1
|
Hao J, Zhu Y, Zhang Y, Li L, Li Z, Wang L, Qu Y, Qi L, Yu H, Wang D. Structural characterization and hypolipidemic activity of a hetero-galactan purified from Sanghuangporus vaninii based on modulation of TLR4/NF-κB pathway. Carbohydr Polym 2025; 347:122702. [PMID: 39486943 DOI: 10.1016/j.carbpol.2024.122702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 09/01/2024] [Accepted: 09/02/2024] [Indexed: 11/04/2024]
Abstract
Sanghuangporus vaninii showed great activities of anti-inflammation and anti-tumor, due to its bioactive macromolecules. However, the hypolipidemic properties of polysaccharides isolated from S. vaninii have not been systematically reported. In this research, a polysaccharide of S. vaninii was obtained and its hypolipidemic activity was investigated. SVP3, a neutral hetero-galactan from S. vaninii, has a →6)-α-Galp-(1→ backbone with partial H-2 branches of α-Manp-(1→ or α-Manp-(1→2)-α-Fucp-(1→. In a hyperlipidemia mouse model, SVP3 significantly inhibited body weight gain and suppressed serum levels of total cholesterol, triglycerides, and low-density lipoprotein cholesterol. SVP3 inhibited the expansion of adipocytes in three types of white adipose tissues and attenuated hepatic injury and hepatic lipid deposition in the mice. The combined analysis of gut microbiota, serum metabolomics, and liver proteomics revealed that SVP3 effectively regulated the abundance of specific gut microbiota and serum metabolites and mediated the inhibitory effect on inflammation-associated toll-like receptor 4/nuclear factor kappa-B pathway by regulating the expression levels of glutathione S-transferase P1, stromal cell derived factor 2-like 1, ribosomal protein L10, thiosulfate sulfurtransferase, and biliverdin reductase A in liver, ultimately realizing the hypolipidemic activity. The results of the present study provide experimental evidence for the development of clinical adjuvant therapeutic drugs to treat hyperlipidemia.
Collapse
Affiliation(s)
- Jie Hao
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yanfeng Zhu
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yongfeng Zhang
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China.
| | - Lanzhou Li
- Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China.
| | - Zhige Li
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Lu Wang
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Yidi Qu
- School of Life Sciences, Jilin University, Changchun 130012, China.
| | - Liangliang Qi
- Microbiology Research Institute, Guangxi Academy of Agricultural Sciences, Nanning 530007, China.
| | - Hailong Yu
- Institute of Edible Fungi, Shanghai Academy of Agricultural Sciences, National Engineering Research Center of Edible Fungi, Shanghai 201403, China.
| | - Di Wang
- School of Life Sciences, Jilin University, Changchun 130012, China; Engineering Research Center of Chinese Ministry of Education for Edible and Medicinal Fungi, Jilin Agricultural University, Changchun 130118, China.
| |
Collapse
|
2
|
Sugino KY, Hernandez TL, Barbour LA, Kofonow JM, Frank DN, Friedman JE. Distinct Plasma Metabolomic and Gut Microbiome Profiles after Gestational Diabetes Mellitus Diet Treatment: Implications for Personalized Dietary Interventions. Microorganisms 2024; 12:1369. [PMID: 39065137 PMCID: PMC11278888 DOI: 10.3390/microorganisms12071369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 06/24/2024] [Accepted: 06/29/2024] [Indexed: 07/28/2024] Open
Abstract
Gestational diabetes mellitus (GDM) triggers alterations in the maternal microbiome. Alongside metabolic shifts, microbial products may impact clinical factors and influence pregnancy outcomes. We investigated maternal microbiome-metabolomic changes, including over 600 metabolites from a subset of the "Choosing Healthy Options in Carbohydrate Energy" (CHOICE) study. Women diagnosed with GDM were randomized to a diet higher in complex carbohydrates (CHOICE, n = 18, 60% complex carbohydrate/25% fat/15% protein) or a conventional GDM diet (CONV, n = 16, 40% carbohydrate/45% fat/15% protein). All meals were provided. Diets were eucaloric, and fiber content was similar. CHOICE was associated with increases in trimethylamine N-oxide, indoxyl sulfate, and several triglycerides, while CONV was associated with hippuric acid, betaine, and indole propionic acid, suggestive of a healthier metabolome. Conversely, the microbiome of CHOICE participants was enriched with carbohydrate metabolizing genes and beneficial taxa such as Bifidobacterium adolescentis, while CONV was associated with inflammatory pathways including antimicrobial resistance and lipopolysaccharide biosynthesis. We also identified latent metabolic groups not associated with diet: a metabolome associated with less of a decrease in fasting glucose, and another associated with relatively higher fasting triglycerides. Our results suggest that GDM diets produce specific microbial and metabolic responses during pregnancy, while host factors also play a role in triglycerides and glucose metabolism.
Collapse
Affiliation(s)
- Kameron Y. Sugino
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
| | - Teri L. Hernandez
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA; (T.L.H.); (L.A.B.)
- College of Nursing, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Linda A. Barbour
- Department of Medicine, Division of Endocrinology, Metabolism and Diabetes, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA; (T.L.H.); (L.A.B.)
- Department of Obstetrics and Gynecology, Division of Maternal-Fetal Medicine, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA
| | - Jennifer M. Kofonow
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA; (J.M.K.); (D.N.F.)
| | - Daniel N. Frank
- Department of Medicine, Division of Infectious Diseases, University of Colorado Anschutz Medical Center, Aurora, CO 80045, USA; (J.M.K.); (D.N.F.)
| | - Jacob E. Friedman
- Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA;
- Department of Biochemistry & Physiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
3
|
Nooti S, Rai V, Radwan MM, Thankam FG, Singh H, Chatzizisis YS, Agrawal DK. Oxidized Low-density Lipoproteins and Lipopolysaccharides Augment Carotid Artery Plaque Vulnerability in Hypercholesterolemic Microswine. CARDIOLOGY AND CARDIOVASCULAR MEDICINE 2023; 7:273-294. [PMID: 37577745 PMCID: PMC10421630 DOI: 10.26502/fccm.92920338] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Atherosclerosis is a chronic inflammatory disease and hypercholesterolemia is a risk factor. This study aims to compare the potency of lipopolysaccharide (LPS) and oxidized low-density lipoproteins (oxLDL) to induce plaque formation and increase plaque vulnerability in the carotid artery of hypercholesterolemic Yucatan microswine. Atherosclerotic lesions at the common carotid artery junction and ascending pharyngeal artery were induced in hypercholesterolemic Yucatan microswine at 5-6 months of age with balloon angioplasty. LPS or oxLDL were administered intraluminally at the site of injury after occluding the arterial flow temporarily. Pre-intervention ultrasound (US), angiography, and optical coherence tomography (OCT) were done at baseline and just before euthanasia to assess post-op parameters. The images from the US, OCT, and angiography in the LPS and the oxLDL-treated group showed increased plaque formation with features suggestive of unstable plaque, including necrotic core, thin fibrous caps, and a signal poor region more with oxLDL compared to LPS. Histomorphology of the carotid artery tissue near the injury corroborated the presence of severe lesions in both LPS and oxLDL-treated pigs but more in the oxLDL group. Vascular smooth muscle and endothelial cells treated with LPS and oxLDL showed increased folds changes in mRNA transcripts of the biomarkers of inflammation and plaque vulnerability compared to untreated cells. Collectively, the results suggest that angioplasty-mediated intimal injury of the carotid arteries in atherosclerotic swine with local administration of LPS or ox-LDL induces vulnerable plaques compared to angioplasty alone and oxLDL is relatively more potent than LPS in inducing vulnerable plaque.
Collapse
Affiliation(s)
- S Nooti
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - V Rai
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - M M Radwan
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - F G Thankam
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - H Singh
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| | - Y S Chatzizisis
- Division of Cardiovascular Medicine, Leonard M. Miller School of Medicine University of Miami, Miami, FL 33136, USA
| | - D K Agrawal
- Department of Translational Research, Western University of Health Sciences, Pomona, California 91763, USA
| |
Collapse
|
4
|
Zhuo X, Luo H, Lei R, Lou X, Bian J, Guo J, Luo H, Zhang X, Jiao Q, Gong W. Association between Intestinal Microecological Changes and Atherothrombosis. Microorganisms 2023; 11:1223. [PMID: 37317197 PMCID: PMC10222604 DOI: 10.3390/microorganisms11051223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 06/16/2023] Open
Abstract
Atherosclerosis (AS) is a chronic inflammatory disease of large- and medium-sized arteries that causes ischemic heart disease, strokes, and peripheral vascular disease, collectively called cardiovascular disease (CVD), and is the leading cause of CVD resulting in a high rate of mortality in the population. AS is pathological by plaque development, which is caused by lipid infiltration in the vessel wall, endothelial dysfunction, and chronic low-grade inflammation. Recently, more and more scholars have paid attention to the importance of intestinal microecological disorders in the occurrence and development of AS. Intestinal G-bacterial cell wall lipopolysaccharide (LPS) and bacterial metabolites, such as oxidized trimethylamine (TMAO) and short-chain fatty acids (SCFAs), are involved in the development of AS by affecting the inflammatory response, lipid metabolism, and blood pressure regulation of the body. Additionally, intestinal microecology promotes the progression of AS by interfering with the normal bile acid metabolism of the body. In this review, we summarize the research on the correlation between maintaining a dynamic balance of intestinal microecology and AS, which may be potentially helpful for the treatment of AS.
