1
|
Huang X, Xie M, Wang Y, Lu X, Mei F, Zhang K, Yang X, Chen G, Yin Y, Feng G, Song W, Dong N, Deng X, Wang S, Chen L. Porphyromonas gingivalis aggravates atherosclerotic plaque instability by promoting lipid-laden macrophage necroptosis. Signal Transduct Target Ther 2025; 10:171. [PMID: 40404630 PMCID: PMC12098900 DOI: 10.1038/s41392-025-02251-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 03/04/2025] [Accepted: 04/28/2025] [Indexed: 05/24/2025] Open
Abstract
At advanced phases of atherosclerosis, the rupture and thrombogenesis of vulnerable plaques emerge as primary triggers for acute cardiovascular events and fatalities. Pathogenic infection such as periodontitis-associated Porphyromonas gingivalis (Pg) has been suspected of increasing the risks of atherosclerotic cardiovascular disease, but its relationship with atherosclerotic plaque destabilization remains elusive. Here we demonstrated that the level of Pg-positive clusters positively correlated with the ratio of necrotic core area to total atherosclerotic plaque area in human clinical samples, which indicates plaque instability. In rabbits and Apoe-/- mice, Pg promoted atherosclerotic plaque necrosis and aggravated plaque instability by triggering oxidative stress, which led to macrophage necroptosis. This process was accompanied by the decreased protein level of forkhead box O3 (FOXO3) in macrophages. The mechanistic dissection showed that Pg lipopolysaccharide (LPS) evoked macrophage oxidative stress via the TLR4 signaling pathway, which subsequently activated MAPK/ERK-mediated FOXO3 phosphorylation and following degradation. While the gingipains, a class of proteases produced by Pg, could effectively hydrolyze FOXO3 in the cytoplasm of macrophages. Both of them decreased the nuclear level of FOXO3, followed by the release of histone deacetylase 2 (HDAC2) from the macrophage scavenger receptor 1 (Msr1) promoter, thus promoting Msr1 transcription. This enhanced MSR1-mediated lipid uptake further amplified oxidative stress-induced necroptosis in lipid-laden macrophages. In summary, Pg exacerbates macrophage oxidative stress-dependent necroptosis, thus enlarges the atherosclerotic plaque necrotic core and ultimately promotes plaque destabilization.
Collapse
Affiliation(s)
- Xiaofei Huang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Mengru Xie
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Yixuan Wang
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China
| | - Xiaofeng Lu
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Feng Mei
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Kaiwen Zhang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Xinlong Yang
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guangjin Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Ying Yin
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Guangxia Feng
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Wencheng Song
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Key Laboratory of Organ Transplantation, Ministry of Education, NHC Key Laboratory of Organ Transplantation, Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, China.
| | - Xuliang Deng
- Department of Geriatric Dentistry, NMPA Key Laboratory for Dental Materials, National Engineering Laboratory for Digital and Material, Technology of Stomatology, Beijing Laboratory of Biomedical Materials, Peking University School and Hospital of Stomatology, Beijing, China.
| | - Songling Wang
- Laboratory of Homeostatic Medicine, School of Medicine, Southern University of Science and Technology, Shenzhen, China.
- Beijing Laboratory of Oral Health, Capital Medical University, Beijing, China.
| | - Lili Chen
- Department of Stomatology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China.
| |
Collapse
|
2
|
Gyriki D, Nikolaidis CG, Bezirtzoglou E, Voidarou C, Stavropoulou E, Tsigalou C. The gut microbiota and aging: interactions, implications, and interventions. FRONTIERS IN AGING 2025; 6:1452917. [PMID: 40438731 PMCID: PMC12116569 DOI: 10.3389/fragi.2025.1452917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 04/25/2025] [Indexed: 06/01/2025]
Abstract
The human microbiota, a complex ecosystem of microorganisms inhabiting various body sites, particularly the gut, plays a crucial role in maintaining health and influencing disease susceptibility. Dysbiosis, characterized by alterations in microbial composition and diversity, has been implicated in numerous diseases, including those associated with aging. This review examines the complex relationship between gut microbiota and aging, highlighting the age-associated gut microbiota alterations, the factors contributing to these changes, the links between microbiota and age-related diseases, and the potential of interventions targeting the microbiome to extend lifespan and improve health outcomes in the elderly. Further research is needed to unravel the intricate mechanisms underlying the interplay between the microbiome and aging, paving the way for innovative strategies to promote healthy aging.
Collapse
Affiliation(s)
- Despoina Gyriki
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | | | - Eugenia Bezirtzoglou
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Chrysa Voidarou
- Department of Agriculture, University of Ioannina, Arta, Greece
| | - Elisavet Stavropoulou
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| | - Christina Tsigalou
- Master Program in “Food, Nutrition and Microbiome”, Department of Medicine, Democritus University of Thrace, Alexandroupolis, Greece
| |
Collapse
|
3
|
Tsalamandris S, Koliastasis L, Miliou A, Oikonomou E, Papageorgiou N, Antonopoulos A, Hatzis G, Mourouzis K, Vogiatzi G, Siasos G, Xaplanteris P, Tousoulis D. Endothelial Function and Pro-Inflammatory Cytokines as Prognostic Markers in Acute Coronary Syndromes. Diagnostics (Basel) 2025; 15:1033. [PMID: 40310443 PMCID: PMC12025570 DOI: 10.3390/diagnostics15081033] [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: 12/26/2024] [Revised: 03/12/2025] [Accepted: 03/28/2025] [Indexed: 05/02/2025] Open
Abstract
Background: Endothelial dysfunction and inflammation are associated with the progression of coronary artery disease (CAD) and the pathophysiology of acute coronary syndrome (ACS). We examined the prognostic role of endothelial function and pro-inflammatory cytokines in patients admitted with ACS. Methods: The study population consisted of 864 subjects. From 663 subjects who presented with chest pain, ACS was diagnosed in 460. We additionally recruited 201 consecutive patients with stable CAD. Endothelial function was assessed using flow-mediated dilatation (FMD). Tumor necrosis factor alpha (TNF-α) and interleukin-6 (IL-6) levels were measured via ELISA. Subjects with ACS were followed up for major adverse cardiovascular events (MACE), defined as cardiovascular death, cardiac arrest, myocardial infarction, stroke, nonfatal stroke, other arterial thrombotic events, and hospitalization due to cardiovascular conditions. Results: There was a stepwise impairment in FMD, logTNF-α, and logIL-6 in patients with chest pain of non-epicardial CAD etiology compared to patients with stable CAD and those with ACS (p < 0.001 for all). Moreover, patients who presented with chest pain had increased odds of ACS in accordance with the increasing levels of TNF-α, IL-6, and impaired FMD (p < 0.05 for all). Interestingly, from all these markers, in patients with ACS, we found that only TNF-α levels above 5.19 pg/mL had a 2.5-times-increased risk of MACE compared to patients with TNF-α levels below 5.19 pg/mL, independently of other confounders. Conclusions: In the current study, we found that patients who presented with ACS had impaired endothelial function and increased levels of IL-6 and TNF-α.
