1
|
Zhou W, Li X, Zhou H, Hu Y, Chen Y, Guo D. TNF-α/IL-1β/IL-1α/IL-12 inflammatory cytokine axes coupled with TLR1/TLR3/TLR5/MYD88 immune signaling pathway over-activation contribute to simultaneous carotid and coronary artery and occlusion in elderly patients. Cytokine 2025; 185:156808. [PMID: 39556941 DOI: 10.1016/j.cyto.2024.156808] [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: 06/11/2024] [Revised: 10/22/2024] [Accepted: 11/10/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND It remains difficult to evaluate the risk factors for concomitant carotid artery as well as coronary artery diseases in elderly patients. The aim of this research was to determine the TNF-α/IL-1β/IL-1α/IL-12 axes-TLR1/TLR3/TLR5/MYD88 immune signaling pathway interactions in coexistent carotid artery occlusion and coronary artery occlusion in elderly patients. METHODS Elderly patients, who underwent carotid ultrasonography and coronary computed tomography angiography, were consecutively included in this research. The analyzed groups consisted of those with coexistent carotid artery occlusion and coronary artery occlusion as well as healthy individuals were enrolled as control group. The circulating levels of tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), interleukin-1α (IL-1α), interleukin-12 (IL-12), toll-like receptor 1 (TLR1), toll-like receptor 3 (TLR3), toll-like receptor 5 (TLR5) and myeloid differentiation factor 88 (MYD88) were measured. RESULTS The biomarkers (TNF-α, IL-1β, IL-1α, IL-12, TLR1, TLR3, TLR5 and MYD88) were significantly increased in carotid artery occlusion + left circumflex coronary artery occlusion group when compared with control group and carotid artery occlusion + right coronary artery occlusion group, respectively (P < 0.001), and were further elevated in carotid artery occlusion + left anterior descending coronary artery occlusion group when compared to carotid artery occlusion + right coronary artery occlusion group and carotid artery occlusion + left circumflex coronary artery occlusion group, respectively (P < 0.001). CONCLUSION This research demonstrated that the TNF-α/IL-1β/IL-1α/IL-12 axes and TLR1/TLR3/TLR5/MYD88 immune signaling pathway implicated in the pathogenesis of carotid artery occlusion with coronary artery occlusion in elderly patients.
Collapse
Affiliation(s)
- Wenhang Zhou
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China
| | - Xia Li
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China
| | - Hualan Zhou
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Youdong Hu
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Ying Chen
- Department of Geriatrics, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223002, China
| | - Dianxuan Guo
- Xiamen Road Branch Hospital, The Affiliated Huaian Hospital of Xuzhou Medical University, Huaian 223005, China.
| |
Collapse
|
2
|
Kawamata T, Wakimoto A, Nishikawa T, Ikezawa M, Hamada M, Inoue Y, Kulathunga K, Salim FN, Kanai M, Nishino T, Gentleman K, Liu C, Mathis BJ, Obana N, Fukuda S, Takahashi S, Taya Y, Sakai S, Hiramatsu Y. Natto consumption suppresses atherosclerotic plaque progression in LDL receptor-deficient mice transplanted with iRFP-expressing hematopoietic cells. Sci Rep 2023; 13:22469. [PMID: 38110459 PMCID: PMC10728071 DOI: 10.1038/s41598-023-48562-y] [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] [Accepted: 11/28/2023] [Indexed: 12/20/2023] Open
Abstract
Natto, known for its high vitamin K content, has been demonstrated to suppress atherosclerosis in large-scale clinical trials through a yet-unknown mechanism. In this study, we used a previously reported mouse model, transplanting the bone marrow of mice expressing infra-red fluorescent protein (iRFP) into LDLR-deficient mice, allowing unique and non-invasive observation of foam cells expressing iRFP in atherosclerotic lesions. Using 3 natto strains, we meticulously examined the effects of varying vitamin K levels on atherosclerosis in these mice. Notably, high vitamin K natto significantly reduced aortic staining and iRFP fluorescence, indicative of decreased atherosclerosis. Furthermore, mice administered natto showed changes in gut microbiota, including an increase in natto bacteria within the cecum, and a significant reduction in serum CCL2 expression. In experiments with LPS-stimulated macrophages, adding natto decreased CCL2 expression and increased anti-inflammatory cytokine IL-10 expression. This suggests that natto inhibits atherosclerosis through suppression of intestinal inflammation and reduced CCL2 expression in macrophages.