Collapse
Affiliation(s)
- Xinyu Zhuo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
| | - Hui Luo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| | - Rumei Lei
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
| | - Xiaokun Lou
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
| | - Jing Bian
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
| | - Junfeng Guo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
| | - Hao Luo
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
| | - Xingwei Zhang
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| | - Qibin Jiao
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
| | - Wenyan Gong
- Department of Clinical Medicine, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Wenzhou Road, Gongshu District, Hangzhou 310000, China; (X.Z.); (H.L.); (R.L.); (X.L.); (J.B.); (J.G.); (H.L.); (X.Z.)
- Hangzhou Institute of Cardiovascular Disease, Hangzhou 310000, China
| |
Collapse
|
5
|
Hypotheses on Atherogenesis Triggering: Does the Infectious Nature of Atherosclerosis Development Have a Substruction? Cells 2023; 12:cells12050707. [PMID: 36899843 PMCID: PMC10001176 DOI: 10.3390/cells12050707] [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: 12/19/2022] [Revised: 02/03/2023] [Accepted: 02/21/2023] [Indexed: 02/25/2023] Open
Abstract
Since the end of the 20th century, it has been clear that atherosclerosis is an inflammatory disease. However, the main triggering mechanism of the inflammatory process in the vascular walls is still unclear. To date, many different hypotheses have been put forward to explain the causes of atherogenesis, and all of them are supported by strong evidence. Among the main causes of atherosclerosis, which underlies these hypotheses, the following can be mentioned: lipoprotein modification, oxidative transformation, shear stress, endothelial dysfunction, free radicals' action, homocysteinemia, diabetes mellitus, and decreased nitric oxide level. One of the latest hypotheses concerns the infectious nature of atherogenesis. The currently available data indicate that pathogen-associated molecular patterns from bacteria or viruses may be an etiological factor in atherosclerosis. This paper is devoted to the analysis of existing hypotheses for atherogenesis triggering, and special attention is paid to the contribution of bacterial and viral infections to the pathogenesis of atherosclerosis and cardiovascular disease.
Collapse
|
6
|
Francisco AJ. Helicobacter Pylori Infection Induces Intestinal Dysbiosis That Could Be Related to the Onset of Atherosclerosis. BIOMED RESEARCH INTERNATIONAL 2022; 2022:9943158. [PMID: 36317116 PMCID: PMC9617700 DOI: 10.1155/2022/9943158] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 10/05/2022] [Accepted: 10/06/2022] [Indexed: 11/17/2022]
Abstract
Cardiovascular diseases represent one of the first causes of death around the world, and atherosclerosis is one of the first steps in the development of them. Although these problems occur mainly in elderly, the incidence in younger people is being reported, and an undetermined portion of patients without the classic risk factors develop subclinical atherosclerosis at earlier stages of life. Recently, both the H. pylori infection and the intestinal microbiota have been linked to atherosclerosis. The mechanisms behind those associations are poorly understood, but some of the proposed explanations are (a) the effect of the chronic systemic inflammation induced by H. pylori, (b) a direct action over the endothelial cells by the cytotoxin associated gene A protein, and (c) alterations of the lipid metabolism and endothelial dysfunction induced by H. pylori infection. Regarding the microbiota, several studies show that induction of atherosclerosis is related to high levels of Trimethylamine N-oxide. In this review, we present the information published about the effects of H. pylori over the intestinal microbiota and their relationship with atherosclerosis and propose a hypothesis to explain the nature of these associations. If H. pylori contributes to atherosclerosis, then interventions for eradication and restoration of the gut microbiota at early stages could represent a way to prevent disease progression.
Collapse
Affiliation(s)
- Avilés-Jiménez Francisco
- Unidad de Investigación Médica en Enfermedades Infecciosas y Parasitarias, UMAE Pediatría. Centro Médico Nacional Siglo XXI. IMSS, Ciudad de México, Mexico
| |
Collapse
|
7
|
Nikolaeva SD, Fock EM, Parnova RG. Lipopolysaccharide Stimulates Triglyceride Accumulation and Lipid Droplet Biogenesis in PC12 Cells: the Role of Carnitine Palmitoyltransferase 1 Down-Regulation and Suppression of Fatty Acid Oxidation. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022040184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Singh H, Rai V, Agrawal DK. LPS and oxLDL-induced S100A12 and RAGE expression in carotid arteries of atherosclerotic Yucatan microswine. Mol Biol Rep 2022; 49:8663-8672. [PMID: 35771356 DOI: 10.1007/s11033-022-07703-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2022] [Accepted: 06/10/2022] [Indexed: 11/24/2022]
Abstract
BACKGROUND S100A12, also known as Calgranulin C, is a ligand for the receptor for advanced glycation end products (RAGE) and plays key roles in cardiovascular and other inflammatory diseases. Interactions between S100A12 and RAGE initiate downstream signaling activating extracellular signal-regulated kinases (ERK1/2), mitogen activated protein kinases (MAPK), and transcription factor NF-κB. This increases the expression of pro-inflammatory cytokines to induce the inflammatory response. S100A12, and RAGE play a critical role in the development and progression of atherosclerosis. There is a well-known relationship between the bacterial endotoxin lipopolysaccharide (LPS) and the lipid antigens oxidized low-density lipoprotein (oxLDL) in driving the immune response in atherosclerosis. METHODS AND RESULTS Our study aimed to compare the potential of LPS and oxLDL in regulating the expression of S100A12 and RAGE in atherosclerosis. The expression of these proteins was assessed in the harvested carotid arteries from LPS- and oxLDL-treated atherosclerotic Yucatan microswine. Tissues were collected from five different treatment groups: (i) angioplasty alone, (ii) LPS alone, (iii) oxLDL alone, (iv) angioplasty with LPS, and (v) angioplasty with oxLDL. Immunohistochemical findings revealed that angioplasty with LPS induced higher expression of S100A12 and RAGE compared to other treatment groups. The results were further corroborated by testing their gene expression through qPCR in cultured vascular smooth muscle cells (VSMCs) isolated from control carotid arteries and LPS- and oxLDL-treated arteries. CONCLUSIONS The results of this study suggest that LPS induces the expression of S100A12 and RAGE more than oxLDL in atherosclerotic artery and both S100A12 and RAGE could be therapeutic targets.
Collapse
Affiliation(s)
- Harbinder Singh
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Vikrant Rai
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 E. Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
9
|
Abstract
Lipid droplets (LDs) are ubiquitous organelles that store and supply lipids for energy metabolism, membrane synthesis and production of lipid-derived signaling molecules. While compositional differences in the phospholipid monolayer or neutral lipid core of LDs impact their metabolism and function, the proteome of LDs has emerged as a major influencer in all aspects of LD biology. The perilipins (PLINs) are the most studied and abundant proteins residing on the LD surface. This Cell Science at a Glance and the accompanying poster summarize our current knowledge of the common and unique features of the mammalian PLIN family of proteins, the mechanisms through which they affect cell metabolism and signaling, and their links to disease.
Collapse
Affiliation(s)
- Charles P. Najt
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Mahima Devarajan
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Douglas G. Mashek
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
- Department of Medicine, Division of Diabetes, Endocrinology and Metabolism, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
10
|
Page MJ, Kell DB, Pretorius E. The Role of Lipopolysaccharide-Induced Cell Signalling in Chronic Inflammation. CHRONIC STRESS (THOUSAND OAKS, CALIF.) 2022; 6:24705470221076390. [PMID: 35155966 PMCID: PMC8829728 DOI: 10.1177/24705470221076390] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Accepted: 01/11/2022] [Indexed: 12/20/2022]
Abstract
Lipopolysaccharide (LPS) is the main structural component of the outer membrane of most Gram-negative bacteria and has diverse immunostimulatory and procoagulant effects. Even though LPS is well described for its role in the pathology of sepsis, considerable evidence demonstrates that LPS-induced signalling and immune dysregulation are also relevant in the pathophysiology of many diseases, characteristically where endotoxaemia is less severe. These diseases are typically chronic and progressive in nature and span broad classifications, including neurodegenerative, metabolic, and cardiovascular diseases. This Review reappraises the mechanisms of LPS-induced signalling and emphasises the crucial contribution of LPS to the pathology of multiple chronic diseases, beyond conventional sepsis. This perspective asserts that new ways of approaching chronic diseases by targeting LPS-driven pathways may be of therapeutic benefit in a wide range of chronic inflammatory conditions.
