 16 Jiangsu Road, Qingdao 266003, Shandong, China.
| |
Collapse
|
16
|
Ding X, Xiang W, He X. IFN-I Mediates Dysfunction of Endothelial Progenitor Cells in Atherosclerosis of Systemic Lupus Erythematosus. Front Immunol 2020; 11:581385. [PMID: 33262760 PMCID: PMC7686511 DOI: 10.3389/fimmu.2020.581385] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a multi-system autoimmune disease including the cardiovascular system. Atherosclerosis is the most common cardiovascular complication of SLE and a significant risk factor for morbidity and mortality. Vascular damage/protection mechanism in SLE patients is out of balance, caused by the cascade reaction among oxidative stress, proinflammatory cytokines, Neutrophil Extracellular Traps, activation of B cells and autoantibodies and abnormal T cells. As a precursor cell repairing vascular endothelium, endothelial progenitor cells (EPCs) belong to the protective mechanism and show the reduced number and impaired function in SLE. However, the pathological mechanism of EPCs dysfunction in SLE remains ill-defined. This paper reviews the latest SLE epidemiology and pathogenesis, discusses the changes in the number and function of EPCs in SLE, expounds the role of EPCs in SLE atherosclerosis, and provides new guidance and theoretical basis for exploring novel targets for SLE treatment.
Collapse
Affiliation(s)
- Xuewei Ding
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Wei Xiang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Medical University, NHC Key Laboratory of Control of Tropical diseases (Hainan Medical University), Haikou, China
| | - Xiaojie He
- Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
- Laboratory of Pediatric Nephrology, Institute of Pediatrics, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
17
|
Elyasi A, Voloshyna I, Ahmed S, Kasselman LJ, Behbodikhah J, De Leon J, Reiss AB. The role of interferon-γ in cardiovascular disease: an update. Inflamm Res 2020; 69:975-988. [PMID: 32699989 DOI: 10.1007/s00011-020-01382-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022] Open
Abstract
PURPOSE Cardiovascular disease (CVD) is the leading cause of death, globally, and its prevalence is only expected to rise due to the increasing incidence of co-morbidities such as obesity and diabetes. Medical treatment of CVD is directed primarily at slowing or reversing the underlying atherosclerotic process by managing circulating lipids with an emphasis on control of low-density lipoprotein (LDL) cholesterol. However, over the past several decades, there has been increasing recognition that chronic inflammation and immune system activation are important contributors to atherosclerosis. This shift in focus has led to the elucidation of the complex interplay between cholesterol and cellular secretion of cytokines involved in CVD pathogenesis. Of the vast array of cytokine promoting atherosclerosis, interferon (IFN)-γ is highly implicated and, therefore, of great interest. METHODS Literature review was performed to further understand the effect of IFN-γ on the development of atherosclerotic CVD. RESULTS IFN-γ, the sole member of the type II IFN family, is produced by T cells and macrophages, and has been found to induce production of other cytokines and to have multiple effects on all stages of atherogenesis. IFN-γ activates a variety of signaling pathways, most commonly the Janus kinase (JAK)/signal transducer and activator of transcription (STAT) pathway, to induce oxidative stress, promote foam cell accumulation, stimulate smooth muscle cell proliferation and migration into the arterial intima, enhance platelet-derived growth factor expression, and destabilize plaque. These are just a few of the contributions of IFN-γ to the initiation and progression of atherosclerotic CVD. CONCLUSION Given the pivotal role of IFN-γ in the advancement of CVD, activation of its signaling pathways is being explored as a driver of atherosclerosis. Manipulation of this key cytokine may lead to novel therapeutic avenues for CVD prevention and treatment. A number of therapies are being explored with IFN-γ as the potential target.