Collapse
Affiliation(s)
- Takeshi Kawamata
- Tsukuba Medical Center Hospital, 1-3-1, Amakubo, Tsukuba, Ibaraki, 305-8558, Japan
- Doctoral Program in Medical Sciences, Graduate School of Comprehensive Human Sciences, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Arata Wakimoto
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
- Ph.D. Program in Human Biology, School of Integrative and Global Majors, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Takanobu Nishikawa
- Department of Natto Research and Development, Takanofoods Corporation, 1542, Noda, Omitama, Ibaraki, 311-3411, Japan.
| | - Masaya Ikezawa
- Department of Natto Research and Development, Takanofoods Corporation, 1542, Noda, Omitama, Ibaraki, 311-3411, Japan
| | - Michito Hamada
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Yuri Inoue
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Kaushalya Kulathunga
- Department of Physiology, Faculty of Medicine, Sabaragamuwa University of Sri Lanka, P.O. Box 01, Hidellana, Ratnapura, Sri Lanka
| | - Filiani Natalia Salim
- Centre for Medical Science and Technology and Healthcare Equity, Parahyangan Catholic University, Bandung, 40141, Indonesia
- Magister Program of Biomedical Sciences, Universitas Padjadjaran, Sumedang, 45363, Indonesia
| | - Maho Kanai
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Teppei Nishino
- Tsukuba Medical Center Hospital, 1-3-1, Amakubo, Tsukuba, Ibaraki, 305-8558, Japan
| | - Kyle Gentleman
- Integrated Master of Science Natural Sciences, University of Southampton, Highfield, Southampton, SO17 1BJ, Hampshire, UK
| | - Chang Liu
- Department of Cardiovascular Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Bryan J Mathis
- Department of Cardiovascular Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan
| | - Nozomu Obana
- Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
- Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, 246-2 Mizukami, Kakuganji, Tsuruoka-shi, Yamagata, 997-0052, Japan
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, 3-25-13 Tonomachi, Kawasaki-ku, Kawasaki, Kanagawa, 210-0821, Japan
- Transborder Medical Research Center, University of Tsukuba, 1-1-1 Tennodai, Tsukuba-shi, Ibaraki, 305-8575, Japan
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, 2-1-1 Hongo, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
- Laboratory Animal Resource Center, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| | - Yuki Taya
- Department of Natto Research and Development, Takanofoods Corporation, 1542, Noda, Omitama, Ibaraki, 311-3411, Japan
| | - Satoshi Sakai
- Department of Cardiovascular Medicine, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
- Faculty of Health Sciences, Tsukuba University of Technology, 4-12-7, Kasuga, Tsukuba, Ibaraki, 305-8521, Japan.
| | - Yuji Hiramatsu
- Department of Cardiovascular Surgery, Faculty of Medicine, University of Tsukuba, 1-1-1, Tennodai, Tsukuba, Ibaraki, 305-8575, Japan.
| |
Collapse
|
3
|
Curini L, Alushi B, Christopher MR, Baldi S, Di Gloria L, Stefano P, Laganà A, Iannone L, Grubitzsch H, Landmesser U, Ramazzotti M, Niccolai E, Lauten A, Amedei A. The first taxonomic and functional characterization of human CAVD-associated microbiota. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:36-48. [PMID: 36789351 PMCID: PMC9896411 DOI: 10.15698/mic2023.02.791] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 12/23/2022] [Accepted: 01/09/2023] [Indexed: 02/05/2023]
Abstract
Introduction Calcific aortic valve disease (CAVD) is the most common heart valve disorder, defined by a remodeling multistep process: namely, valve fibrosis with its area narrowing, impaired blood flow, and final calcification phase. Nowadays, the only treatment is the surgical valve replacement. As for other cardiovascular diseases, growing evidence suggest an active role of the immune system in the calcification process that could be modulated by the microbiota. To address this point, we aimed to investigate and characterize, for the first time, the presence of a valve microbiota and associated immune response in human CAVD. Method Calcified aortic valve (CAV) samples from twenty patients (11 from Germany and 9 from Italy) with diagnosis of severe symptomatic CAVD were used to assess the presence of infiltrating T cells, by cloning approach, and to characterize the valve microbiota, by 16S rRNA gene sequencing (NGS). Results We documented the presence of infiltrating T lymphocytes, especially the T helper subset, in CAV samples. Moreover, we found a tissue-associated microbiota in freshly collected CAV samples, which was significantly different in Italian and German patients, suggesting potential correlation with other cardiovascular risk factors. Conclusion The presence of microbiota in inflamed CAV samples represents the right trigger point to explain the valve calcification process, encouraging further studies to explore the potential link between bacteria and adaptive immune response and to define the critical role of local microbiota-immunity axis on CAVD development.