Collapse
Affiliation(s)
| | - Douglas B Kell
- Stellenbosch University, Stellenbosch, South Africa.,Institute of Integrative Biology, University of Liverpool, Liverpool, UK.,The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Lyngby, Denmark
| | | |
Collapse
|
11
|
Lin S, Zhang H, Wang X, Lin T, Chen Z, Liu J, Wang J. Abundance of Lipopolysaccharide Heptosyltransferase I in Human Gut Microbiome and Its Association With Cardiovascular Disease and Liver Cirrhosis. Front Microbiol 2021; 12:756976. [PMID: 34917047 PMCID: PMC8669917 DOI: 10.3389/fmicb.2021.756976] [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: 08/11/2021] [Accepted: 11/09/2021] [Indexed: 11/13/2022] Open
Abstract
Lipopolysaccharide (LPS) is a potent endotoxin on the outer membrane of gram-negative bacteria. Heptosyltransferase I (HpeI) takes part in the synthesis of LPS. In this study, we first collected the protein sequences of HpeI homologs from the human microbiome. The collected HpeI sequences was classified based on sequence similarity, and seven clusters of HpeI were obtained. Among these clusters, proteins from Cluster 3 were abundant in the human mouth, while Clusters 1, 6, and 7 were abundant in the human gut. In addition, proteins from Cluster 1 were mainly from the order of Enterobacterales, while Cluster 6 and 7 were from Burkholderiales. The correlation analysis indicated that the total abundance of HpeIs was increased in patients with cardiovascular disease and liver cirrhosis, and HpeI in Cluster 1 contributed to this increase. These data suggest that HpeI homologs in Cluster 1 can be recognized as biomarkers for cardiovascular disease and liver cirrhosis, and that reducing the bacterial load in Cluster 1 may contribute to disease therapy.
Collapse
Affiliation(s)
- Shujin Lin
- Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Hui Zhang
- Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Xueke Wang
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Ting Lin
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Zihan Chen
- College of Biological Science and Engineering, Fuzhou University, Fuzhou, China
| | - Jingfeng Liu
- Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| | - Jianmin Wang
- Fujian Cancer Hospital, Fujian Medical University Cancer Hospital, Fuzhou, China
| |
Collapse
|
12
|
Feng V, Bawa KK, Marzolini S, Kiss A, Oh P, Herrmann N, Lanctôt KL, Gallagher D. Impact of 12-week exercise program on biomarkers of gut barrier integrity in patients with coronary artery disease. PLoS One 2021; 16:e0260165. [PMID: 34797867 PMCID: PMC8604291 DOI: 10.1371/journal.pone.0260165] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 10/28/2021] [Indexed: 12/03/2022] Open
Abstract
Introduction Breakdown of gut barrier integrity has been associated with inflammatory activation and is implicated in the etiology of several chronic medical conditions. Acute exercise is known to increase gut barrier permeability but the impact of chronic exercise is not clear. Most studies to date have examined how acute exercise impacts gut barrier integrity in healthy adults, while few studies have examined the impact of chronic exercise in older adults with comorbidities. We aim to investigate the impact of a 12-week program of aerobic and resistance training on biomarkers of gut barrier integrity in a sample of older adults with coronary artery disease. Methods Participants were adults with coronary artery disease undergoing a moderate-intensity 12-week cardiac rehabilitation exercise program. Fasting blood samples were taken at baseline and study termination. Serum levels of biomarkers of gut barrier integrity (zonulin and fatty acid-binding protein 2 (FABP2)) were measured by ELISA. Cardiorespiratory fitness was assessed by peak oxygen uptake (VO2peak) at study start & completion. Data analyses were performed using SPSS software version 24.0. Results Among study participants (n = 41, 70% male, age = 62.7± 9.35) we found a significant negative association between baseline FABP2 levels and baseline VO2peak in a multiple linear regression model adjusting for covariates (B = -0.3, p = 0.009). Over the course of the exercise program an increase in VO2peak (≥ 5 mL/kg/min) was independently associated with a relative decrease in FABP2 (B = -0.45, p = 0.018) after controlling for medical covariates. Conclusion Our findings indicate that an increase in cardiorespiratory fitness during a 12-week exercise program resulted in a relative improvement in a biomarker of gut barrier integrity. This indicates a potential mechanism by which longer term exercise may improve gut barrier integrity.
Collapse
Affiliation(s)
- Vivian Feng
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Neuropsychopharmacology Research Group, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kritleen K. Bawa
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Neuropsychopharmacology Research Group, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Susan Marzolini
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada
- KITE Toronto Rehabilitation Institute, University Health Network, East York, Ontario, Canada
| | - Alex Kiss
- ICES, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Paul Oh
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Nathan Herrmann
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Neuropsychopharmacology Research Group, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Krista L. Lanctôt
- Department of Pharmacology & Toxicology, University of Toronto, Toronto, Ontario, Canada
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Neuropsychopharmacology Research Group, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Heart and Stroke Foundation Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute, Toronto, Ontario, Canada
- KITE Toronto Rehabilitation Institute, University Health Network, East York, Ontario, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Damien Gallagher
- Neuropsychopharmacology Research Group, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
13
|
Karlsen TR, Kong XY, Holm S, Quiles-Jiménez A, Dahl TB, Yang K, Sagen EL, Skarpengland T, S Øgaard JD, Holm K, Vestad B, Olsen MB, Aukrust P, Bjørås M, Hov JR, Halvorsen B, Gregersen I. NEIL3-deficiency increases gut permeability and contributes to a pro-atherogenic metabolic phenotype. Sci Rep 2021; 11:19749. [PMID: 34611194 PMCID: PMC8492623 DOI: 10.1038/s41598-021-98820-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis and its consequences cause considerable morbidity and mortality world-wide. We have previously shown that expression of the DNA glycosylase NEIL3 is regulated in human atherosclerotic plaques, and that NEIL3-deficiency enhances atherogenesis in Apoe-/- mice. Herein, we identified a time point prior to quantifiable differences in atherosclerosis between Apoe-/-Neil3-/- mice and Apoe-/- mice. Mice at this age were selected to explore the metabolic and pathophysiological processes preceding extensive atherogenesis in NEIL3-deficient mice. Untargeted metabolomic analysis of young Apoe-/-Neil3-/- mice revealed significant metabolic disturbances as compared to mice expressing NEIL3, particularly in metabolites dependent on the gut microbiota. 16S rRNA gene sequencing of fecal bacterial DNA indeed confirmed that the NEIL3-deficient mice had altered gut microbiota, as well as increased circulating levels of the bacterially derived molecule LPS. The mice were challenged with a FITC-conjugated dextran to explore gut permeability, which was significantly increased in the NEIL3-deficient mice. Further, immunohistochemistry showed increased levels of the proliferation marker Ki67 in the colonic epithelium of NEIL3-deficient mice, suggesting increased proliferation of intestinal cells and gut leakage. We suggest that these metabolic alterations serve as drivers of atherosclerosis in NEIL3-deficient mice.
Collapse
Affiliation(s)
- Tom Rune Karlsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Xiang Yi Kong
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Sverre Holm
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ana Quiles-Jiménez
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tuva B Dahl
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Research and Development, Division of Emergencies and Critical Care, Oslo University Hospital HF, Rikshospitalet, Oslo, Norway
| | - Kuan Yang
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Ellen L Sagen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Tonje Skarpengland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Jonas D S Øgaard
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Kristian Holm
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Beate Vestad
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Maria B Olsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Magnar Bjørås
- Department of Microbiology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Johannes R Hov
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Section of Gastroenterology, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
- Norwegian PSC Research Center, Department of Transplantation Medicine, Oslo University Hospital, Oslo, Norway
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway.
| | - Ida Gregersen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway.
| |
Collapse
|
14
|
Native Low-Density Lipoproteins Act in Synergy with Lipopolysaccharide to Alter the Balance of Human Monocyte Subsets and Their Ability to Produce IL-1 Beta, CCR2, and CX3CR1 In Vitro and In Vivo: Implications in Atherogenesis. Biomolecules 2021; 11:biom11081169. [PMID: 34439835 PMCID: PMC8391227 DOI: 10.3390/biom11081169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/29/2021] [Accepted: 08/04/2021] [Indexed: 12/17/2022] Open
Abstract
Increasing evidence has demonstrated that oxidized low-density lipoproteins (oxLDL) and lipopolysaccharide (LPS) enhance accumulation of interleukin (IL)-1 beta-producing macrophages in atherosclerotic lesions. However, the potential synergistic effect of native LDL (nLDL) and LPS on the inflammatory ability and migration pattern of monocyte subpopulations remains elusive and is examined here. In vitro, whole blood cells from healthy donors (n = 20) were incubated with 100 μg/mL nLDL, 10 ng/mL LPS, or nLDL + LPS for 9 h. Flow cytometry assays revealed that nLDL significantly decreases the classical monocyte (CM) percentage and increases the non-classical monocyte (NCM) subset. While nLDL + LPS significantly increased the number of NCMs expressing IL-1 beta and the C-C chemokine receptor type 2 (CCR2), the amount of NCMs expressing the CX3C chemokine receptor 1 (CX3CR1) decreased. In vivo, patients (n = 85) with serum LDL-cholesterol (LDL-C) >100 mg/dL showed an increase in NCM, IL-1 beta, LPS-binding protein (LBP), and Castelli’s atherogenic risk index as compared to controls (n = 65) with optimal LDL-C concentrations (≤100 mg/dL). This work demonstrates for the first time that nLDL acts in synergy with LPS to alter the balance of human monocyte subsets and their ability to produce inflammatory cytokines and chemokine receptors with prominent roles in atherogenesis.