Collapse
Affiliation(s)
- Ailin Elyasi
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Iryna Voloshyna
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Saba Ahmed
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Lora J Kasselman
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Jennifer Behbodikhah
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Joshua De Leon
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA
| | - Allison B Reiss
- Department of Medicine and Biomedical Research Institute, NYU Long Island School of Medicine, NYU Winthrop Hospital, 101 Mineola Boulevard, Suite 4-004, Mineola, NY, 11501, USA.
| |
Collapse
|
18
|
Cyclodextrin Ameliorates the Progression of Atherosclerosis via Increasing High-Density Lipoprotein Cholesterol Plasma Levels and Anti-inflammatory Effects in Rabbits. J Cardiovasc Pharmacol 2020; 73:334-342. [PMID: 30855405 DOI: 10.1097/fjc.0000000000000660] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
To investigate the therapeutic effects of cyclodextrin on the development of atherosclerosis in rabbits, we evaluated the effects of (2-hydroxypropyl)-β-cyclodextrin (HPβCD) therapy on the organ coefficient, lipid profiles, inflammatory cytokines, and atherosclerotic plaques in rabbits fed a high-fat diet. Our results demonstrated that HPβCD therapy reduced plasma triglyceride levels and inflammatory cytokine levels but increased plasma high-density lipoprotein cholesterol levels. HPβCD therapy produced a significant decrease in the atherosclerotic lesion area and reduced macrophage and collagen content in the lesions. The expression levels of inflammatory genes in aortic plaques were significantly reduced by HPβCD treatment, but the expression of ATP-binding cassette (ABC) transporters A1 (ABCA1) and G1 (ABCG1) in aortic plaques and livers increased significantly. HPβCD therapy may produce additional antiatherosclerotic benefits likely via increasing high-density lipoprotein cholesterol levels.
Collapse
|
19
|
Martínez-Hervás S, González-Navarro H. Terapias antiinflamatorias para la enfermedad cardiovascular: vías de señalización y mecanismos. Rev Esp Cardiol 2019. [DOI: 10.1016/j.recesp.2019.02.022] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Anti-inflammatory Therapies for Cardiovascular Disease: Signaling Pathways and Mechanisms. ACTA ACUST UNITED AC 2019; 72:767-773. [PMID: 31155366 DOI: 10.1016/j.rec.2019.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 03/15/2019] [Indexed: 12/23/2022]
Abstract
Cardiovascular diseases (CVD) are the clinical manifestation of atherosclerosis, a chronic inflammatory disease promoted by several risk factors such as dyslipidemia, type 2 diabetes mellitus, hypertension, and smoking. Acute CVD events are the result of an unresolved inflammatory chronic state that promotes the rupture of unstable plaque lesions. Of note, the existing intensive therapies modify risk factors but do not prevent life-threatening recurrent ischemic events in high-risk patients, who have a residual inflammatory risk displayed by increased C-reactive protein (CRP) levels. Better understanding of the role of innate and adaptive immunity in plaque development and rupture has led to intensive investigation of anti-inflammatory strategies for CVD. Some of them are being tested in specific clinical trials and use lower doses of existing medications originally developed for other inflammatory diseases such as rheumatoid arthritis and psoriasis, which have high CVD risk. Other investigations are retrospective and meta-analyses of existing clinical trials that evaluate the incidence of CVD in these inflammatory diseases. Others are based on preclinical testing such as vaccines. In this article, we summarize the main anti-inflammatory strategies and associated molecular mechanisms that are being evaluated in preclinical or clinical CVD studies.