Collapse
Affiliation(s)
- Lavinia Curini
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Brunilda Alushi
- Department of Cardiology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, and German Centre for Cardiovascular Research (DZHK); Department of Interventional Cardiology, Klinik Vincentinum Augsburg, Germany
| | - Mary Roxana Christopher
- Department of Cardiology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, and German Centre for Cardiovascular Research (DZHK)
| | - Simone Baldi
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Leandro Di Gloria
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | | | - Anna Laganà
- Cardiac Surgery, Careggi University Hospital, 50134 Florence, Italy
| | - Luisa Iannone
- Cardiac Surgery, Careggi University Hospital, 50134 Florence, Italy
| | - Herko Grubitzsch
- Berlin Institute of Health; Department of Cardiology, German Heart Centre Berlin (DHZB)
| | - Ulf Landmesser
- Department of Cardiology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin and German Centre for Cardiovascular Research (DZHK); Berlin Institute of Health
| | - Matteo Ramazzotti
- Department of Biomedical, Experimental and Clinical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Elena Niccolai
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy
| | - Alexander Lauten
- Department of Cardiology, Campus Benjamin Franklin, Charité Universitätsmedizin Berlin, and German Centre for Cardiovascular Research (DZHK); Department of Interventional Cardiology, Klinik Vincentinum Augsburg, Germany
| | - Amedeo Amedei
- Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy.
,SOD of Interdisciplinary Internal Medicine, Azienda Ospedaliera Universitaria Careggi (AOUC), 50139 Florence, Italy.
,* Corresponding Author: Amedeo Amedei, Department of Clinical and Experimental Medicine, University of Florence, 50139 Florence, Italy; E-mail:
| |
Collapse
|
4
|
Ren H, Zhu B, An Y, Xie F, Wang Y, Tan Y. Immune communication between the intestinal microbiota and the cardiovascular system. Immunol Lett 2023; 254:13-20. [PMID: 36693435 DOI: 10.1016/j.imlet.2023.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/27/2022] [Accepted: 01/19/2023] [Indexed: 01/22/2023]
Abstract
The intestine hosts a large number of microbial communities. Recent studies have shown that gut microbiota-mediated immune responses play a vital role in developing cardiovascular diseases (CVD). Immune cells are extensively infiltrated in the gut and heart tissues, such as T cells, B cells, and macrophages. They play a crucial role in the crosstalk between the heart and gut microbiota. And the microbiota influences the bidirectional function of immune cells in CVD such as myocardial infarction and atherosclerosis, including through metabolites. The mapping of immune cell-mediated immune networks in the heart and gut provides us with new targets for treating CVD. This review discusses the role of immune cells in gut microbiota and cardiac communication during health and CVD.
Collapse
Affiliation(s)
- Hao Ren
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Botao Zhu
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Yuze An
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Feng Xie
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Yichuan Wang
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China
| | - Yurong Tan
- Department of Medical Microbiology, Central South University Changsha, Hunan Provinces, China.