Collapse
|
15
|
Cho SH, Kim SR, Jeong MS, Choi M, Park S, Kim KN. Protective Effect of Brassica napus L. Hydrosols against Inflammation Response in RAW 264.7 Cells. Chin J Integr Med 2021; 27:273-279. [PMID: 33759044 DOI: 10.1007/s11655-021-3330-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2019] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To demonstrate the anti-inflammatory activity of Brassica napus L. hydrosols (BNH) in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. METHODS Composition analysis of BNH was conducted via gas chromatography-mass spectrometry after BNH were extracted. The nitric oxide (NO) production was measured using the Griess assay. Prostaglandin E2 (PGE2) production was evaluated with enzyme-linked immunosorbent assay. The effects of BNH on LPS-induced pro-inflammatory enzymes including inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) were evaluated using Western blot analysis. Furthermore, phosphorylation of nuclear factor-kappa B (NF-κB) and nuclear translocation of NF-κB p65 were evaluated with Western blot analysis and immunofluorescence staining, respectively. RESULTS Compared with LPS-stimulated cells, BNH markedly decreased the generation of NO and PGE2 in LPS-stimulated RAW 264.7 cells (P<0.01 or P<0.05). Moreover, BNH inhibited protein levels of iNOS and COX-2 (P<0.01). Phosphorylation of NF-κB and nuclear translocation of NF-κB p65 was significantly inhibited by BNH (P<0.01 or P<0.05). CONCLUSION The anti-inflammatory activities of BNH were mediated via blockage of the NF-κB signaling pathways in LPS-stimulated RAW 264.7 cells.
Collapse
Affiliation(s)
- Su-Hyeon Cho
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, Republic of Korea
| | - Song Rae Kim
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, Republic of Korea
| | - Myeong Seon Jeong
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, Republic of Korea
| | - Miri Choi
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, Republic of Korea
| | - SeonJu Park
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, Republic of Korea
| | - Kil-Nam Kim
- Chuncheon Center, Korea Basic Science Institute, Chuncheon, 24341, Republic of Korea.
| |
Collapse
|
16
|
Kurilenko N, Fatkhullina AR, Mazitova A, Koltsova EK. Act Locally, Act Globally-Microbiota, Barriers, and Cytokines in Atherosclerosis. Cells 2021; 10:cells10020348. [PMID: 33562334 PMCID: PMC7915371 DOI: 10.3390/cells10020348] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 12/12/2022] Open
Abstract
Atherosclerosis is a lipid-driven chronic inflammatory disease that is characterized by the formation and progressive growth of atherosclerotic plaques in the wall of arteries. Atherosclerosis is a major predisposing factor for stroke and heart attack. Various immune-mediated mechanisms are implicated in the disease initiation and progression. Cytokines are key mediators of the crosstalk between innate and adaptive immune cells as well as non-hematopoietic cells in the aortic wall and are emerging players in the regulation of atherosclerosis. Progression of atherosclerosis is always associated with increased local and systemic levels of pro-inflammatory cytokines. The role of cytokines within atherosclerotic plaque has been extensively investigated; however, the cell-specific role of cytokine signaling, particularly the role of cytokines in the regulation of barrier tissues tightly associated with microbiota in the context of cardiovascular diseases has only recently come to light. Here, we summarize the knowledge about the function of cytokines at mucosal barriers and the interplay between cytokines, barriers, and microbiota and discuss their known and potential implications for atherosclerosis development.
Collapse
Affiliation(s)
- Natalia Kurilenko
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
| | | | - Aleksandra Mazitova
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
| | - Ekaterina K. Koltsova
- Department of Medicine and Department of Biomedical Sciences, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA 90048, USA; (N.K.); (A.M.)
- Correspondence:
| |
Collapse
|
17
|
Sun J, Xu X, Huang X, Ji S, Bian C, Ji H. Nuclear factor-κB subunit p65 is involved in lipopolysaccharide-induced lipid accumulation via regulating DGAT1b in Ctenopharyngodon idellus kidney cells. FISH & SHELLFISH IMMUNOLOGY 2020; 105:71-77. [PMID: 32585360 DOI: 10.1016/j.fsi.2020.05.071] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 05/24/2020] [Accepted: 05/26/2020] [Indexed: 06/11/2023]
Abstract
Lipopolysaccharide (LPS) can promote the accumulation of triglycerides (TGs) in CIK (Ctenopharyngodon idellus kidney) cells, but the underlying mechanism is unclear. In this study, two genes involved TG synthesis, DGAT1a and DGAT1b, were isolated and characterized from grass carp Ctenopharyngodon idella, which encode peptides of 498 and 501 amino acids, respectively. Phylogenetic and synteny analyses indicated that DGAT1a and DGAT1b could have originated from the teleost-specific genome duplication event. Analysis of the exon-intron structures clarified that genomic structures of all DGAT1 proteins are conserved in vertebrates. DGAT1a mRNA was highly expressed in gut, adipose tissue and heart, while DGAT1b mRNA was highly expressed in liver and kidney. After LPS treatment, only expression of DGAT1b was up-regulated and knockdown of DGAT1b reduced the content of TG, suggesting that DGAT1b is involved in LPS-induced lipid accumulation. To explore the mechanism underlying the transcriptional regulation of DGAT1b in response to LPS, we cloned DGAT1b promoter sequence. Its promoter sequence consists of IRF7, RelA (p65) and RelB binding elements. Dual luciferase assay and q-PCR suggested that the promoter of DGAT1b can be activated by the overexpression of p65, but cannot be triggered by IRF7 and RelB. Mutational analysis shows that the potential p65 binding sites may locate in the region -111/-100 bp of the DGAT1b promoter. These results indicated that DGAT1b is the target gene of NF-κB p65. Finally, inhibiting p65 effectively decreased LPS-induced lipid accumulation. Taken together, we demonstrate that NF-κB p65 takes part in the lipid accumulation by regulating DGAT1b-induced TG synthesis in LPS signalling in CIK cells. The finding that NF-κB p65 links LPS signalling and TG synthesis adds to our growing appreciation of the interplay between immunity and lipid metabolism in fish.
Collapse
Affiliation(s)
- Jian Sun
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xinxin Xu
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Xiaocheng Huang
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Shanghong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Chenchen Bian
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Hong Ji
- College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW This review aims to highlight the association between gut microbiome and cardiovascular disease (CVD) with emphasis on the possible molecular mechanisms by which how gut microbiome contributes to CVD. RECENT FINDINGS Increasingly, the roles of gut microbiome in cardiovascular health and disease have gained much attention. Most of the investigations focus on how the gut dysbiosis contributes to CVD risk factors and which gut microbial-derived metabolites mediate such effects. SUMMARY In this review, we discuss the molecular mechanisms of gut microbiome contributing to CVD, which include gut microbes translocalization to aortic artery because of gut barrier defect to initiate inflammation and microbial-derived metabolites inducing inflammation-signaling pathway and renal insufficiency. Specifically, we categorize beneficial and deleterious microbial-derived metabolites in cardiovascular health. We also summarize recent findings in the gut microbiome modulation of drug efficacy in treatment of CVD and the microbiome mechanisms by which how physical exercise ameliorates cardiovascular health. Gut microbiome has become an essential component of cardiovascular research and a crucial consideration factor in cardiovascular health and disease.
Collapse
|
19
|
Kashtanova DA, Tkacheva ON. The phenomenon of intestinal permeability and its association with cardiovascular disease. Current status. КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2020. [DOI: 10.15829/1728-8800-2020-2474] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Changes in the intestinal permeability in various pathologies are widely discussed in the scientific community. There is still no consensus on whether high intestinal permeability can lead to chronic noncommunicable diseases, but there is much evidence that increased permeability can aggravate some of them. The article discusses a modern vision of the intestinal permeability including its potential contribution to the development of cardiovascular pathologies, which are the number one mortality cause both in Russia and around the world.
Collapse
Affiliation(s)
- D. A. Kashtanova
- Russian Clinical and Research Center of Gerontology, Pirogov Russian National Research Medical University
| | - O. N. Tkacheva
- Russian Clinical and Research Center of Gerontology, Pirogov Russian National Research Medical University
| |
Collapse
|
20
|
Zhou Y, Little PJ, Downey L, Afroz R, Wu Y, Ta HT, Xu S, Kamato D. The Role of Toll-like Receptors in Atherothrombotic Cardiovascular Disease. ACS Pharmacol Transl Sci 2020; 3:457-471. [PMID: 32566912 PMCID: PMC7296543 DOI: 10.1021/acsptsci.9b00100] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Indexed: 02/06/2023]
Abstract
Toll-like receptors (TLRs) are dominant components of the innate immune system. Activated by both pathogen-associated molecular patterns and damage-associated molecular patterns, TLRs underpin the pathology of numerous inflammation related diseases that include not only immune diseases, but also cardiovascular disease (CVD), diabetes, obesity, and cancers. Growing evidence has demonstrated that TLRs are involved in multiple cardiovascular pathophysiologies, such as atherosclerosis and hypertension. Specifically, a trial called the Canakinumab Anti-inflammatory Thrombosis Outcomes Study showed the use of an antibody that neutralizes interleukin-1β, reduces the recurrence of cardiovascular events, demonstrating inflammation as a therapeutic target and also the research value of targeting the TLR system in CVD. In this review, we provide an update of the interplay between TLR signaling, inflammatory mediators, and atherothrombosis, with an aim to identify new therapeutic targets for atherothrombotic CVD.