Collapse
Affiliation(s)
- Sotirios Tsalamandris
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| | - Leonidas Koliastasis
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| | - Antigoni Miliou
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, “Sotiria” Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.O.); (K.M.); (G.S.)
| | - Nikos Papageorgiou
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| | - Alexis Antonopoulos
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| | - George Hatzis
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| | - Konstantinos Mourouzis
- 3rd Department of Cardiology, “Sotiria” Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.O.); (K.M.); (G.S.)
| | - Georgia Vogiatzi
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| | - Gerasimos Siasos
- 3rd Department of Cardiology, “Sotiria” Chest Disease Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece; (E.O.); (K.M.); (G.S.)
| | - Panagiotis Xaplanteris
- Department of Cardiology, Centre Hospitalier Universitaire Saint-Pierre, Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Dimitris Tousoulis
- 1st Department of Cardiology, School of Medicine, ‘Hippokration’ General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece; (S.T.); (L.K.); (A.M.); (N.P.); (A.A.); (G.H.); (G.V.); (D.T.)
| |
Collapse
|
4
|
Gillan JL, Jaeschke L, Kuebler WM, Grune J. Immune mediators in heart-lung communication. Pflugers Arch 2025; 477:17-30. [PMID: 39256247 PMCID: PMC11711577 DOI: 10.1007/s00424-024-03013-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 09/12/2024]
Abstract
It is often the case that serious, end-stage manifestations of disease result from secondary complications in organs distinct from the initial site of injury or infection. This is particularly true of diseases of the heart-lung axis, given the tight anatomical connections of the two organs within a common cavity in which they collectively orchestrate the two major, intertwined circulatory pathways. Immune cells and the soluble mediators they secrete serve as effective, and targetable, messengers of signals between different regions of the body but can also contribute to the spread of pathology. In this review, we discuss the immunological basis of interorgan communication between the heart and lung in various common diseases, and in the context of organ crosstalk more generally. Gaining a greater understanding of how the heart and lung communicate in health and disease, and viewing disease progression generally from a more holistic, whole-body viewpoint have the potential to inform new diagnostic approaches and strategies for better prevention and treatment of comorbidities.
Collapse
Affiliation(s)
- Jonathan L Gillan
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Lara Jaeschke
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Wolfgang M Kuebler
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Jana Grune
- Department of Cardiothoracic and Vascular Surgery, Deutsches Herzzentrum Der Charité (DHZC), Virchowweg 6, 10117, Berlin, Germany.
- Institute of Physiology, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| |
Collapse
|
5
|
Yang L, Xia B, Qian T, Wang J, Wang Y, Dai J, Le C, Yang X, Wu J, Wu W, Xu J, Liu Y, Wang J. hUC-MSCs mitigate atherosclerosis induced by a high-fat diet in ApoE -/- mice by regulating the intestinal microbiota. Heliyon 2024; 10:e38698. [PMID: 39559240 PMCID: PMC11570454 DOI: 10.1016/j.heliyon.2024.e38698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 11/20/2024] Open
Abstract
Background The mechanism underlying human umbilical cord mesenchymal stem cells (hUC-MSCs) regulating the stability of atherosclerotic plaque was explored by establishing mice models of atherosclerosis induced by a high-fat diet and hUC-MSCs intervention. Methods The ApoE-/- mice atherosclerosis model was constructed using a high-fat diet, and mice were divided into a normal diet group (ND), high-fat diet group (HFD), hUC-MSCs treatment group (HFDM), while the blank control (BC) consisted of C57BL/6J mice. After successful establishment of the model, the feces, hearts, and aorta of mice were collected. Morphological features were detected using HE, oil red O, and Masson staining. Afterward, 16s rRNA gene sequences was used to detect the species and abundance of the intestinal flora in mice, and an atomic force microscope (AFM) was used to detect the Young's modulus of the fibrous cap of atherosclerotic plaques. Lastly, the expression level of the inflammatory factors NLRP3, IL-1β, and IL-18 were detected via immunohistochemistry and immunofluorescence assays. Results In terms of morphological characteristics, the expression level of NLRP3, Young's modulus of the fibrous cap, and plaque stability were significantly reduced in HFD, whereas the ratio of Firmicutes to Bacteroidetes (F/B) was significantly increased. Interestingly, hUC-MSCs treatment reversed the above indices, thus enhancing plaque stability. Conclusion HFD led to dysregulation of intestinal flora homeostasis and induced aberrant expression levels of NLRP3, resulting in a decrease in the Young's modulus of plaques. However, hUC-MSCs treatment improved the biomechanical properties of plaque by modulating the intestinal flora and NLRP3, thereby elevating plaque stability and minimizing the risk of plaque rupture.
Collapse
Affiliation(s)
- Lin Yang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Bing Xia
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Tianbao Qian
- School of Biology and Engineering, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jie Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Yuanhe Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jialin Dai
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Cuiyun Le
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Xiaorong Yang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jun Wu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Wenxin Wu
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
| | - Jianwei Xu
- Center for Tissue Engineering and Stem Cell Research, Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Youbin Liu
- Cardiovascular Department of Ganzhou People's Hospital, Ganzhou, 341099, Jiangxi, China
| | - Jiawen Wang
- School of Forensic Medicine, Guizhou Medical University, Guiyang, 550000, Guizhou, China
- School of Basic Medicine/Institute of Forensic Medicine, Gannan Medical University, Ganzhou, 341000, Jiangxi, China
| |
Collapse
|
6
|
Ronen D, Rokach Y, Abedat S, Qadan A, Daana S, Amir O, Asleh R. Human Gut Microbiota in Cardiovascular Disease. Compr Physiol 2024; 14:5449-5490. [PMID: 39109979 DOI: 10.1002/cphy.c230012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
Abstract
The gut ecosystem, termed microbiota, is composed of bacteria, archaea, viruses, protozoa, and fungi and is estimated to outnumber human cells. Microbiota can affect the host by multiple mechanisms, including the synthesis of metabolites and toxins, modulating inflammation and interaction with other organisms. Advances in understanding commensal organisms' effect on human conditions have also elucidated the importance of this community for cardiovascular disease (CVD). This effect is driven by both direct CV effects and conditions known to increase CV risk, such as obesity, diabetes mellitus (DM), hypertension, and renal and liver diseases. Cardioactive metabolites, such as trimethylamine N -oxide (TMAO), short-chain fatty acids (SCFA), lipopolysaccharides, bile acids, and uremic toxins, can affect atherosclerosis, platelet activation, and inflammation, resulting in increased CV incidence. Interestingly, this interaction is bidirectional with microbiota affected by multiple host conditions including diet, bile acid secretion, and multiple diseases affecting the gut barrier. This interdependence makes manipulating microbiota an attractive option to reduce CV risk. Indeed, evolving data suggest that the benefits observed from low red meat and Mediterranean diet consumption can be explained, at least partially, by the changes that these diets may have on the gut microbiota. In this article, we depict the current epidemiological and mechanistic understanding of the role of microbiota and CVD. Finally, we discuss the potential therapeutic approaches aimed at manipulating gut microbiota to improve CV outcomes. © 2024 American Physiological Society. Compr Physiol 14:5449-5490, 2024.
Collapse
Affiliation(s)
- Daniel Ronen
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Yair Rokach
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Abed Qadan
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Samar Daana
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| |
Collapse
|
7
|
Beloglazov VA, Yatskov IA, Useinova RK. Low-grade inflammation in the post-COVID period as a strategic goal of treatment and rehabilitation. ACTA BIOMEDICA SCIENTIFICA 2024; 9:24-34. [DOI: 10.29413/abs.2024-9.2.3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2025] Open
Abstract
As of the beginning of 2023, there are more than 660 million convalescents of a new coronavirus infection in the world, however, even despite successful treatment of the acute period of the disease, such patients have a high risk of developing long-term complications in the post-COVID period, primarily cardiovascular events. One factor that seriously increases the risk of these complications is the state of lowgrade systemic inflammation (LGSI). LGSI is not a clinical diagnosis, it is characterized by a level of C-reactive protein in peripheral blood in the range of 3–10 mg/l and is most often detected during routine examination of patients, who in most cases have no clinical symptoms. In this regard, the condition of LGSI most often remains unnoticed and unreasonably ignored, despite quite extensive literature data on the effect of LGSI on the pathogenesis of many cardiovascular diseases. The development of drug therapy for LGSI is complicated by the multifactorial etiology of this condition. The causes of LGSI can be both genetic factors, which are practically impossible to correct, and conditions that are amenable to drug and non-drug treatment, such as, for example, increased intestinal permeability to pro-inflammatory agents, including lipopolysaccharide of gram-negative flora, the presence of a chronic untreated infection site and endocrine pathology (obesity and type 2 diabetes). This review presents the main information to date on the state of LGSI in patients who had a new coronavirus infection, including the results of our own observations of patients who have undergone a course of rehabilitation measures, as well as the most significant, in our opinion, factors predisposing to the development of LGSI in such patients.