| |
Collapse
|
5
|
Orzuna-Orzuna JF, Dorantes-Iturbide G, Lara-Bueno A, Chay-Canul AJ, Miranda-Romero LA, Mendoza-Martínez GD. Meta-analysis of flavonoids use into beef and dairy cattle diet: Performance, antioxidant status, ruminal fermentation, meat quality, and milk composition. Front Vet Sci 2023; 10:1134925. [PMID: 36876000 PMCID: PMC9975267 DOI: 10.3389/fvets.2023.1134925] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 01/30/2023] [Indexed: 02/17/2023] Open
Abstract
The objective of this study was to evaluate the effects of dietary supplementation with flavonoids (FLAs) on animal performance, diet digestibility, antioxidant status in blood serum, rumen parameters, meat quality, and milk composition in beef and dairy cattle through a meta-analysis. Thirty-six peer-reviewed publications were included in the data set. The weighted mean differences (WMD) between the FLAs treatments and the control treatment were used to assess the effect size. Dietary supplementation with FLAs decreased feed conversion ratio (WMD = -0.340 kg/kg; p = 0.050) and increased (p < 0.05) dry matter intake (WMD = 0.191 kg/d), dry matter digestibility (WMD = 15.283 g/kg of DM), and daily weight gain (WMD = 0.061 kg/d). In blood serum, FLAs supplementation decreased the serum concentration of malondialdehyde (WMD = -0.779 nmol/mL; p < 0.001) and increased (p < 0.01) the serum concentration of superoxide dismutase (WMD = 8.516 U/mL), glutathione peroxidase (WMD = 12.400 U/mL) and total antioxidant capacity (WMD = 0.771 U/mL). A higher ruminal propionate concentration (WMD = 0.926 mol/100 mol; p = 008) was observed in response to FLAs supplementation. In meat, the dietary inclusion of FLAs decreased (p < 0.05) shear force (WMD = -1.018 kgf/cm2), malondialdehyde content (WMD = -0.080 mg/kg of meat), and yellowness (WMD = -0.460). Supplementation with FLAs decreased milk somatic cell count (WMD = -0.251 × 103 cells/mL; p < 0.001) and increased (p < 0.01) milk production (WMD = 1.348 kg/d), milk protein content (WMD = 0.080/100 g) and milk fat content (WMD = 0.142/100 g). In conclusion, dietary supplementation with FLAs improves animal performance and nutrient digestibility in cattle. In addition, FLAs improve the antioxidant status in blood serum and the quality of meat and milk.
Collapse
|
6
|
Wang C, Deng H, Liu F, Yin Q, Xia L. The Role of Gut Microbiota in the Immunopathology of Atherosclerosis: focus on immune cells. Scand J Immunol 2022; 96:e13174. [PMID: 35474231 DOI: 10.1111/sji.13174] [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: 02/06/2022] [Revised: 03/27/2022] [Accepted: 04/12/2022] [Indexed: 11/27/2022]
Abstract
Gut microbiota (GM) play important roles in multiple organ function, homeostasis and several diseases. More recently, increasing evidences have suggested that the compositional and functional alterations of GM play a crucial role in the accumulation of foam cells and the formation of atherosclerotic plaque in atherosclerosis. In particular, the effects of bacterial components and metabolites on innate and adaptive immune cells have been explored as the underlying mechanisms. Understanding the effects of GM and metabolites on immunoregulation are important for clinical therapy for atherosclerosis. Herein, we summarize the potential role of the GM (such as bacterial components lipopolysaccharide and peptidoglycan) and GM-derived metabolites (such as short-chain fatty acids, trimethylamine N-oxide and bile acids) in the immunopathology of atherosclerosis. Based on that, we further discuss the anti-atherosclerotic effects of GM-directed dietary bioactive factors such as dietary fibers, dietary polyphenols and probiotics. Because of drug-induced adverse events in anti-inflammatory therapies, personalized dietary interventions would be potential therapies for atherosclerosis, and the interactions between GM-derived products and immune cells should be studied further.