Collapse
Affiliation(s)
- Ying Zhou
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Peter J. Little
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| | - Liam Downey
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Rizwana Afroz
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
| | - Yuao Wu
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Hang T. Ta
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Australian
Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, St Lucia, Queensland 4072, Australia
| | - Suowen Xu
- Aab
Cardiovascular Research Institute, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, New York 14642, United States
| | - Danielle Kamato
- School
of Pharmacy, University of Queensland, Pharmacy
Australia Centre of Excellence, Woolloongabba, Queensland 4102, Australia
- Department
of Pharmacy, Xinhua College of Sun Yat-Sen
University, Tianhe District, Guangzhou, Guangdong Province 510520, China
| |
Collapse
|
21
|
Guo J, Li Y, Zhao R, Yang X. Adipokine zinc-α2-glycoprotein alleviates lipopolysaccharide-induced inflammatory responses through the β3-AR/PKA/CREB pathway. Cytokine 2019; 123:154742. [PMID: 31260855 DOI: 10.1016/j.cyto.2019.154742] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/25/2022]
Abstract
Humans and animals frequently experience dysmetabolism induced by inflammation. Zinc-α2-glycoprotein (ZAG), a newly identified adipokine, is potentially involved in lipid metabolism. Our previous study revealed that the ZAG content increased after lipopolysaccharide (LPS) treatment. To clarify ZAG's possible effects on inflammatory responses and lipid metabolism, we used gene overexpression and knockout mice as models to investigate the function of ZAG during inflammation. The results showed that LPS increased plasma triglyceride, non-esterified fatty acid and hepatic triglyceride, while ZAG overexpression decreased these effects. Furthermore, ZAG overexpression weakened inflammatory responses, suppressed lipogenesis, and improved mitochondrial function during inflammation. ZAG overexpression also increased β3-adrenoreceptor, protein kinase A, and phosphorylated cyclic adenosine monophosphate-response element binding protein (CREB), promoted the combination of CREB and CREB-binding protein (CBP), and competitively inhibited the combination of nuclear factor-κB and CBP. After ZAG knockout, LPS-induced the hyperlipidemia worsened. ZAG knockout aggravated inflammatory responses, promoted lipogenesis, and weakened mitochondrial function during inflammation. ZAG knockout also decreased β3-adrenoreceptor and protein kinase A. The present study demonstrated that ZAG alleviated lipid metabolism disorders by weakening inflammatory responses. The β3-adrenoreceptor/protein kinase A/CREB pathway mediated the effects of ZAG on inflammation. These results will provide new insight for research on anti-inflammation.
Collapse
Affiliation(s)
- Jun Guo
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yanfei Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Ruqian Zhao
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Xiaojing Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
22
|
Fock E, Lavrova E, Bachteeva V, Nikolaeva S, Parnova R. Suppression of fatty acid β-oxidation and energy deficiency as a cause of inhibitory effect of E. coli lipopolysaccharide on osmotic water transport in the frog urinary bladder. Comp Biochem Physiol C Toxicol Pharmacol 2019; 218:81-87. [PMID: 30660802 DOI: 10.1016/j.cbpc.2019.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 12/29/2018] [Accepted: 01/02/2019] [Indexed: 12/23/2022]
Abstract
Previously we showed that arginine-vasotocin (AVT)-stimulated osmotic water permeability (OWP) of the frog urinary bladder was decreased if the mucosal side of the bladder has been naturally colonized by Gram-negative bacteria, or if bacterial lipopolysaccharide (LPS) was introduced into the lumen of the isolated bladder (J. Exp. Zool., 2013, 319, 487-494). Taking into account that in different tissues and cell types, challenge with LPS causes significant metabolic shift and energy deficiency, we hypothesized that an LPS-induced decrease of AVT-stimulated OWP could depend on the reduction of fatty acid oxidation (FAO), which is important for generation of ATP in epithelia. Using an isolated frog Rana temporaria urinary bladder we showed that the AVT-induced increase of OWP did not depend on the external glucose, but was inhibited by oligomycin, an ATP-synthase inhibitor, and by etomoxir, an inhibitor of carnitine palmitoyltransferase-1. In primary cultured epithelial cells isolated from the bladder mucosa, LPS E. coli (25 μg/ml, 21 h), as well as etomoxir (100 μM), decreased FAO accompanied by triacylglycerol accumulation. Both drugs impaired mitochondrial functions demonstrated by decreased ATP production and a reduced maximal oxygen consumption rate (OCR) and OCR directed at ATP synthesis. Additionally, we found that LPS decreased the expression of peroxisome proliferator-activated receptor alpha, a key player in the regulation of FAO. These data indicate that the impairment of AVT-induced water transport in osmoregulatory epithelium caused by LPS depends at least partly on defects in FAO and FAO-dependent energy production.
Collapse
Affiliation(s)
- Ekaterina Fock
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 Saint-Petersburg, Russia
| | - Elena Lavrova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 Saint-Petersburg, Russia
| | - Vera Bachteeva
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 Saint-Petersburg, Russia
| | - Svetlana Nikolaeva
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 Saint-Petersburg, Russia
| | - Rimma Parnova
- I.M. Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, 194223 Saint-Petersburg, Russia.
| |
Collapse
|
23
|
Elkahloun AG, Rodriguez Y, Alaiyed S, Wenzel E, Saavedra JM. Telmisartan Protects a Microglia Cell Line from LPS Injury Beyond AT1 Receptor Blockade or PPARγ Activation. Mol Neurobiol 2018; 56:3193-3210. [PMID: 30105672 DOI: 10.1007/s12035-018-1300-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 08/02/2018] [Indexed: 01/12/2023]
Abstract
The Angiotensin II Receptor Blocker (ARB) Telmisartan reduces inflammation through Angiotensin II AT1 receptor blockade and peroxisome proliferator-activated receptor gamma (PPARγ) activation. However, in a mouse microglia-like BV2 cell line, imitating primary microglia responses with high fidelity and devoid of AT1 receptor gene expression or PPARγ activation, Telmisartan reduced gene expression of pro-injury factors, enhanced that of anti-inflammatory genes, and prevented LPS-induced increase in inflammatory markers. Using global gene expression profiling and pathways analysis, we revealed that Telmisartan normalized the expression of hundreds of genes upregulated by LPS and linked with inflammation, apoptosis and neurodegenerative disorders, while downregulating the expression of genes associated with oncological, neurodegenerative and viral diseases. The PPARγ full agonist Pioglitazone had no neuroprotective effects. Surprisingly, the PPARγ antagonists GW9662 and T0070907 were neuroprotective and enhanced Telmisartan effects. GW9226 alone significantly reduced LPS toxic effects and enhanced Telmisartan neuroprotection, including downregulation of pro-inflammatory TLR2 gene expression. Telmisartan and GW9662 effects on LPS injury negatively correlated with pro-inflammatory factors and upstream regulators, including TLR2, and positively with known neuroprotective factors and upstream regulators. Gene Set Enrichment Analysis (GSEA) of the Telmisartan and GW9662 data revealed negative correlations with sets of genes associated with neurodegenerative and metabolic disorders and toxic treatments in cultured systems, while demonstrating positive correlations with gene sets associated with neuroprotection and kinase inhibition. Our results strongly suggest that novel neuroprotective effects of Telmisartan and GW9662, beyond AT1 receptor blockade or PPARγ activation, include downregulation of the TLR2 signaling pathway, findings that may have translational relevance.