Collapse
|
21
|
Secreted Factors From Intervertebral Disc Cells and Infiltrating Macrophages Promote Degenerated Intervertebral Disc Catabolism. Spine (Phila Pa 1976) 2019; 44:E520-E529. [PMID: 30540714 DOI: 10.1097/brs.0000000000002953] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
STUDY DESIGN Rat nucleus pulposus (NP) cells or annulus fibrosus (AF) cells were stimulated with conditioned media of RAW 264.7 macrophages and vice versa under healthy culture conditions and in the presence of pro-inflammatory mediators. The gene expression of pro-inflammatory mediators, extracellular matrix (ECM)-modifying enzymes, and chemokines, which play important roles in intervertebral disc degeneration (IDD), was determined. OBJECTIVE To test whether the interaction between native disc cells and infiltrating macrophages accelerates inflammation state, disrupts matrix homeostasis, and promotes inflammatory cells infiltration. SUMMARY OF BACKGROUND DATA With macrophages infiltration, the disc resident cells would be inevitably exposed to macrophages. Macrophages have been shown to play pro-inflammatory role in the cellular interactions with disc cells under healthy culture conditions. However, the biologic interactions between macrophages and disc cells under degenerated disc inflammatory environment remain unknown. METHODS Murine Macrophages RAW 264.7 were cultured in the conditioned media of Rat AF or NP cells culture in the presence or absence of IL-1β stimulation. Similarly, Rat AF or NP cells were also cultured in the conditioned media of Murine Macrophages RAW 264.7 culture in the presence or absence of IFN-γ stimulation. The mRNA levels difference of pro-inflammatory genes, catabolic genes and chemokines genes for AF cells, NP cells and Macrophages RAW 264.7 were analyzed by qRT-PCR, respectively. RESULTS Compared with serum-free media exposure, RAW 264.7 macrophages exposed to AF or NP cells conditioned media selectively modestly upregulated mRNA levels of the aforementioned cytokines. Exposure of RAW 264.7 macrophages to conditioned media from AF or NP cells with IL-1β stimulation dramatically increased mRNA levels of all the investigated cytokines. Similarly, compared with serum-free media exposure, AF or NP cells exposed to RAW 264.7 macrophages conditioned media selectively modestly upregulated mRNA levels of the aforementioned cytokines. Exposure of AF or NP cells to conditioned media from RAW 264.7 macrophages with IFN-γ stimulation dramatically increased mRNA levels of all the investigated cytokines. CONCLUSION The biologic interactions between infiltrating macrophages and native disc cells under degenerated disc inflammatory environment lead to an increasingly severe inflammatory conditions, which may be a self-stimulated process from the macrophages infiltration occurrenceLevel of Evidence: 5.
Collapse
|
22
|
Wang F, Zhang Z, Fang A, Jin Q, Fang D, Liu Y, Wu J, Tan X, Wei Y, Jiang C, Song X. Macrophage Foam Cell-Targeting Immunization Attenuates Atherosclerosis. Front Immunol 2019; 9:3127. [PMID: 30687328 PMCID: PMC6335275 DOI: 10.3389/fimmu.2018.03127] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 12/18/2018] [Indexed: 02/05/2023] Open
Abstract
Background: Macrophage foam cells (FCs) play a crucial role in the initiation and progression of atherosclerosis. Reducing the formation or inducing the removal of FCs could ameliorate atherosclerosis. The present study examined whether the whole-cell vaccination using FCs could be used as novel prevention and treatment strategies to battle atherosclerosis. Methods: ApoE−/− mice with initial or established atherosclerosis were subcutaneously immunized three times with FCs in Freund's adjuvant. Results: Immunization with FCs resulted in an overt reduction of atherosclerotic lesion in the whole aorta and the aortic root with enhanced lesion stability. Subsequent study in mechanism showed that FCs vaccination dramatically increased CD4+ T cell and CD8+ T cell populations. Immunization with FCs significantly raised the plasma FCs-specific IgG antibodies. Of note, the FCs immune plasma could selectively recognize and bind to FC. FCs immune plasma significantly blocked the process of FCs formation, finally reduced the accumulation of FCs in plaque. Additionally, it was observed that FCs immunization down-regulated the expression level of atherosclerosis related pro-inflammatory cytokines, including IFN-γ, MCP-1, and IL-6 and enhanced the lesion stability with a significant increase in TGF-β1 level and collagen content. Conclusions: These findings demonstrate that the whole-cell vaccination using FCs significantly decreased lesion development and positively modulated lesion progression and stability by targeting FCs. The whole-cell FCs vaccine might represent a potential novel strategy for development of new antibodies and vaccines to the prevention or treatment of atherosclerosis.