Collapse
Affiliation(s)
- Chong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.,International Genome Center, Jiangsu University, Zhenjiang, China
| | - Hualing Deng
- Operating room, Weihai Municipal Hospital, Weihai, China
| | - Fang Liu
- International Genome Center, Jiangsu University, Zhenjiang, China
| | - Qing Yin
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lin Xia
- Department of Laboratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, China.,International Genome Center, Jiangsu University, Zhenjiang, China
| |
Collapse
|
7
|
Anto L, Blesso CN. Interplay Between Diet, the Gut Microbiome, and Atherosclerosis: Role of Dysbiosis and Microbial Metabolites on Inflammation and Disordered Lipid Metabolism. J Nutr Biochem 2022; 105:108991. [DOI: 10.1016/j.jnutbio.2022.108991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/21/2021] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
|
8
|
Han L, Fu Q, Deng C, Luo L, Xiang T, Zhao H. Immunomodulatory potential of flavonoids for the treatment of autoimmune diseases and tumour. Scand J Immunol 2021. [DOI: 10.1111/sji.13106] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Limin Han
- Department of Pathophysiology Zunyi Medical University Zunyi China
- Department of Endocrinology People’s Hospital of Changshou Chongqing Chongqing China
| | - Qiang Fu
- Organ Transplantation Center Sichuan Academy of Medical Sciences and Sichuan Provincial People’s Hospital School of Medicine University of Electronic Science and Technology of China Chengdu China
| | - Chuan Deng
- Department of Neurology People’s Hospital of Changshou Chongqing Chongqing China
| | - Li Luo
- Department of Forensic Medicine Zunyi Medical University Zunyi China
| | - Tengxiao Xiang
- Department of Endocrinology People’s Hospital of Changshou Chongqing Chongqing China
| | - Hailong Zhao
- Department of Pathophysiology Zunyi Medical University Zunyi China
| |
Collapse
|
9
|
Karlsen TR, Kong XY, Holm S, Quiles-Jiménez A, Dahl TB, Yang K, Sagen EL, Skarpengland T, S Øgaard JD, Holm K, Vestad B, Olsen MB, Aukrust P, Bjørås M, Hov JR, Halvorsen B, Gregersen I. NEIL3-deficiency increases gut permeability and contributes to a pro-atherogenic metabolic phenotype. Sci Rep 2021; 11:19749. [PMID: 34611194 PMCID: PMC8492623 DOI: 10.1038/s41598-021-98820-0] [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: 07/02/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis and its consequences cause considerable morbidity and mortality world-wide. We have previously shown that expression of the DNA glycosylase NEIL3 is regulated in human atherosclerotic plaques, and that NEIL3-deficiency enhances atherogenesis in Apoe-/- mice. Herein, we identified a time point prior to quantifiable differences in atherosclerosis between Apoe-/-Neil3-/- mice and Apoe-/- mice. Mice at this age were selected to explore the metabolic and pathophysiological processes preceding extensive atherogenesis in NEIL3-deficient mice. Untargeted metabolomic analysis of young Apoe-/-Neil3-/- mice revealed significant metabolic disturbances as compared to mice expressing NEIL3, particularly in metabolites dependent on the gut microbiota. 16S rRNA gene sequencing of fecal bacterial DNA indeed confirmed that the NEIL3-deficient mice had altered gut microbiota, as well as increased circulating levels of the bacterially derived molecule LPS. The mice were challenged with a FITC-conjugated dextran to explore gut permeability, which was significantly increased in the NEIL3-deficient mice. Further, immunohistochemistry showed increased levels of the proliferation marker Ki67 in the colonic epithelium of NEIL3-deficient mice, suggesting increased proliferation of intestinal cells and gut leakage. We suggest that these metabolic alterations serve as drivers of atherosclerosis in NEIL3-deficient mice.
Collapse
Affiliation(s)
- Tom Rune Karlsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ana Quiles-Jiménez
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ellen L Sagen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tonje Skarpengland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jonas D S Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kristian Holm
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Beate Vestad
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Maria B Olsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Magnar Bjørås
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| |
Collapse
|
10
|
Lipid accumulation and novel insight into vascular smooth muscle cells in atherosclerosis. J Mol Med (Berl) 2021; 99:1511-1526. [PMID: 34345929 DOI: 10.1007/s00109-021-02109-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 06/03/2021] [Accepted: 06/29/2021] [Indexed: 12/15/2022]
Abstract
Atherosclerosis is a chronic and progressive process. It is the most important pathological basis of cardiovascular disease and stroke. Vascular smooth muscle cells (VSMCs) are an essential cell type in atherosclerosis. Previous studies have revealed that VSMCs undergo phenotypic transformation in atherosclerosis to participate in the retention of atherogenic lipoproteins as well as the formation of the fibrous cap and the underlying necrotic core in plaques. The emergence of lineage-tracing studies indicates that the function and number of VSMCs in plaques have been greatly underestimated. In addition, recent studies have revealed that VSMCs make up at least 50% of the foam cell population in human and mouse atherosclerotic lesions. Therefore, understanding the formation of lipid-loaded VSMCs and their regulatory mechanisms is critical to elucidate the pathogenesis of atherosclerosis and to explore potential therapeutic targets. Moreover, combination of many complementary technologies such as lineage tracing, single-cell RNA sequencing (scRNA-seq), flow cytometry, and mass cytometry (CyTOF) with immunostaining has been performed to further understand the complex VSMC function. Correct identification of detrimental and beneficial processes may reveal successful therapeutic treatments targeting VSMCs and their derivatives during atherosclerosis. The purpose of this review is to summarize the process of lipid-loaded VSMC formation in atherosclerosis and to describe novel insight into VSMCs gained by using multiple advanced methods.