Collapse
Affiliation(s)
- V. A. Beloglazov
- Medical Institute named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University
| | - I. A. Yatskov
- Medical Institute named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University
| | - R. Kh. Useinova
- Medical Institute named after S.I. Georgievsky, V.I. Vernadsky Crimean Federal University
| |
Collapse
|
8
|
Zheng S, Liu Z, Liu H, Lim JY, Li DWH, Zhang S, Luo F, Wang X, Sun C, Tang R, Zheng W, Xie Q. Research development on gut microbiota and vulnerable atherosclerotic plaque. Heliyon 2024; 10:e25186. [PMID: 38384514 PMCID: PMC10878880 DOI: 10.1016/j.heliyon.2024.e25186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 01/21/2024] [Accepted: 01/22/2024] [Indexed: 02/23/2024] Open
Abstract
The relationship between gut microbiota and its metabolites with cardiovascular disease (CVD) has been proven. In this review, we aim to conclude the potential mechanism of gut microbiota and its metabolites on inducing the formation of vulnerable atherosclerotic plaque, and to discuss the effect of intestinal metabolites, including trimethylamine-N-oxide (TMAO), lipopolysaccharide (LPS), phenylacetylglutamine (PAG), short-chain fatty acids (SCFAs) on plaque stability. Finally, we include the impact of gut microbiota and its metabolites on plaque stability, to propose a new therapeutic direction for coronary heart disease. Gut microbiota regulation intervenes the progress of arteriosclerosis, especially on coronary atherosclerosis, by avoiding or reducing the formation of vulnerable plaque, to lower the morbidity rate of myocardial infarction.
Collapse
Affiliation(s)
- Shujiao Zheng
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| | - Zuheng Liu
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Haiyue Liu
- Xiamen Key Laboratory of Genetic Testing, The Department of Laboratory Medicine, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Jie Ying Lim
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Dolly Wong Hui Li
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Shaofeng Zhang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Fang Luo
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiujing Wang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Changqing Sun
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Rong Tang
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Wuyang Zheng
- Department of Cardiology, Xiamen Key Laboratory of Cardiac Electrophysiology, Xiamen Institute of Cardiovascular Diseases, The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qiang Xie
- The School of Clinical Medicine, Fujian Medical University, Fuzhou, China
| |
Collapse
|
9
|
Montone RA, Camilli M, Calvieri C, Magnani G, Bonanni A, Bhatt DL, Rajagopalan S, Crea F, Niccoli G. Exposome in ischaemic heart disease: beyond traditional risk factors. Eur Heart J 2024; 45:419-438. [PMID: 38238478 PMCID: PMC10849374 DOI: 10.1093/eurheartj/ehae001] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 12/22/2023] [Accepted: 01/03/2024] [Indexed: 02/09/2024] Open
Abstract
Ischaemic heart disease represents the leading cause of morbidity and mortality, typically induced by the detrimental effects of risk factors on the cardiovascular system. Although preventive interventions tackling conventional risk factors have helped to reduce the incidence of ischaemic heart disease, it remains a major cause of death worldwide. Thus, attention is now shifting to non-traditional risk factors in the built, natural, and social environments that collectively contribute substantially to the disease burden and perpetuate residual risk. Of importance, these complex factors interact non-linearly and in unpredictable ways to often enhance the detrimental effects attributable to a single or collection of these factors. For this reason, a new paradigm called the 'exposome' has recently been introduced by epidemiologists in order to define the totality of exposure to these new risk factors. The purpose of this review is to outline how these emerging risk factors may interact and contribute to the occurrence of ischaemic heart disease, with a particular attention on the impact of long-term exposure to different environmental pollutants, socioeconomic and psychological factors, along with infectious diseases such as influenza and COVID-19. Moreover, potential mitigation strategies for both individuals and communities will be discussed.
Collapse
Affiliation(s)
- Rocco A Montone
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | | | - Giulia Magnani
- Department of Medicine, University of Parma, Parma, Italy
| | - Alice Bonanni
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
| | - Deepak L Bhatt
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sanjay Rajagopalan
- Cardiovascular Research Institute, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Filippo Crea
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168 Rome, Italy
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | | |
Collapse
|
10
|
Byun DJ, Lee J, Yu JW, Hyun YM. NLRP3 Exacerbate NETosis-Associated Neuroinflammation in an LPS-Induced Inflamed Brain. Immune Netw 2023; 23:e27. [PMID: 37416934 PMCID: PMC10320420 DOI: 10.4110/in.2023.23.e27] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/27/2023] [Accepted: 04/15/2023] [Indexed: 07/08/2023] Open
Abstract
Neutrophil extracellular traps (NETs) exert a novel function of trapping pathogens. Released NETs can accumulate in inflamed tissues, be recognized by other immune cells for clearance, and lead to tissue toxicity. Therefore, the deleterious effect of NET is an etiological factor, causing several diseases directly or indirectly. NLR family pyrin domain containing 3 (NLRP3) in neutrophils is pivotal in signaling the innate immune response and is associated with several NET-related diseases. Despite these observations, the role of NLRP3 in NET formation in neuroinflammation remains elusive. Therefore, we aimed to explore NET formation promoted by NLRP3 in an LPS-induced inflamed brain. Wild-type and NLRP3 knockout mice were used to investigate the role of NLRP3 in NET formation. Brain inflammation was systemically induced by administering LPS. In such an environment, the NET formation was evaluated based on the expression of its characteristic indicators. DNA leakage and NET formation were analyzed in both mice through Western blot, flow cytometry, and in vitro live cell imaging as well as two-photon imaging. Our data revealed that NLRP3 promotes DNA leakage and facilitates NET formation accompanied by neutrophil death. Moreover, NLRP3 is not involved in neutrophil infiltration but is predisposed to boost NET formation, which is accompanied by neutrophil death in the LPS-induced inflamed brain. Furthermore, either NLRP3 deficiency or neutrophil depletion diminished pro-inflammatory cytokine, IL-1β, and alleviated blood-brain barrier damage. Overall, the results suggest that NLRP3 exacerbates NETosis in vitro and in the inflamed brain, aggravating neuroinflammation. These findings provide a clue that NLRP3 would be a potential therapeutic target to alleviate neuroinflammation.
Collapse
Affiliation(s)
- Da Jeong Byun
- Department of Anatomy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Jaeho Lee
- Department of Anatomy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Je-Wook Yu
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| | - Young-Min Hyun
- Department of Anatomy, Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Korea
| |
Collapse
|
11
|
Zhang Q, Zhang L, Chen C, Li P, Lu B. The gut microbiota-artery axis: A bridge between dietary lipids and atherosclerosis? Prog Lipid Res 2023; 89:101209. [PMID: 36473673 DOI: 10.1016/j.plipres.2022.101209] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/30/2022] [Accepted: 12/02/2022] [Indexed: 12/09/2022]
Abstract
Atherosclerotic cardiovascular disease is one of the major leading global causes of death. Growing evidence has demonstrated that gut microbiota (GM) and its metabolites play a pivotal role in the onset and progression of atherosclerosis (AS), now known as GM-artery axis. There are interactions between dietary lipids and GM, which ultimately affect GM and its metabolites. Given these two aspects, the GM-artery axis may play a mediating role between dietary lipids and AS. Diets rich in saturated fatty acids (SFAs), omega-6 polyunsaturated fatty acids (n-6 PUFAs), industrial trans fatty acids (TFAs), and cholesterol can increase the levels of atherogenic microbes and metabolites, whereas monounsaturated fatty acids (MUFAs), ruminant TFAs, and phytosterols (PS) can increase the levels of antiatherogenic microbes and metabolites. Actually, dietary phosphatidylcholine (PC), sphingomyelin (SM), and omega-3 polyunsaturated fatty acids (n-3 PUFAs) have been demonstrated to affect AS via the GM-artery axis. Therefore, that GM-artery axis acts as a communication bridge between dietary lipids and AS. Herein, we will describe the molecular mechanism of GM-artery axis in AS and discuss the complex interactions between dietary lipids and GM. In particular, we will highlight the evidence and potential mechanisms of dietary lipids affecting AS via GM-artery axis.