Collapse
Affiliation(s)
- Abdel G Elkahloun
- Microarray Core, Cancer Genetics and Comparative Genomics Branch, National Human Genome Research Institute, National Institutes of Health, 50 South Dr, MSC 4435, Bethesda, MD, 20892-4435, USA
| | - Yara Rodriguez
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA
| | - Seham Alaiyed
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA
| | - Erin Wenzel
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA
| | - Juan M Saavedra
- Laboratory of Neuroprotection, Department of Pharmacology and Physiology, Georgetown University Medical Center, SE402 Med/Dent, 3900 Reservoir Road, Washington, DC, 20057, USA.
| |
Collapse
|
24
|
Li J, Hu SB, He YM, Zhuo CF, Zhou RL, Chen F, Li HY, Deng ZY. 9c11tCLA modulates 11t18:1 and 9t18:1 induced inflammations differently in human umbilical vein endothelial cells. Sci Rep 2018; 8:1535. [PMID: 29367652 PMCID: PMC5784167 DOI: 10.1038/s41598-018-19729-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 01/04/2018] [Indexed: 02/02/2023] Open
Abstract
Endothelial inflammation is recognized as the initial stage of a multistep process leading to coronary heart disease (CHD). Recently, the different effects of industrial trans fatty acids (elaidic acid, 9t18:1) and ruminant trans fatty acids (vaccenic acid, 11t18:1) on CHD have been reported in epidemiological and animal studies, however, the mechanism was not fully studied. Therefore, the objective of this study was to explore the underlying mechanism by which 9t18:1 and 11t18:1 affect human umbilical vein endothelial cells (HUVECs) inflammation. We found that 9c11t-CLA modulated the inflammation of HUVECs induced by 9t18:1 and 11t18:1. Fatty acid composition, pro-inflammatory factors, phosphorylation of MAPKs, and the TLR4 level in HUVECs altered by 11t18:1 induction, collectively suggest that the bio-conversion of 11t18:1 to 9c11tCLA might be the cause why 11t18:1 and 9t18:1 have distinct influences on endothelial injuries. It was concluded that it is biosynthesis of 9c11t CLA from11t18:1, and the modulation of TLR4-MAPK pathway by 9c11t CLA, which at least partially account for the slight effect of 11t18:1 on endothelial inflammation.
Collapse
Affiliation(s)
- Jing Li
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Sheng-Ben Hu
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Yue-Ming He
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Cheng-Fei Zhuo
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Ruo-Lin Zhou
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Fang Chen
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Hong-Yan Li
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China
| | - Ze-Yuan Deng
- State Key Lab of Food Science and Technology, Nanchang University, Nanchang, 330047, China. .,Institute for Advanced Study, Nanchang University, Nanchang, 330031, China.
| |
Collapse
|
25
|
Tan Y, Zhang H, Guo D, Wang J, Yuan X, Yuan Z. Adipophilin Involved in Lipopolysaccharide-Induced Inflammation in RAW264.7 Cell via Extracellular Signal-Regulated Kinase 1/2-Peroxisome Proliferator-Activated Receptor Gamma Pathway. DNA Cell Biol 2017; 36:1159-1167. [DOI: 10.1089/dna.2017.3706] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Affiliation(s)
- Yanmei Tan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical School, University of South China, Hengyang, China
- Department of Pathology, Changde Vocational Technical College, Changde, China
| | - Hai Zhang
- Department of Pathology, The First Affiliated Hospital of University of South China, Hengyang, China
| | - Dongming Guo
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical School, University of South China, Hengyang, China
| | - Jiangbo Wang
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical School, University of South China, Hengyang, China
| | - Xu Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical School, University of South China, Hengyang, China
| | - Zhonghua Yuan
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Medical School, University of South China, Hengyang, China
| |
Collapse
|
26
|
Ma SC, Hao YJ, Jiao Y, Wang YH, Xu LB, Mao CY, Yang XL, Yang AN, Tian J, Zhang MH, Jin SJ, Xu H, Jiang YD, Zhang HP. Homocysteine‑induced oxidative stress through TLR4/NF‑κB/DNMT1‑mediated LOX‑1 DNA methylation in endothelial cells. Mol Med Rep 2017; 16:9181-9188. [PMID: 29039510 DOI: 10.3892/mmr.2017.7753] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Accepted: 08/22/2017] [Indexed: 11/05/2022] Open
Abstract
Atherosclerosis (AS) is a progressive disease of multifactorial origin, which occurs in response to endothelial injury. Increased homocysteine (Hcy) is considered a major cause of endothelial dysfunction, oxidative stress and DNA methylation; however, the mechanisms remain to be fully elucidated. The aim of the present study was to investigate whether Hcy causes injury to endothelial cells (ECs) by the effect of lectin‑like oxidized‑low density lipoprotein receptor‑1 (LOX‑1) DNA methylation through toll‑like receptor 4(TLR4)/nuclear factor (NF)‑κB/DNA methyltransferase (DNMT)1. The ECs were treated with different concentrations of Hcy, and it was found that Hcy promoted the expression of TLR4, leading to EC injury. The effect of oxidative stress was analyzed by measuring superoxide dismutase, malondialdehyde and hydrogen peroxide in the ECs. In addition, the association between NF‑κB and DNMT1 was examined by treatment of the ECs with pyrrolidine dithiocarbamate (PDTC). The results suggested that Hcy induced LOX‑1 DNA hypomethyaltion to promote the expression levels of LOX‑1. Taken together, Hcy injured the ECs through the effect of methylation and trans‑sulfuration metabolism of LOX‑1 through TLR4/NF‑κB/DNMT1. Following injury to the ECs, lipids, particularly ox‑LDL, accumulated in the sub‑endothelial layer to promote the formation of AS.
Collapse
Affiliation(s)
- Sheng-Chao Ma
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yin-Ju Hao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yun Jiao
- Department of Infectious Diseases, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yan-Hua Wang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ling-Bo Xu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Cai-Yan Mao
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Xiao-Ling Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - An-Ning Yang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Jue Tian
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Ming-Hao Zhang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Shao-Ju Jin
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Hua Xu
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Yi-Deng Jiang
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| | - Hui-Ping Zhang
- Department of Prenatal Diagnosis Center, General Hospital of Ningxia Medical University, Yinchuan, Ningxia 750004, P.R. China
| |
Collapse
|
27
|
Zhao H, Han T, Hong X, Sun D. Adipose differentiation‑related protein knockdown inhibits vascular smooth muscle cell proliferation and migration and attenuates neointima formation. Mol Med Rep 2017; 16:3079-3086. [PMID: 28713961 PMCID: PMC5548019 DOI: 10.3892/mmr.2017.6997] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2016] [Accepted: 04/06/2017] [Indexed: 12/31/2022] Open
Abstract
Vascular smooth muscle cells (VSMCs) have an important role in atherosclerosis development. Evidence has demonstrated that adipose differentiation-related protein (ADRP) is associated with foam cell formation and atherosclerosis progression. However, to the best of our knowledge, no previous studies have investigated the role of ADRP knockdown in platelet-derived growth factor (PDGF)-stimulated proliferation and migration of VSMCs in vitro. Furthermore, the effect of ADRP knockdown on neointima formation in vivo remains unclear. In the present study, primary human aortic VSMCs were incubated with PDGF following ADRP small interfering (si)RNA transfection. Cell viability, migration and cell cycle distribution were analyzed by MTT, wound healing and Transwell assays and flow cytometry, respectively. Extracellular signal-regulated kinase (ERK), phosphorylated (p)-ERK, Akt, p-Akt, proliferating cell nuclear antigen (PCNA), matrix metalloproteinase (MMP)-2 and MMP-9 protein levels were determined by western blotting. Apolipoprotein E−/− mice fed an atherogenic diet were injected with siADRP or control siRNA twice a week. After 3 weeks of therapy, aortas were excised and ADRP mRNA and protein expression was determined. Neointima formation was assessed by hematoxylin and eosin staining. The results of the current study demonstrated that ADRP knockdown significantly inhibited PDGF-induced increases in VSMC viability, caused G1 phase cell cycle arrest and decreased PCNA expression. Knockdown of ADRP inhibited PDGF-induced migration of VSMCs by reducing MMP protein expression and activity. In addition, the present study also demonstrated that ADRP knockdown inhibited ERK and Akt signaling pathways in response to PDGF. Furthermore, siADRP administration suppressed neointima formation in the mouse model. The results of the present study indicate that ADRP may be a potential target for the treatment of atherosclerosis.