Collapse
Affiliation(s)
- Fazhan Wang
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Zhi Zhang
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China.,School of Chemical and Pharmaceutical Engineering, Sichuan University of Science and Engineering, Zigong, China
| | - Aiping Fang
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China.,West China School of Public Health, Sichuan University, Chengdu, China
| | - Quansheng Jin
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Dailong Fang
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Yongmei Liu
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Jinhui Wu
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiaoyue Tan
- Department of Pathology/Collaborative Innovation Center of Biotherapy, Medical School of Nankai University, Tianjin, China
| | - Yuquan Wei
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Chunling Jiang
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| | - Xiangrong Song
- State Key Laboratory of Biotherapy, Department of Anesthesiology and Translational Neuroscience Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
23
|
Hong R, Wang Z, Sui A, Liu X, Fan C, Lipkind S, Xu Q. Gingival mesenchymal stem cells attenuate pro-inflammatory macrophages stimulated with oxidized low-density lipoprotein and modulate lipid metabolism. Arch Oral Biol 2018; 98:92-98. [PMID: 30468993 DOI: 10.1016/j.archoralbio.2018.11.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 11/08/2018] [Accepted: 11/09/2018] [Indexed: 12/24/2022]
Abstract
OBJECTIVE To examine the effects of gingival mesenchymal stem cells (GMSCs) on inflammatory macrophages upon oxidized low-density lipoprotein (ox-LDL) stimulation and evaluate therapeutic potential of GMSCs on mouse model of periodontitis associated with hyperlipidemia. METHODS in vitro, GMSCs were co-cultured with macrophages for 48 h in the absence or presence of M1 polarizing conditions and oxidized low-density lipoprotein in the transwell system. The supernatants were collected for ELISA. M1 and M2 markers of macrophages were analyzed by flow cytometry and PCR, and lipid accumulation was assessed by oil red O staining. in vivo, eighteen mice were divided into three groups (n = 6): Group A (periodontally healthy mice as control), Group B (periodontitis mice with hyperlipidemia), Group C (periodontitis mice with hyperlipidemia with the transplantation of GMSCs). The serum levels of cholesterol and inflammatory factors were measured by automatic analyzer. Bone regeneration was evaluated by Masson staining. RESULTS When co-cultured with GMSCs, the M1 markers of Tumor Necrosis Factor (TNF) -α, Interleukin (IL) -6, Interleukin (IL) -1β, CD86, and Human Leukocyte Antigen (HLA) -DR were significantly reduced. In contrast, M2 markers such as Interleukin(IL) -10 and CD206 were moderately increased. Similar results were obtained in the cell culture supernatants. In animal experiment, GMSCs suppressed the expression of sterol regulatory element binding transcription factor 1c (SREBP-1c) and elevated the levels of peroxisome proliferator-activated receptor alpha (PPARα) and peroxisome proliferator activator receptor- coactivator 1(PGC-1α) in the liver, attenuated cholesterol dysfunction via the downregulation of low-density lipoprotein (LDL) and total cholesterol (TC), and the upregulation of high-density lipoprotein (HDL), and decreased the levels of TNF-α and IL-6. Moreover, GMSC treatment improved bone regeneration. CONCLUSION GMSCs inhibit the activation of M1 macrophages, regulate lipid metabolism and reduce inflammatory response, and promote bone regeneration in mouse model of periodontitis associated with hyperlipidemia.
Collapse
Affiliation(s)
- Rundan Hong
- Department of Stomatology, Affiliated Hospital of Qingdao University, College of Stomatology, Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong, China
| | - Zhiguo Wang
- Department of Burn and Plastic Surgery, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Aihua Sui
- Department of the Key Lab, Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xiaoxuan Liu
- Department of Stomatology, Affiliated Hospital of Qingdao University, College of Stomatology, Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong, China; Department of Stomatology, Yidu Central Hospital of Weifang, 4138 Linglong Mountain South Road, Qingzhou, 262500, Shandong, China
| | - Chun Fan
- Department of Stomatology, Affiliated Hospital of Qingdao University, College of Stomatology, Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong, China
| | - Sofya Lipkind
- Department of Molecular and Cellular Biology, University of California, Davis, California, United States
| | - Quanchen Xu
- Department of Stomatology, Affiliated Hospital of Qingdao University, College of Stomatology, Qingdao University, 16 Jiangsu Road, Qingdao, 266003, Shandong, China.