Collapse
Affiliation(s)
- Qinjun Zhang
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China
| | - Liangxiao Zhang
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Cheng Chen
- Center for Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peiwu Li
- Oil Crops Research Institute, Chinese Academy of Agricultural Sciences, Wubhan, China
| | - Baiyi Lu
- College of Biosystems Engineering and Food Science, National-Local Joint Engineering Laboratory of Intelligent Food Technology and Equipment, Key Laboratory for Agro-Products Nutritional Evaluation of Ministry of Agriculture and Rural Affairs, Key Laboratory of Agro-Products Postharvest Handling of Ministry of Agriculture and Rural Affairs, Zhejiang Key Laboratory for Agro-Food Processing, Zhejiang International Scientific and Technological Cooperation Base of Health Food Manufacturing and Quality Control, Zhejiang University, Hangzhou, China; Hangzhou Global Scientific and Technological Innovation Center, Zhejiang University, Hangzhou, China.
| |
Collapse
|
12
|
Abstract
Systemic inflammation has been suggested to have a pivotal role in atherothrombosis, but the factors that trigger systemic inflammation have not been fully elucidated. Lipopolysaccharide (LPS) is a component of the membrane of Gram-negative bacteria present in the gut that can translocate into the systemic circulation, causing non-septic, low-grade endotoxaemia. Gut dysbiosis is a major determinant of low-grade endotoxaemia via dysfunction of the intestinal barrier scaffold, which is a prerequisite for LPS translocation into the systemic circulation. Experimental studies have demonstrated that LPS is present in atherosclerotic arteries but not in normal arteries. In atherosclerotic plaques, LPS promotes a pro-inflammatory status that can lead to plaque instability and thrombus formation. Low-grade endotoxaemia affects several cell types, including leukocytes, platelets and endothelial cells, leading to inflammation and clot formation. Low-grade endotoxaemia has been described in patients at risk of or with overt cardiovascular disease, in whom low-grade endotoxaemia was associated with atherosclerotic burden and its clinical sequelae. In this Review, we describe the mechanisms favouring the development of low-grade endotoxaemia, focusing on gut dysbiosis and changes in gut permeability; the plausible biological mechanisms linking low-grade endotoxaemia and atherothrombosis; the clinical studies suggesting that low-grade endotoxaemia is a risk factor for cardiovascular events; and the potential therapeutic tools to improve gut permeability and eventually eliminate low-grade endotoxaemia.
Collapse
|
13
|
Nabeh OA. Gut microbiota and cardiac arrhythmia: a pharmacokinetic scope. Egypt Heart J 2022; 74:87. [PMID: 36583819 PMCID: PMC9803803 DOI: 10.1186/s43044-022-00325-2] [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: 09/22/2022] [Accepted: 12/27/2022] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Dealing with cardiac arrhythmia is a difficult challenge. Choosing between different anti-arrhythmic drugs (AADs) while being cautious about the pro-arrhythmic characteristics of some of these drugs and their diverse interaction with other drugs is a real obstacle. MAIN BODY Gut microbiota (GM), in our bodies, are now being considered as a hidden organ which can regulate our immune system, digest complex food, and secrete bioactive compounds. Yet, GM are encountered in the pathophysiology of arrhythmia and can affect the pharmacokinetics of AADs, as well as some anti-thrombotics, resulting in altering their bioavailability, therapeutic function and may predispose to some of their unpleasant adverse effects. CONCLUSIONS Knowledge of the exact role of GM in the pharmacokinetics of these drugs is now essential for better understanding of the art of arrhythmia management. Also, it will help deciding when to consider probiotics as an adjunctive therapy while treating arrhythmia. This should be discovered in the near future.
Collapse
Affiliation(s)
- Omnia Azmy Nabeh
- grid.7776.10000 0004 0639 9286Department of Medical Pharmacology, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
14
|
Serum Lipopolysaccharide Is Associated with the Recurrence of Atrial Fibrillation after Radiofrequency Ablation by Increasing Systemic Inflammation and Atrial Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:2405972. [PMID: 36285296 PMCID: PMC9588364 DOI: 10.1155/2022/2405972] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/13/2022] [Accepted: 09/28/2022] [Indexed: 11/30/2022]
Abstract
Objectives The gut microbiota and its metabolites are linked to inflammation and contribute to the progression of atrial fibrillation (AF), but the predictive value of the gut microbiota-derived metabolite lipopolysaccharide (LPS) for AF recurrence (RAF) is unknown. This study is aimed at investigating (1) the correlation between LPS and RAF and (2) its relationship with inflammation and atrial fibrosis. Method We performed a single-centre retrospective analysis in 159 AF patients. Fasting plasma samples were collected, and an enzyme-linked immunosorbent assay was used to determine the levels of serum LPS, interleukin-6 (IL-6), collagen type-1 C-terminal telopeptide (CITP), and transforming growth factor-β1 (TGFβ1). The cumulative risk for RAF was evaluated with Kaplan–Meier analysis. Cox proportional hazard analysis was carried out to predict the hazard of RAF. The correlations among LPS and IL-6, CITP, TGFβ1, and left atrial diameter (LAD) were analysed by Pearson's correlation coefficient. Subsequent univariate and multivariable linear regression analyses were carried out to evaluate the connection between clinical variables and Log-LPS. Results All 159 AF patients were included in this study. The proportion of persistent atrial fibrillation was 40.3%, the mean age was 61.9 ± 10.1 years, the proportion of males was 61.6%, and the mean LPS was 56.5 ± 29.5 pg/mL. After all patients were divided into tertiles according to the circulating LPS level, a total of 44 RAF occurred: 10 in the first tertile, 15 in the second tertile, and 19 in the third tertile (log-rank test P = 0.037). Heart failure (hazard ratio 2.029, P = 0.041), LAD (hazard ratio 1.064, P = 0.022), Log-LPS (hazard ratio 5.686, P = 0.043), and CITP (hazard ratio 6.841, P = 0.033) independently predicted the risk of RAF. In all patients, univariate analysis showed that heart failure, LAD, hs-CRP, IL-6, CITP, and TGF-β1 were connected with Log-LPS. Multivariate linear regression analysis indicated that IL-6 and hs-CRP were independently and positively connected with Log-LPS. Conclusions Our results indicated that circulating LPS was a predictor of RAF and may contribute to RAF incidence after ablation by increasing systemic inflammation and atrial fibrosis.
Collapse
|
15
|
Pletz MW, Jensen AV, Bahrs C, Davenport C, Rupp J, Witzenrath M, Barten-Neiner G, Kolditz M, Dettmer S, Chalmers JD, Stolz D, Suttorp N, Aliberti S, Kuebler WM, Rohde G. Unmet needs in pneumonia research: a comprehensive approach by the CAPNETZ study group. Respir Res 2022; 23:239. [PMID: 36088316 PMCID: PMC9463667 DOI: 10.1186/s12931-022-02117-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 07/15/2022] [Indexed: 11/10/2022] Open
Abstract
Abstract
Introduction
Despite improvements in medical science and public health, mortality of community-acquired pneumonia (CAP) has barely changed throughout the last 15 years. The current SARS-CoV-2 pandemic has once again highlighted the central importance of acute respiratory infections to human health. The “network of excellence on Community Acquired Pneumonia” (CAPNETZ) hosts the most comprehensive CAP database worldwide including more than 12,000 patients. CAPNETZ connects physicians, microbiologists, virologists, epidemiologists, and computer scientists throughout Europe. Our aim was to summarize the current situation in CAP research and identify the most pressing unmet needs in CAP research.
Methods
To identify areas of future CAP research, CAPNETZ followed a multiple-step procedure. First, research members of CAPNETZ were individually asked to identify unmet needs. Second, the top 100 experts in the field of CAP research were asked for their insights about the unmet needs in CAP (Delphi approach). Third, internal and external experts discussed unmet needs in CAP at a scientific retreat.