Collapse
Affiliation(s)
- Haomin Zhao
- Department of Vascular Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Tao Han
- Department of Vascular Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Xin Hong
- Department of Vascular Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| | - Dajun Sun
- Department of Vascular Surgery, China‑Japan Union Hospital of Jilin University, Changchun, Jilin 130033, P.R. China
| |
Collapse
|
28
|
Balistreri CR, Ruvolo G, Lio D, Madonna R. Toll-like receptor-4 signaling pathway in aorta aging and diseases: "its double nature". J Mol Cell Cardiol 2017; 110:38-53. [PMID: 28668304 DOI: 10.1016/j.yjmcc.2017.06.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/20/2017] [Accepted: 06/27/2017] [Indexed: 12/20/2022]
Abstract
Recent advances in the field of innate immunity have revealed a complex role of innate immune signaling pathways in both tissue homeostasis and disease. Among them, the Toll-like receptor 4 (TLR-4) pathways has been linked to various pathophysiological conditions, such as cardiovascular diseases (CVDs). This has been interrogated by developing multiple laboratory tools that have shown in animal models and clinical conditions, the involvement of the TLR-4 signaling pathway in the pathophysiology of different CVDs, such as atherosclerosis, ischemic heart disease, heart failure, ischemia-reperfusion injury and aorta aneurysm. Among these, aorta aneurysm, a very complex pathological condition with uncertain etiology and fatal complications (i.e. dissection and rupture), has been associated with the occurrence of high risk cardiovascular conditions, including thrombosis and embolism. In this review, we discuss the possible role of TLR-4 signaling pathway in the development of aorta aneurysm, considering the emerging evidence from ongoing investigations. Our message is that emphasizing the role of TLR-4 signaling pathway in aorta aneurysm may serve as a starting point for future studies, leading to a better understanding of the pathophysiological basis and perhaps the effective treatment of this difficult human disease.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Giovanni Ruvolo
- Department of Cardiac Surgery, University of Rome 'Tor Vergata', Rome, Italy
| | - Domenico Lio
- Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy
| | - Rosalinda Madonna
- Heart Failure Research, Texas Heart Institute, St. Luke's Episcopal Hospital, Houston, TX, United States; Department of Internal Medicine, Cardiology, The University of Texas Health Science Center at Houston, Houston, TX, United States; Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences "G. D'Annunzio" University, 66100 Chieti, Italy
| |
Collapse
|
29
|
Endotoxemia accelerates diaphragm dysfunction in ventilated rabbits. J Surg Res 2016; 206:507-516. [PMID: 27884349 DOI: 10.1016/j.jss.2016.08.023] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Revised: 06/13/2016] [Accepted: 08/04/2016] [Indexed: 01/30/2023]
Abstract
BACKGROUND Ventilators may induce diaphragm dysfunction, and most of the septic population who are admitted to the intensive care unit require mechanical ventilation. However, there is no evidence that sepsis accelerates the onset of ventilator-induced diaphragm dysfunction or affects the microcirculation. Our study investigated whether lipopolysaccharide (LPS)-induced endotoxemia accelerated diaphragm dysfunction in ventilated rabbits by evaluating microcirculation, lipid accumulation, and diaphragm contractility. METHODS After anesthesia and tracheostomy, 25 invasively monitored and mechanically ventilated New Zealand white rabbits were randomized to control (n = 5), controlled mechanical ventilation (CMV) (n = 5), pressure support ventilation (PSV; n = 5), CMV or PSV with LPS-induced endotoxemia (CMV-LPS and PSV-LPS, respectively; n = 5 for each). Rabbits were anesthetized and ventilated for 24 h, except the control rabbits (30 min). Diaphragmatic contractility was evaluated using neuromechanical and neuroventilatory efficiency. We evaluated the following at the end of the protocol: (1) diaphragm microcirculation; (2) lipid accumulation; and (3) diaphragm muscular fibers structure. RESULTS Diaphragm contractility, microcirculation, lipid accumulation, and fiber structures were severely compromised in endotoxemic animals after 24 h compared to nonendotoxemic rabbits. Moreover, a slight but significant increase in lipid accumulation was observed in CMV and PSV groups compared with controls (P < 0.05). CONCLUSIONS Endotoxemia accelerates the diaphragm dysfunction process in ventilated rabbits, affects the microcirculation, and results in diaphragmatic lipid accumulation and contractility impairment.
Collapse
|
30
|
Qi M, Zheng L, Qi Y, Han X, Xu Y, Xu L, Yin L, Wang C, Zhao Y, Sun H, Liu K, Peng J. Dioscin attenuates renal ischemia/reperfusion injury by inhibiting the TLR4/MyD88 signaling pathway via up-regulation of HSP70. Pharmacol Res 2015; 100:341-352. [PMID: 26348276 DOI: 10.1016/j.phrs.2015.08.025] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Revised: 08/28/2015] [Accepted: 08/31/2015] [Indexed: 11/21/2022]
Abstract
We previously reported the effect of dioscin against hepatic ischemia/reperfusion injury (IRI) in rats. However, little is known concerning the role of dioscin in renal IRI. In the present study, rats were subjected to IRI and dioscin was intragastrically administered for seven consecutive days before surgery. In vitro models of hypoxia/reoxygenation were developed in NRK-52E and HK-2 cells, which were prophylactically treated with or without dioscin. The results showed that dioscin significantly decreased serum BUN and Cr levels, and markedly attenuated cell injury. Mechanistic studies showed that dioscin significantly increased HSP70 levels, decreased the levels of TLR4, MyD88, TRAF6, COX-2, JNK, ERK and p38 MAPK phosphorylation, suppressed the nuclear translocation of NF-κB and HMGB1, and subsequently decreased the mRNA levels of IL-1β, IL-6, TNF-α, ICAM-1 and IFN-γ. Moreover, HSP70 siRNA or TLR4 DNA reversed the nephroprotective effects of dioscin, while dioscin still significantly down-regulated the TLR4 signaling pathway. Furthermore, by inhibiting MyD88 with ST2825 (a MyD88 inhibitor), renal IRI was significantly attenuated, suggesting that the effect of dioscin against renal IRI depended on MyD88. Our results suggested that dioscin had a potent effect against renal IRI through suppressing the TLR4/MyD88 signaling pathway by up-regulating HSP70. These data provide new insights for investigating the natural product with the nephroprotective effect against IRI, which should be developed as a new therapeutic agent for the treatment of acute kidney injury in the future.
Collapse
Affiliation(s)
- Meng Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lingli Zheng
- Department of Pharmaceuticals, The First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Yan Qi
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Xu Han
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Youwei Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lina Xu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Lianhong Yin
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Changyuan Wang
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Yanyan Zhao
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Huijun Sun
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Kexin Liu
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China
| | - Jinyong Peng
- College of Pharmacy, Dalian Medical University, Western 9 Lvshunnan Road, Dalian 116044, China.
| |
Collapse
|
31
|
Bai X, Qi Z, Song G, Zhao X, Zhao H, Meng X, Liu C, Bing W, Bi Y. Effects of Monocyte Chemotactic Protein-1 and Nuclear Factor of Kappa B Pathway in Rejection of Cardiac Allograft in Rat. Transplant Proc 2015; 47:2010-6. [DOI: 10.1016/j.transproceed.2015.05.014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 04/26/2015] [Accepted: 05/14/2015] [Indexed: 01/04/2023]
|
32
|
Luo H, Wang J, Qiao C, Zhang X, Zhang W, Ma N. ATF3 Inhibits Tenascin-C-induced Foam Cell Formation in LPS-Stimulated THP-1 Macrophages by Suppressing TLR-4. J Atheroscler Thromb 2015; 22:1214-23. [PMID: 26133317 DOI: 10.5551/jat.28415] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
AIM Efficiently inhibiting the formation of macrophage foam cells is indispensable for mitigating and treating atherosclerosis. Tenascin-C (TN-C) plays an important role in promoting atherosclerosis; therefore, it is essential to inhibit foam cell formation associated with TN-C for controlling atherosclerosis. Activating transcription factor 3 (ATF3) is one of the factors involved in regulating the complex process of foam cell formation. This study aimed to explore the role of TN-C and ATF3 in LPS-stimulated THP-1-derived macrophages. METHODS RT-PCR was used for evaluating the expression of TN-C in LPS-stimulated THP-1 macrophages. Further, exogenous TN-C was introduced and incubated with cultured THP-1 macrophages to confirm the effect of TN-C on LPS-stimulated THP-1 macrophages. ATF3-modified THP-1 macrophages were constructed and verified by western blot. High performance liquid chromatography (HPLC) assay and Oil red O staining were applied for detecting cholesteryl ester/total cholesterol (CE/TC) and lipid formation in THP-1 macrophages. RESULTS The expression of TN-C was determined to be upregulated in LPS-stimulated THP-1 macrophages in a dose- and time-dependent manner. HPLC assay and Oil red O staining confirmed that TN-C can enhance LPS-induced THP-1 macrophage foam cell formation. Moreover, ATF3 can act as a negative regulatory factor for inhibiting TN-C-induced foam cell formation by suppressing TLR-4 in LPS-stimulated THP-1 macrophages. CONCLUSION ATF3 can inhibit TN-C-induced foam cell formation in LPS-stimulated THP-1 macrophages by suppressing TLR-4. It may be a useful molecular target to control TN-C-induced foam cell formation in atherosclerosis.
Collapse
Affiliation(s)
- Hong Luo
- Key Disciplines Laboratory Clinical-Medicine Henan, Department of Cardiovascular Surgery, the First Affiliated Hospital of Zhengzhou University
| | | | | | | | | | | |
Collapse
|
33
|
Yu XH, Zheng XL, Tang CK. Nuclear Factor-κB Activation as a Pathological Mechanism of Lipid Metabolism and Atherosclerosis. Adv Clin Chem 2015; 70:1-30. [PMID: 26231484 DOI: 10.1016/bs.acc.2015.03.004] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Atherosclerosis is a chronic inflammatory disease of the arterial wall with lipid-laden lesions, involving a complex interaction between multiple different cell types and cytokine networks. Inflammatory responses mark all stages of atherogenesis: from lipid accumulation in the intima to plaque formation and eventual rupture. One of the most important regulators of inflammation is the transcription factor nuclear factor-κB (NF-κB), which is activated through the canonical and noncanonical pathways in response to various stimuli. NF-κB has long been regarded as a proatherogenic factor, because it is implicated in multiple pathological processes during atherogenesis, including foam cell formation, vascular inflammation, proliferation of vascular smooth muscle cells, arterial calcification, and plaque progression. In contrast, inhibition of NF-κB signaling has been shown to protect against atherosclerosis. This chapter aims to discuss recent progress on the roles of NF-κB in lipid metabolism and atherosclerosis and also to highlight its potential therapeutic benefits.