| |
Collapse
|
24
|
New Insight on a Combination of Policosanol and 10-Dehydrogingerdione Phytochemicals as Inhibitors for Platelet Activation Biomarkers and Atherogenicity Risk in Dyslipidemic Rabbits: Role of CETP and PCSK9 Inhibition. Appl Biochem Biotechnol 2018; 186:805-815. [PMID: 29740798 DOI: 10.1007/s12010-018-2776-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2018] [Accepted: 04/30/2018] [Indexed: 02/05/2023]
|
25
|
Sanjadi M, Rezvanie Sichanie Z, Totonchi H, Karami J, Rezaei R, Aslani S. Atherosclerosis and autoimmunity: a growing relationship. Int J Rheum Dis 2018; 21:908-921. [PMID: 29671956 DOI: 10.1111/1756-185x.13309] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Atherosclerosis is regarded as one of the leading causes of mortality and morbidity in the world. Nowadays, it seems that atherosclerosis cannot be defined merely through the Framingham traditional risk factors and that autoimmunity settings exert a remarkable role in its mechanobiology. Individuals with autoimmune disorders show enhanced occurrence of cardiovascular complications and subclinical atherosclerosis. The mechanisms underlying the atherosclerosis in disorders like rheumatoid arthritis, systemic lupus erythematosus, antiphospholipid syndrome, systemic sclerosis and Sjögren's syndrome, seem to be the classical risk factors. However, chronic inflammatory processes and abnormal immune function may also be involved in atherosclerosis development. Autoantigens, autoantibodies, infectious agents and pro-inflammatory mediators exert a role in that process. Being armed with the mechanisms underlying autoimmunity in the etiopathogenesis of atherosclerosis in rheumatic autoimmune disorders and the shared etiologic pathway may result in substantial developing therapeutics for these patients.
Collapse
Affiliation(s)
- Maryam Sanjadi
- Department of Biochemistry, Islamic Azad University, Falavarjan Branch, Tehran, Iran
| | | | - Hamidreza Totonchi
- Department of Biochemistry, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jafar Karami
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ramazan Rezaei
- Department of Immunology, Medical School, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Aslani
- Rheumatology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
26
|
Kasselman LJ, Vernice NA, DeLeon J, Reiss AB. The gut microbiome and elevated cardiovascular risk in obesity and autoimmunity. Atherosclerosis 2018. [DOI: 10.1016/j.atherosclerosis.2018.02.036] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
27
|
Pérez‐Baos S, Barrasa JI, Gratal P, Larrañaga‐Vera A, Prieto‐Potin I, Herrero‐Beaumont G, Largo R. Tofacitinib restores the inhibition of reverse cholesterol transport induced by inflammation: understanding the lipid paradox associated with rheumatoid arthritis. Br J Pharmacol 2017; 174:3018-3031. [PMID: 28646516 PMCID: PMC5573422 DOI: 10.1111/bph.13932] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 06/06/2017] [Accepted: 06/17/2017] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND AND PURPOSE Patients with active rheumatoid arthritis (RA) have increased cardiovascular mortality, paradoxically associated with reduced circulating lipid levels. The JAK inhibitor tofacitinib ameliorates systemic and joint inflammation in RA with a concomitant increase in serum lipids. We analysed the effect of tofacitinib on the lipid profile of hyperlipidaemic rabbits with chronic arthritis (CA) and on the changes in reverse cholesterol transport (RCT) during chronic inflammation. EXPERIMENTAL APPROACH CA was induced in previously immunized rabbits, fed a high-fat diet, by administering four intra-articular injections of ovalbumin. A group of rabbits received tofacitinib (10 mg·kg-1 ·day-1 ) for 2 weeks. Systemic and synovial inflammation and lipid content were evaluated. For in vitro studies, THP-1-derived macrophages were exposed to high lipid concentrations and then stimulated with IFNγ in the presence or absence of tofacitinib in order to study mediators of RCT. KEY RESULTS Tofacitinib decreased systemic and synovial inflammation and increased circulating lipid levels. Although it did not modify synovial macrophage density, it reduced the lipid content within synovial macrophages. In foam macrophages in culture, IFNγ further stimulated intracellular lipid accumulation, while the JAK/STAT inhibition provoked by tofacitinib induced lipid release by increasing the levels of cellular liver X receptor α and ATP-binding cassette transporter (ABCA1) synthesis. CONCLUSIONS AND IMPLICATIONS Active inflammation could be associated with lipid accumulation within macrophages of CA rabbits. JAK inhibition induced lipid release through RCT activation, providing a plausible explanation for the effect of tofacitinib on the lipid profile of RA patients.