Results
Eleven topics for future CAP research were identified: detection of causative pathogens, next generation sequencing for antimicrobial treatment guidance, imaging diagnostics, biomarkers, risk stratification, antiviral and antibiotic treatment, adjunctive therapy, vaccines and prevention, systemic and local immune response, comorbidities, and long-term cardio-vascular complications.
Conclusion
Pneumonia is a complex disease where the interplay between pathogens, immune system and comorbidities not only impose an immediate risk of mortality but also affect the patients’ risk of developing comorbidities as well as mortality for up to a decade after pneumonia has resolved. Our review of unmet needs in CAP research has shown that there are still major shortcomings in our knowledge of CAP.
Collapse
|
16
|
Gawałko M, Agbaedeng TA, Saljic A, Müller DN, Wilck N, Schnabel R, Penders J, Rienstra M, van Gelder I, Jespersen T, Schotten U, Crijns HJGM, Kalman JM, Sanders P, Nattel S, Dobrev D, Linz D. Gut microbiota, dysbiosis and atrial fibrillation. Arrhythmogenic mechanisms and potential clinical implications. Cardiovasc Res 2022; 118:2415-2427. [PMID: 34550344 PMCID: PMC9400433 DOI: 10.1093/cvr/cvab292] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/08/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
Recent preclinical and observational cohort studies have implicated imbalances in gut microbiota composition as a contributor to atrial fibrillation (AF). The gut microbiota is a complex and dynamic ecosystem containing trillions of microorganisms, which produces bioactive metabolites influencing host health and disease development. In addition to host-specific determinants, lifestyle-related factors such as diet and drugs are important determinants of the gut microbiota composition. In this review, we discuss the evidence suggesting a potential bidirectional association between AF and gut microbiota, identifying gut microbiota-derived metabolites as possible regulators of the AF substrate. We summarize the effect of gut microbiota on the development and progression of AF risk factors, including heart failure, hypertension, obesity, and coronary artery disease. We also discuss the potential anti-arrhythmic effects of pharmacological and diet-induced modifications of gut microbiota composition, which may modulate and prevent the progression to AF. Finally, we highlight important gaps in knowledge and areas requiring future investigation. Although data supporting a direct relationship between gut microbiota and AF are very limited at the present time, emerging preclinical and clinical research dealing with mechanistic interactions between gut microbiota and AF is important as it may lead to new insights into AF pathophysiology and the discovery of novel therapeutic targets for AF.
Collapse
Affiliation(s)
- Monika Gawałko
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Duisburg, Germany
- Department of Cardiology, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas A Agbaedeng
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Arnela Saljic
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Dominik N Müller
- Experimental and Clinical Research Center, Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
| | - Nicola Wilck
- Experimental and Clinical Research Center, Cooperation of Charité-Universitätsmedizin Berlin and Max Delbrück Centre for Molecular Medicine, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Max Delbrück Centre for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- Medizinische Klinik mit Schwerpunkt Nephrologie und Internistische Intensivmedizin, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Renate Schnabel
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Department of General and Interventional Cardiology, University Heart Center Hamburg, Hamburg, Germany
| | - John Penders
- Department of Medical Microbiology, Care and Public Health Research Institute (CAPHRI), School for Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Centre, Maastricht, The Netherlands
| | - Michiel Rienstra
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Isabelle van Gelder
- Department of Cardiology, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Thomas Jespersen
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulrich Schotten
- Department of Physiology, University Maastricht, Maastricht, The Netherlands
| | - Harry J G M Crijns
- Department of Cardiology, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
| | - Jonathan M Kalman
- Department of Cardiology, Royal Melbourne Hospital, Melbourne, Australia
- Department of Medicine, University of Melbourne, Melbourne, Australia
| | - Prashanthan Sanders
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
| | - Stanley Nattel
- Department of Pharmacology, Medicine and Research Centre, Montréal Heart Institute, University de Montréal, McGill University, Montréal, QC, Canada
- IHU LIRYC and Fondation Bordeaux Université, Bordeaux, France
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Centre, University Duisburg-Essen, Duisburg, Germany
- Department of Pharmacology, Medicine and Research Centre, Montréal Heart Institute, University de Montréal, McGill University, Montréal, QC, Canada
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Dominik Linz
- Department of Cardiology, Maastricht University Medical Centre, Cardiovascular Research Institute Maastricht, Universiteitssingel 50, 6229 ER Maastricht, The Netherlands
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Centre for Heart Rhythm Disorders, Royal Adelaide Hospital, University of Adelaide, Adelaide, Australia
- Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| |
Collapse
|
17
|
Pussinen PJ, Kopra E, Pietiäinen M, Lehto M, Zaric S, Paju S, Salminen A. Periodontitis and cardiometabolic disorders: The role of lipopolysaccharide and endotoxemia. Periodontol 2000 2022; 89:19-40. [PMID: 35244966 PMCID: PMC9314839 DOI: 10.1111/prd.12433] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Lipopolysaccharide is a virulence factor of gram-negative bacteria with a crucial importance to the bacterial surface integrity. From the host's perspective, lipopolysaccharide plays a role in both local and systemic inflammation, activates both innate and adaptive immunity, and can trigger inflammation either directly (as a microbe-associated molecular pattern) or indirectly (by inducing the generation of nonmicrobial, danger-associated molecular patterns). Translocation of lipopolysaccharide into the circulation causes endotoxemia, which is typically measured as the biological activity of lipopolysaccharide to induce coagulation of an aqueous extract of blood cells of the assay. Apparently healthy subjects have a low circulating lipopolysaccharide activity, since it is neutralized and cleared rapidly. However, chronic endotoxemia is involved in the pathogenesis of many inflammation-driven conditions, especially cardiometabolic disorders. These include atherosclerotic cardiovascular diseases, obesity, liver diseases, diabetes, and metabolic syndrome, where endotoxemia has been recognized as a risk factor. The main source of endotoxemia is thought to be the gut microbiota. However, the oral dysbiosis in periodontitis, which is typically enriched with gram-negative bacterial species, may also contribute to endotoxemia. As endotoxemia is associated with an increased risk of cardiometabolic disorders, lipopolysaccharide could be considered as a molecular link between periodontal microbiota and cardiometabolic diseases.
Collapse
Affiliation(s)
- Pirkko J Pussinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Elisa Kopra
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Milla Pietiäinen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Markku Lehto
- Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland.,Abdominal Center, Nephrology, University of Helsinki and Helsinki University Hospital, Helsinki, Finland.,Clinical and Molecular Metabolism, Faculty of Medicine Research Programs, University of Helsinki, Helsinki, Finland
| | - Svetislav Zaric
- Faculty of Dentistry, Oral & Craniofacial Sciences, Kings College London, London, UK
| | - Susanna Paju
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Aino Salminen
- Oral and Maxillofacial Diseases, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
18
|
Shen X, Li L, Sun Z, Zang G, Zhang L, Shao C, Wang Z. Gut Microbiota and Atherosclerosis-Focusing on the Plaque Stability. Front Cardiovasc Med 2021; 8:668532. [PMID: 34414217 PMCID: PMC8368126 DOI: 10.3389/fcvm.2021.668532] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are major causes of mortality and morbidity in the modern society. The rupture of atherosclerotic plaque can induce thrombus formation, which is the main cause of acute cardiovascular events. Recently, many studies have demonstrated that there are some relationships between microbiota and atherosclerosis. In this review, we will focus on the effect of the microbiota and the microbe-derived metabolites, including trimethylamine-N-oxide (TMAO), short-chain fatty acids (SCFAs), and lipopolysaccharide (LPS), on the stability of atherosclerotic plaque. Finally, we will conclude with some therapies based on the microbiota and its metabolites.