Collapse
|
11
|
Chen L, Ishigami T, Doi H, Arakawa K, Tamura K. The Types and Proportions of Commensal Microbiota Have a Predictive Value in Coronary Heart Disease. J Clin Med 2021; 10:3120. [PMID: 34300286 PMCID: PMC8303676 DOI: 10.3390/jcm10143120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/12/2021] [Accepted: 06/24/2021] [Indexed: 12/13/2022] Open
Abstract
Previous clinical studies have suggested that commensal microbiota play an important role in atherosclerotic cardiovascular disease; however, a synthetic analysis of coronary heart disease (CHD) has yet to be performed. Therefore, we aimed to investigate the specific types of commensal microbiota associated with CHD by performing a systematic review of prospective observational studies that have assessed associations between commensal microbiota and CHD. Of the 544 published articles identified in the initial search, 16 publications with data from 16 cohort studies (2210 patients) were included in the analysis. The combined data showed that Bacteroides and Prevotella were commonly identified among nine articles (n = 13) in the fecal samples of patients with CHD, while seven articles commonly identified Firmicutes. Moreover, several types of commensal microbiota were common to atherosclerotic plaque and blood or gut samples in 16 cohort studies. For example, Veillonella, Proteobacteria, and Streptococcus were identified among the plaque and fecal samples, whereas Clostridium was commonly identified among blood and fecal samples of patients with CHD. Collectively, our findings suggest that several types of commensal microbiota are associated with CHD, and their presence may correlate with disease markers of CHD.
Collapse
Affiliation(s)
- Lin Chen
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, Kanagawa 236-0027, Japan; (L.C.); (H.D.); (K.A.); (K.T.)
- Department of Cardiology, Sir Run Run Hospital, Nanjing Medical University, Nanjing 210029, China
| | - Tomoaki Ishigami
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, Kanagawa 236-0027, Japan; (L.C.); (H.D.); (K.A.); (K.T.)
| | - Hiroshi Doi
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, Kanagawa 236-0027, Japan; (L.C.); (H.D.); (K.A.); (K.T.)
| | - Kentaro Arakawa
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, Kanagawa 236-0027, Japan; (L.C.); (H.D.); (K.A.); (K.T.)
| | - Kouichi Tamura
- Department of Medical Science and Cardio-Renal Medicine, Graduate School of Medicine, Yokohama City University, Kanagawa 236-0027, Japan; (L.C.); (H.D.); (K.A.); (K.T.)
| |
Collapse
|
12
|
Gui DD, Luo W, Yan BJ, Ren Z, Tang ZH, Liu LS, Zhang JF, Jiang ZS. Effects of gut microbiota on atherosclerosis through hydrogen sulfide. Eur J Pharmacol 2021; 896:173916. [PMID: 33529724 DOI: 10.1016/j.ejphar.2021.173916] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 01/14/2021] [Accepted: 01/26/2021] [Indexed: 12/23/2022]
Abstract
Cardiovascular diseases are the leading cause of death and morbidity worldwide. Atherosclerotic cardiovascular disease (ASCVD) is affected by both environmental and genetic factors. Microenvironmental disorders of the human gut flora are associated with a variety of health problems, not only gastrointestinal diseases, such as inflammatory bowel disease, but also extralintestinal organs. Hydrogen sulfide (H2S) is the third gas signaling molecule other than nitric oxide and carbon monoxide. In the cardiovascular system, H2S plays important roles in the regulation of blood pressure, angiogenesis, smooth muscle cell proliferation and apoptosis, anti-oxidative stress, cardiac functions. This review is aiming to explore the potential role of gut microbiota in the development of atherosclerosis through hydrogen sulfide production as a novel therapeutic direction for atherosclerosis.
Collapse
Affiliation(s)
- Dan-Dan Gui
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Wen Luo
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Bin-Jie Yan
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Zhong Ren
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Zhi-Han Tang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Lu-Shan Liu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China
| | - Ji-Feng Zhang
- Cardiovascular Center, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, 421001, China.
| |
Collapse
|