Collapse
Affiliation(s)
- S Pérez‐Baos
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - J I Barrasa
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
| | - P Gratal
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - A Larrañaga‐Vera
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - I Prieto‐Potin
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - G Herrero‐Beaumont
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| | - R Largo
- Bone and Joint Research Unit, Rheumatology DepartmentIIS‐Fundación Jiménez Díaz UAMMadridSpain
- Thematic Network on Aging and Frailty (RETICEF)MadridSpain
| |
Collapse
|
28
|
Orekhov AN, Ivanova EA, Melnichenko AA, Sobenin IA. Circulating desialylated low density lipoprotein. COR ET VASA 2017. [DOI: 10.1016/j.crvasa.2016.10.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Sukhorukov VN, Karagodin VP, Orekhov AN. [Atherogenic modification of low-density lipoproteins]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2017; 62:391-402. [PMID: 27562992 DOI: 10.18097/pbmc20166204391] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
One of the first manifestations of atherosclerosis is accumulation of extra- and intracellular cholesterol esters in the arterial intima. Formation of foam cells is considered as a trigger in the pathogenesis of atherosclerosis. Low density lipoprotein (LDL) circulating in human blood is the source of lipids accumulated in the arterial walls. This review considered features and role in atherogenesis different modified forms of LDL: oxidized, small dense, electronegative and especially desialylated LDL. Desialylated LDL of human blood plasma is capable to induce lipid accumulation in cultured cells and it is atherogenic. LDL possesses numerous alterations of protein, carbohydrate and lipid moieties and therefore can be termed multiple-modified LDL. Multiple modification of LDL occurs in human blood plasma and represents a cascade of successive changes in the lipoprotein particle: desialylation, loss of lipids, reduction in the particle size, increase of surface electronegative charge, etc. In addition to intracellular lipid accumulation, stimulatory effects of naturally occurring multiple-modified LDL on other processes involved in the development of atherosclerotic lesions, namely cell proliferation and fibrosis, were shown.
Collapse
Affiliation(s)
- V N Sukhorukov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - V P Karagodin
- Plekhanov Russian University of Economics, Moscow, Russia
| | - A N Orekhov
- Institute of General Pathology and Pathophysiology, Moscow, Russia; Institute for Atherosclerosis Research, Skolkovo Innovative Center, PO Box #21, Moscow, Russia
| |
Collapse
|
30
|
Shen J, Hafeez A, Stevenson J, Yang J, Yin C, Li F, Wang S, Du H, Ji X, Rafols JA, Geng X, Ding Y. Omega-3 fatty acid supplement prevents development of intracranial atherosclerosis. Neuroscience 2016; 334:226-235. [DOI: 10.1016/j.neuroscience.2016.08.013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2016] [Revised: 07/15/2016] [Accepted: 08/07/2016] [Indexed: 11/26/2022]
|
31
|
Ding Y, Chen J, Cui G, Wei Y, Lu C, Wang L, Diao H. Pathophysiological role of osteopontin and angiotensin II in atherosclerosis. Biochem Biophys Res Commun 2016; 471:5-9. [PMID: 26828266 DOI: 10.1016/j.bbrc.2016.01.142] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Accepted: 01/22/2016] [Indexed: 01/13/2023]
Affiliation(s)
- Yulong Ding
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003 Hangzhou, China
| | - Jianing Chen
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003 Hangzhou, China
| | - Guangying Cui
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003 Hangzhou, China
| | - Yingfeng Wei
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003 Hangzhou, China
| | - Chong Lu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003 Hangzhou, China
| | - Lin Wang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003 Hangzhou, China
| | - Hongyan Diao
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, College of Medicine, Zhejiang University, 310003 Hangzhou, China.
| |
Collapse
|