Collapse
Affiliation(s)
- Xinyi Shen
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Guangyao Zang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Lili Zhang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Chen Shao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| |
Collapse
|
19
|
Abstract
Pneumonia is a common acute respiratory infection that affects the alveoli and distal airways; it is a major health problem and associated with high morbidity and short-term and long-term mortality in all age groups worldwide. Pneumonia is broadly divided into community-acquired pneumonia or hospital-acquired pneumonia. A large variety of microorganisms can cause pneumonia, including bacteria, respiratory viruses and fungi, and there are great geographical variations in their prevalence. Pneumonia occurs more commonly in susceptible individuals, including children of <5 years of age and older adults with prior chronic conditions. Development of the disease largely depends on the host immune response, with pathogen characteristics having a less prominent role. Individuals with pneumonia often present with respiratory and systemic symptoms, and diagnosis is based on both clinical presentation and radiological findings. It is crucial to identify the causative pathogens, as delayed and inadequate antimicrobial therapy can lead to poor outcomes. New antibiotic and non-antibiotic therapies, in addition to rapid and accurate diagnostic tests that can detect pathogens and antibiotic resistance will improve the management of pneumonia.
Collapse
|
20
|
Cinar R, Iyer MR, Kunos G. Dual inhibition of CB 1 receptors and iNOS, as a potential novel approach to the pharmacological management of acute and long COVID-19. Br J Pharmacol 2021; 179:2121-2127. [PMID: 33769552 PMCID: PMC8251289 DOI: 10.1111/bph.15461] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 03/15/2021] [Accepted: 03/21/2021] [Indexed: 12/14/2022] Open
Abstract
COVID‐19 (SARS‐CoV‐2) causes multiple inflammatory complications, resulting not only in severe lung inflammation but also harm to other organs. Although the current focus is on the management of acute COVID‐19, there is growing concern about long‐term effects of COVID‐19 (Long Covid), such as fibroproliferative changes in the lung, heart and kidney. Therefore, the identification of therapeutic targets not only for the management of acute COVID‐19 but also for preventing Long Covid are needed, and would mitigate against long‐lasting health burden and economic costs, in addition to saving lives. COVID‐19 induces pathological changes via multiple pathways, which could be targeted simultaneously for optimal effect. We discuss the potential pathologic function of increased activity of the endocannabinoid/CB1 receptor system and inducible NO synthase (iNOS). We advocate a polypharmacology approach, wherein a single chemical entity simultaneously interacts with CB1 receptors and iNOS causing inhibition, as a potential therapeutic strategy for COVID‐19‐related health complications.
Collapse
Affiliation(s)
- Resat Cinar
- Section on Fibrotic Disorders, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - Malliga R Iyer
- Section on Medicinal Chemistry, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland, USA
| | - George Kunos
- Laboratory of Physiologic Studies, National Institute on Alcohol Abuse and Alcoholism (NIAAA), National Institutes of Health (NIH), Bethesda, Maryland, USA
| |
Collapse
|
21
|
Khoshkhouy F, Farshbaf A, Mahmoudabady M, Gholamnezhad Z. Effects of moderate exercise on lipopolysaccharide-induced inflammatory responses in rat's cardiac tissue. Cytokine 2020; 138:155409. [PMID: 33360764 DOI: 10.1016/j.cyto.2020.155409] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 12/13/2020] [Accepted: 12/14/2020] [Indexed: 01/03/2023]
Abstract
The effects of moderate exercise on cardiac tissue inflammation, oxidative stress markers and apoptosis in lipopolysaccharide (LPS)-administered rats were evaluated. Wistar rats were divided into three groups (N = 8): (1) control; (2) LPS (1 mg/kg); and (3) LPS + moderate training (LPS + EX: 15 m/min, 30 min/day, for 9 weeks (week 1-9)). LPS was injected intraperitoneally for 5 days during week 9. Finally, the rats' heart were removed for biochemical and expression assessments. LPS increased the levels of tumor necrosis factor α (TNF-α), interleukin (IL)- 1β, C-reactive protein (CRP), malondialdehyde (MDA) and nitric oxide (NO) metabolites in cardiac tissue, but decreased thiol contents and catalase (CAT) and superoxide dismutase (SOD) activity in cardiac tissue compared to the control group (p < 0.05-p < 0.001). In LPS + EX group, the level of NO metabolites was increased (p < 0.05) and thiol contents were decreased (p < 0.001) compared to the control group. Moderate training decreased the levels of TNF-α, IL-1β, CRP and NO metabolites while increased CAT activity in the LPS + EX group compared to the LPS group (p < 0.05-p < 0.001). The mRNA level of BAX in the LPS group and the BCL2/BAX ratio in both LPS and LPS + EX groups increased compared to the control group (p < 0.05-p < 0.01). These results indicated that moderate training improved LPS-induced deleterious effects on cardiac tissue by attenuating proinflammatory cytokine levels, apoptosis and oxidative damage.
Collapse
Affiliation(s)
- Fatemeh Khoshkhouy
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Alieh Farshbaf
- Dental Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Mahmoudabady
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Gholamnezhad
- Department of Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
22
|
Piscitelli F, Silvestri C. Role of the Endocannabinoidome in Human and Mouse Atherosclerosis. Curr Pharm Des 2020; 25:3147-3164. [PMID: 31448709 DOI: 10.2174/1381612825666190826162735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022]
Abstract
The Endocannabinoid (eCB) system and its role in many physiological and pathological conditions is well described and accepted, and includes cardiovascular disorders. However, the eCB system has been expanded to an "-ome"; the endocannabinoidome (eCBome) that includes endocannabinoid-related mediators, their protein targets and metabolic enzymes, many of which significantly impact upon cardiometabolic health. These recent discoveries are here summarized with a special focus on their potential involvement in atherosclerosis. We described the role of classical components of the eCB system (eCBs, CB1 and CB2 receptors) and eCB-related lipids, their regulatory enzymes and molecular targets in atherosclerosis. Furthermore, since increasing evidence points to significant cross-talk between the eCBome and the gut microbiome and the gut microbiome and atherosclerosis, we explore the possibility that a gut microbiome - eCBome axis has potential implications in atherosclerosis.
Collapse
Affiliation(s)
- Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Council of Research, Pozzuoli (NA), Italy
| | - Cristoforo Silvestri
- Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada.,Department of Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
23
|
Sin DD. Contemporary Concise Review 2019: Chronic obstructive pulmonary disease. Respirology 2020; 25:449-454. [PMID: 32040982 DOI: 10.1111/resp.13782] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 01/29/2020] [Indexed: 12/14/2022]
Affiliation(s)
- Don D Sin
- UBC Centre for Heart and Lung Innovation, St. Paul's Hospital, Vancouver, BC, Canada.,Division of Respiratory Medicine (Department of Medicine), University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
24
|
Bhatt SP. Acute Exacerbations of Chronic Lung Disease: Cardiac Considerations. CARDIAC CONSIDERATIONS IN CHRONIC LUNG DISEASE 2020. [PMCID: PMC7282481 DOI: 10.1007/978-3-030-43435-9_12] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
The importance of appropriately recognizing and managing patients with cardiovascular and pulmonary comorbidities is underscored by the poor outcomes described in complex comorbid patients. Patients with chronic obstructive pulmonary disease (COPD) have an increased risk, up to one-third greater than the general population, of cardiovascular comorbidities including hypertension and diabetes [1].
Collapse
Affiliation(s)
- Surya P. Bhatt
- Division of Pulmonary, Allergy and Critical Care Medicine, University of Alabama at Birmingham, Birmingham, AL USA
| |
Collapse
|
25
|
Hurst JR, Sin DD. Chronic Obstructive Pulmonary Disease as a Risk Factor for Cardiovascular Disease. A View from the SUMMIT. Am J Respir Crit Care Med 2019. [PMID: 29533679 DOI: 10.1164/rccm.201802-0347ed] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- John R Hurst
- 1 UCL Respiratory Medicine University College London London, United Kingdom
| | - Don D Sin
- 2 Centre for Heart Lung Innovation St. Paul's Hospital Vancouver, British Columbia, Canada and.,3 Division of Respiratory Medicine University of British Columbia Vancouver, British Columbia, Canada
| |
Collapse
|
26
|
Crisan L, Wong N, Sin DD, Lee HM. Karma of Cardiovascular Disease Risk Factors for Prevention and Management of Major Cardiovascular Events in the Context of Acute Exacerbations of Chronic Obstructive Pulmonary Disease. Front Cardiovasc Med 2019; 6:79. [PMID: 31294030 PMCID: PMC6603127 DOI: 10.3389/fcvm.2019.00079] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
There is compelling epidemiological evidence that airway exposure to cigarette smoke, air pollution particles, as well as bacterial and viral pathogens is strongly related to acute ischemic events. Over the years, there have been important animal and human studies that have provided experimental evidence to support a causal link. Studies show that patients with cardiovascular diseases (CVDs) or risk factors for CVD are more likely to have major adverse cardiovascular events (MACEs) after an acute exacerbation of chronic obstructive pulmonary disease (COPD), and patients with more severe COPD have higher cardiovascular mortality and morbidity than those with less severe COPD. The risk of MACEs in acute exacerbation of COPD is determined by the complex interactions between genetics, behavioral, metabolic, infectious, and environmental risk factors. To date, there are no guidelines regarding the prevention, screening, and management of the modifiable risk factors for MACEs in the context of COPD or COPD exacerbations, and there is insufficient CVD risk control in those with COPD. A deeper insight of the modifiable risk factors shared by CVD, COPD, and acute exacerbations of COPD may improve the strategies for reduction of MACEs in patients with COPD through vaccination, tight control of traditional CV risk factors and modifying lifestyle. This review summarizes the most recent studies regarding the pathophysiology and epidemiology of modifiable risk factors shared by CVD, COPD, and COPD exacerbations that could influence overall morbidity and mortality due to MACEs in patients with acute exacerbations of COPD.
Collapse
Affiliation(s)
- Liliana Crisan
- Heart Disease Prevention Program, Division of Cardiology, University of California, Irvine, Irvine, CA, United States
| | - Nathan Wong
- Heart Disease Prevention Program, Division of Cardiology, University of California, Irvine, Irvine, CA, United States
| | - Don D. Sin
- Division of Respiratory Medicine, Department of Medicine, University of British Columbia and Centre for Heart Lung Innovation, Vancouver, BC, Canada
| | - Hwa Mu Lee
- Heart Disease Prevention Program, Division of Cardiology, University of California, Irvine, Irvine, CA, United States
- Division of Pulmonary and Critical Care Medicine, University of California, Irvine, Irvine, CA, United States
| |
Collapse
|
27
|
|
28
|
Mawhin MA, Tilly P, Zirka G, Charles AL, Slimani F, Vonesch JL, Michel JB, Bäck M, Norel X, Fabre JE. Neutrophils recruited by leukotriene B4 induce features of plaque destabilization during endotoxaemia. Cardiovasc Res 2018; 114:1656-1666. [DOI: 10.1093/cvr/cvy130] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/17/2018] [Indexed: 12/22/2022] Open
Affiliation(s)
- Marie-Anne Mawhin
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
- IGMBC, Illkirch, France
- UMR 7104 CNRS, Illkirch, France
- U964 INSERM, Illkirch, France
- Strasbourg University, Strasbourg, France
| | - Peggy Tilly
- IGMBC, Illkirch, France
- UMR 7104 CNRS, Illkirch, France
- U964 INSERM, Illkirch, France
- Strasbourg University, Strasbourg, France
| | - Gaia Zirka
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
| | - Anne-Laure Charles
- Equipe d'accueil 3072, Faculty of Medicine, Translational Medicine Federation, Strasbourg University, Strasbourg, France
| | - Farid Slimani
- IGMBC, Illkirch, France
- UMR 7104 CNRS, Illkirch, France
- U964 INSERM, Illkirch, France
- Strasbourg University, Strasbourg, France
| | | | | | - Magnus Bäck
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
- INSERM U1116, University of Lorraine and CHRU, Nancy, France
| | - Xavier Norel
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
| | - Jean-Etienne Fabre
- UMR 1148 INSERM, Xavier Bichat Hospital, 46 rue Henri Huchard, Paris, France
| |
Collapse
|
29
|
Leitao Filho FS, Sin DD. COPD and cardiovascular diseases: now is the time for action! Thorax 2018; 73:799-800. [PMID: 29622696 DOI: 10.1136/thoraxjnl-2018-211553] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2018] [Indexed: 01/09/2023]
Affiliation(s)
- Fernando Sergio Leitao Filho
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada.,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Don D Sin
- Centre for Heart Lung Innovation, St. Paul's Hospital, Vancouver, British Columbia, Canada.,Division of Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| |
Collapse
|
30
|
Sigala F, Oikonomou E, Antonopoulos AS, Galyfos G, Tousoulis D. Coronary versus carotid artery plaques. Similarities and differences regarding biomarkers morphology and prognosis. Curr Opin Pharmacol 2018; 39:9-18. [PMID: 29202375 DOI: 10.1016/j.coph.2017.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 11/14/2017] [Accepted: 11/20/2017] [Indexed: 01/09/2023]
Abstract
Carotid and coronary artery disease are two major atherosclerotic conditions that have shown an increased prevalence in the last three decades that is associated with high morbidity and mortality. Recent data have revealed that the development of the atherosclerotic plaque-the basic entity in both diseases-may share similar characteristics and mechanisms irrespective of the location site. Even though the biology of atherosclerotic process is similar, there are differences in plaque morphology and characteristics. Indeed, plaque erosion, calcified nodules, fibrous cap thickness and macrophage accumulation may be different in the setting of coronary and carotid artery disease. The perivascular adipose tissue surrounding the coronary arteries (but not carotids) could also affect plaque biology. In this review we focus on comparative the characteristics of both types of atherosclerotic plaques and summarize existing knowledge to provide useful conclusions about current and future treatment strategies.
Collapse
Affiliation(s)
- Fragiska Sigala
- 1st Department of Propaedeutic Surgery, University of Athens Medical School, Hippocration Hospital, Athens, Greece
| | - Evangelos Oikonomou
- 1st Department of Propaedeutic Surgery, University of Athens Medical School, Hippocration Hospital, Athens, Greece.
| | - Alexis S Antonopoulos
- 1st Cardiology Clinic, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - George Galyfos
- 1st Department of Propaedeutic Surgery, University of Athens Medical School, Hippocration Hospital, Athens, Greece
| | - Dimitris Tousoulis
- 1st Cardiology Clinic, 'Hippokration' Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| |
Collapse
|
31
|
Carnevale R, Nocella C, Petrozza V, Cammisotto V, Pacini L, Sorrentino V, Martinelli O, Irace L, Sciarretta S, Frati G, Pastori D, Violi F. Localization of lipopolysaccharide from Escherichia Coli into human atherosclerotic plaque. Sci Rep 2018; 8:3598. [PMID: 29483584 PMCID: PMC5826929 DOI: 10.1038/s41598-018-22076-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Accepted: 02/16/2018] [Indexed: 12/28/2022] Open
Abstract
Experimental studies showed that gut-derived lipopolysaccharide (LPS) is pro-atherogenic, however, its relationship with human atherosclerosis is still to be defined. We investigate if gut-derived LPS from Escherichia Coli localizes in human carotid plaque and its potential role as pro-inflammatory molecule in the atherosclerotic lesion. LPS from Escherichia Coli and Toll-like receptor 4 (TLR4) were studied in specimens from carotid and thyroid arteries of 10 patients undergoing endarterectomy and 15 controls matched for demographic and clinical characteristics. Blood LPS were significantly higher in patients compared to controls. Immunochemistry analysis revealed positivity for antibodies against LPS and TLR4 coincidentally with positivity for CD68 only in the atherosclerotic plaque of carotid arteries but not in thyroid arteries; the positivity for LPS and TLR4 was greater in the area with activated macrophages. LPS concentration similar to that detected in atherosclerotic plaque resulted in a dose-dependent TLR4-mediated Nox2 up-regulation by human monocytes. These data provide the first evidence that LPS from Escherichia Coli localizes in human plaque and may contribute to atherosclerotic damage via TLR4-mediated oxidative stress.
Collapse
Affiliation(s)
- Roberto Carnevale
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cristina Nocella
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Vincenzo Petrozza
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Vittoria Cammisotto
- I Medical Clinic, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Luca Pacini
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Veronica Sorrentino
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Ombretta Martinelli
- Unit of Vascular Surgery, Department "Paride Stefanini", Sapienza University of Rome, Rome, Italy
| | - Luigi Irace
- Unit of Vascular Surgery, Department "Paride Stefanini", Sapienza University of Rome, Rome, Italy
| | - Sebastiano Sciarretta
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Giacomo Frati
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Daniele Pastori
- I Medical Clinic, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy
| | - Francesco Violi
- I Medical Clinic, Atherothrombosis Center, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
32
|
Oikonomou E, Tsalamandris S, Mourouzis K, Tousoulis D. Biology of the Vessel Wall. Coron Artery Dis 2018. [DOI: 10.1016/b978-0-12-811908-2.00001-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
33
|
|
34
|
Abstract
Hypertension is the leading risk factor for heart disease and stroke, and is estimated to cause 9.4 million deaths globally every year. The pathogenesis of hypertension is complex, but lifestyle factors such as diet are important contributors to the disease. High dietary intake of fruit and vegetables is associated with reduced blood pressure and lower cardiovascular mortality. A critical relationship between dietary intake and the composition of the gut microbiota has been described in the literature, and a growing body of evidence supports the role of the gut microbiota in the regulation of blood pressure. In this Review, we describe the mechanisms by which the gut microbiota and its metabolites, including short-chain fatty acids, trimethylamine N-oxide, and lipopolysaccharides, act on downstream cellular targets to prevent or contribute to the pathogenesis of hypertension. These effects have a direct influence on tissues such as the kidney, the endothelium, and the heart. Finally, we consider the role of the gut microbiota in resistant hypertension, the possible intergenerational effect of the gut microbiota on blood pressure regulation, and the promising therapeutic potential of gut microbiota modification to improve health and prevent disease.
Collapse
Affiliation(s)
- Francine Z Marques
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia.,Department of Pharmacology, Faculty of Medicine, Nursing and Health Sciences, Monash University, Wellington Road, Clayton Victoria 3800, Australia
| | - Charles R Mackay
- Infection and Immunity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, 23 Innovation Walk, Clayton, Victoria 3800, Australia
| | - David M Kaye
- Heart Failure Research Group, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, Victoria 3004, Australia.,Heart Centre, Alfred Hospital, Philip Block, Level 3, 55 Commercial Road, Melbourne, Victoria 3004, Australia.,Central Clinical School, Faculty of Medicine, Nursing and Health Sciences, Monash University, 99 Commercial Road, Melbourne, Victoria 3004, Australia
| |
Collapse
|
35
|
Rai V, Agrawal DK. The role of damage- and pathogen-associated molecular patterns in inflammation-mediated vulnerability of atherosclerotic plaques. Can J Physiol Pharmacol 2017; 95:1245-1253. [PMID: 28746820 DOI: 10.1139/cjpp-2016-0664] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease resulting in the formation of the atherosclerotic plaque. Plaque formation starts with the inflammation in fatty streaks and progresses through atheroma, atheromatous plaque, and fibroatheroma leading to development of stable plaque. Hypercholesterolemia, dyslipidemia, and hyperglycemia are the risk factors for atherosclerosis. Inflammation, infection with viruses and bacteria, and dysregulation in the endothelial and vascular smooth muscle cells leads to advanced plaque formation. Death of the cells in the intima due to inflammation results in secretion of damage-associated molecular patterns (DAMPs) such as high mobility group box 1 (HMGB1), receptor for advanced glycation end products (RAGE), alarmins (S100A8, S100A9, S100A12, and oxidized low-density lipoproteins), and infection with pathogens leads to secretion of pathogen-associated molecular patterns (PAMPs) such as lipopolysaccharides, lipoteichoic acids, and peptidoglycans. DAMPs and PAMPs further activate the inflammatory surface receptors such as TREM-1 and toll-like receptors and downstream signaling kinases and transcription factors leading to increased secretion of pro-inflammatory cytokines such as tumor necrosis factor α, interleukin (IL)-1β, IL-6, and interferon-γ and matrix metalloproteinases (MMPs). These mediators and cytokines along with MMPs render the plaque vulnerable for rupture leading to ischemic events. In this review, we have discussed the role of DAMPs and PAMPs in association with inflammation-mediated plaque vulnerability.
Collapse
Affiliation(s)
- Vikrant Rai
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA.,Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Devendra K Agrawal
- Department of Clinical and Translational Science, Creighton University School of Medicine, Omaha, NE 68178, USA
| |
Collapse
|
36
|
Pastori D, Carnevale R, Nocella C, Novo M, Santulli M, Cammisotto V, Menichelli D, Pignatelli P, Violi F. Gut-Derived Serum Lipopolysaccharide is Associated With Enhanced Risk of Major Adverse Cardiovascular Events in Atrial Fibrillation: Effect of Adherence to Mediterranean Diet. J Am Heart Assoc 2017; 6:e005784. [PMID: 28584074 PMCID: PMC5669181 DOI: 10.1161/jaha.117.005784] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/12/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Gut microbiota is emerging as a novel risk factor for atherothrombosis, but the predictive role of gut-derived lipopolysaccharide (LPS) is unknown. We analyzed (1) the association between LPS and major adverse cardiovascular events (MACE) in atrial fibrillation (AF) and (2) its relationship with adherence to a Mediterranean diet (Med-diet). METHODS AND RESULTS This was a prospective single-center study including 912 AF patients treated with vitamin K antagonists (3716 patient-years). The primary end point was a composite of MACE. Baseline serum LPS, adherence to Med-diet (n=704), and urinary excretion of 11-dehydro-thromboxane B2 (TxB2, n=852) were investigated. Mean age was 73.5 years; 42.9% were women. A total of 187 MACE (5.0% per year) occurred: 54, 59, and 74 in the first, second, and third tertile of LPS, respectively (log-rank test P=0.004). Log-LPS (hazard ratio 1.194, P=0.009), age (hazard ratio 1.083, P<0.001), and previous cerebrovascular (hazard ratio 1.634, P=0.004) and cardiac events (hazard ratio 1.822, P<0.001) were predictors of MACE. In the whole cohort, AF (versus sinus rhythm) (β 0.087, P=0.014) and low-density lipoprotein cholesterol (β 0.069, P=0.049) were associated with circulating LPS. Furthermore, Med-diet score (β -0.137, P<0.001) was predictive of log-LPS, with fruits (β -0.083, P=0.030) and legumes (β -0.120, P=0.002) negatively associated with log-LPS levels. Log-LPS and log-TxB2 were highly correlated (r=0.598, P<0.001). Log-LPS (β 0.574, P<0.001) and Med-diet score (β -0.218, P<0.001) were significantly associated with baseline urinary excretion of TxB2. CONCLUSIONS In this cohort of AF patients, LPS levels were predictive of MACE and negatively affected by high adherence to Med-diet. LPS may contribute to MACE incidence in AF by increasing platelet activation.
Collapse
Affiliation(s)
- Daniele Pastori
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
- Department of Anatomical, Histological, Forensic Medicine and Orthopedics Sciences, Sapienza University of Rome, Italy
| | - Roberto Carnevale
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cristina Nocella
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Marta Novo
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Maria Santulli
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Italy
| | - Vittoria Cammisotto
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Danilo Menichelli
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Pasquale Pignatelli
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| | - Francesco Violi
- I Clinica Medica, Department of Internal Medicine and Medical Specialties, Sapienza University of Rome, Italy
| |
Collapse
|
37
|
Hirano Y, Choi A, Tsuruta M, Jaw JE, Oh Y, Ngan D, Moritani K, Chen YWR, Tam S, Li Y, Vasilescu DM, Hogg JC, Francis G, Bernatchez P, Man SFP, Sin DD. Surfactant protein-D deficiency suppresses systemic inflammation and reduces atherosclerosis in ApoE knockout mice. Cardiovasc Res 2017; 113:1208-1218. [DOI: 10.1093/cvr/cvx067] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Accepted: 04/29/2017] [Indexed: 01/18/2023] Open
|