Collapse
Affiliation(s)
- Xiao-Hua Yu
- Key Laboratory for Atherosclerology of Hunan Province, Molecular Target New Drug Discovery and Cooperative Innovation Center of Hunan Province, Life Science Research Center, University of South China, Hengyang, PR China
| | - Xi-Long Zheng
- Department of Biochemistry and Molecular Biology, The Libin Cardiovascular Institute of Alberta, Cumming School of Medicine, The University of Calgary, Health Sciences Center, Calgary, Alberta, Canada
| | - Chao-Ke Tang
- Key Laboratory for Atherosclerology of Hunan Province, Molecular Target New Drug Discovery and Cooperative Innovation Center of Hunan Province, Life Science Research Center, University of South China, Hengyang, PR China.
| |
Collapse
|
34
|
Yuan M, Fu H, Ren L, Wang H, Guo W. Soluble CD40 ligand promotes macrophage foam cell formation in the etiology of atherosclerosis. Cardiology 2015; 131:1-12. [PMID: 25825037 DOI: 10.1159/000374105] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 01/08/2015] [Indexed: 11/19/2022]
Abstract
OBJECTIVE High levels of soluble CD40 ligand (sCD40L) in the circulation have been suggested as an important indicator of cardiovascular diseases such as atherosclerosis and acute coronary syndromes. In the present study, we explored the role of sCD40L in the formation of foam cells. METHODS Lipid deposition and foam cell formation was measured by high-performance liquid chromatography and Nile Red staining, respectively. Gene expressions were detected by quantitative real-time PCR and Western blot analysis. The interaction between CD40 and sCD40L were blocked by CD40 small interfering RNA or anti-CD40 antibody. RESULTS sCD40L significantly increased lipid deposition and foam cell formation associated with upregulation of scavenger receptor type A and CD36. Additionally, sCD40L increased adipocyte enhancer-binding protein 1 and cholesterol efflux, and activated NF-κB in macrophages. sCD40L promoted foam cell formation via CD40 ligation and disruption of the ligation between CD40 and CD40L either by small interfering RNA or by a blocking anti-CD40 antibody apparently inhibiting foam cell formation in response to sCD40L. CONCLUSION Our data suggests a novel insight into the role of sCD40L in foam cell formation during atherosclerosis, which further confirms the importance of sCD40L in atherosclerosis and as a target for the treatment of this disease.
Collapse
Affiliation(s)
- Ming Yuan
- Department of Cardiology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | | | | | | |
Collapse
|
35
|
Jia SJ, Niu PP, Cong JZ, Zhang BK, Zhao M. TLR4 signaling: A potential therapeutic target in ischemic coronary artery disease. Int Immunopharmacol 2014; 23:54-9. [DOI: 10.1016/j.intimp.2014.08.011] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/06/2014] [Accepted: 08/13/2014] [Indexed: 01/12/2023]
|
36
|
Critical role of TLR2 and MyD88 for functional response of macrophages to a group IIA-secreted phospholipase A2 from snake venom. PLoS One 2014; 9:e93741. [PMID: 24718259 PMCID: PMC3981733 DOI: 10.1371/journal.pone.0093741] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 03/06/2014] [Indexed: 01/10/2023] Open
Abstract
The snake venom MT-III is a group IIA secreted phospholipase A2 (sPLA2) enzyme with functional and structural similarities with mammalian pro-inflammatory sPLA2s of the same group. Previously, we demonstrated that MT-III directly activates the innate inflammatory response of macrophages, including release of inflammatory mediators and formation of lipid droplets (LDs). However, the mechanisms coordinating these processes remain unclear. In the present study, by using TLR2−/− or MyD88−/− or C57BL/6 (WT) male mice, we report that TLR2 and MyD88 signaling have a critical role in MT-III-induced inflammatory response in macrophages. MT-III caused a marked release of PGE2, PGD2, PGJ2, IL-1β and IL-10 and increased the number of LDs in WT macrophages. In MT-III-stimulated TLR2−/− macrophages, formation of LDs and release of eicosanoids and cytokines were abrogated. In MyD88−/− macrophages, MT-III-induced release of PGE2, IL-1β and IL-10 was abrogated, but release of PGD2 and PGJ2 was maintained. In addition, COX-2 protein expression seen in MT-III-stimulated WT macrophages was abolished in both TLR2−/− and MyD88−/− cells, while perilipin 2 expression was abolished only in MyD88−/− cells. We further demonstrated a reduction of saturated, monounsaturated and polyunsaturated fatty acids and a release of the TLR2 agonists palmitic and oleic acid from MT-III-stimulated WT macrophages compared with WT control cells, thus suggesting these fatty acids as major messengers for MT-III-induced engagement of TLR2/MyD88 signaling. Collectively, our findings identify for the first time a TLR2 and MyD88-dependent mechanism that underlies group IIA sPLA2-induced inflammatory response in macrophages.
Collapse
|
37
|
Wang X, Li L, Niu X, Dang X, Li P, Qu L, Bi X, Gao Y, Hu Y, Li M, Qiao W, Peng Z, Pan L. mTOR enhances foam cell formation by suppressing the autophagy pathway. DNA Cell Biol 2014; 33:198-204. [PMID: 24512183 PMCID: PMC3967384 DOI: 10.1089/dna.2013.2164] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2011] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 02/05/2023] Open
Abstract
Recently, autophagy has drawn more attention in cardiovascular disease as it has important roles in lipid metabolism. Mammalian target of rapamycin (mTOR) is a key regulator of autophagy; however, its effect on atherosclerosis and the underlying mechanism remains undefined. In this study, an obvious upregulation of mTOR and p-mTOR protein was observed in macrophage-derived foam cells. Blocking mTOR expression with specific small interference RNA (siRNA) dramatically suppressed foam cell formation, accompanied by a decrease of lipid deposition. Further mechanistic analysis indicated that suppressing mTOR expression significantly upregulated autophagic marker LC3 expression and downregulated autophagy substrate p62 levels, indicating that mTOR silencing triggered autophagosome formation. Moreover, blocking mTOR expression obviously accelerated neutral lipid delivery to lysosome and cholesterol efflux from foam cells, implying that mTOR could induce macrophage foam cell formation by suppressing autophagic pathway. Further, mTOR silencing significantly upregulated ULK1 expression, which was accounted for mTOR-induced foam cell formation via autophagic pathway as treatment with ULK1 siRNA dampened LC3-II levels and increased p62 expression, concomitant with lipid accumulation and decreased cholesterol efflux from foam cells. Together, our data provide an insight into how mTOR accelerates the pathological process of atherosclerosis. Accordingly, blocking mTOR levels may be a promising therapeutic agent against atherosclerotic complications.
Collapse
Affiliation(s)
- Xiaochuang Wang
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Lingxia Li
- The Cadre Ward, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiaolin Niu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiaoyan Dang
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Ping Li
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Li Qu
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Xiaoju Bi
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yanxia Gao
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Yanfen Hu
- The Cadre Ward, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Manxiang Li
- Department of Respiratory Diseases, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Wanhai Qiao
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Zhuo Peng
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Longfei Pan
- Department of Emergency Medicine, The Second Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an, People's Republic of China
| |
Collapse
|
38
|
Dural fibroblasts play a potential role in headache pathophysiology. Pain 2014; 155:1238-1244. [PMID: 24657451 DOI: 10.1016/j.pain.2014.03.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 03/16/2014] [Accepted: 03/17/2014] [Indexed: 12/24/2022]
Abstract
Nociceptive signaling from the meninges is proposed to contribute to many forms of headache. However, the events within the meninges that drive afferent activity are not clear. Meningeal fibroblasts are traditionally thought to produce extracellular proteins that constitute the meninges but not to contribute to headache. The purpose of these studies was to determine whether dural fibroblasts release factors that activate/sensitize dural afferents and produce headache-like behavior in rats. Dura mater was removed from male rats and dural fibroblasts were cultured. Fibroblast cultures were stimulated with vehicle or lipopolysaccharide (LPS), washed, and conditioned media was collected. Fibroblast media conditioned with vehicle or LPS was applied to retrogradely labeled rat dural trigeminal ganglion neurons in vitro. Patch-clamp electrophysiology was performed to determine whether conditioned media activated/sensitized dural afferents. A preclinical behavioral model was used where conditioned media was applied directly to the rat dura to determine the presence of cutaneous facial and hind-paw allodynia. Conditioned media was also tested for interleukin-6 (IL-6) content using an enzyme-linked immunosorbent assay. Application of LPS-conditioned fibroblast media to dural afferents produced a significant increase in action potential firing as well as cutaneous facial and hind-paw allodynia when this media was applied to the dura. Finally, stimulation of cultured fibroblasts with LPS increased IL-6 levels in the media. These findings demonstrate that fibroblasts stimulated with LPS release factors capable of activating/sensitizing dural afferents. Further, they suggest that fibroblasts play a potential role in the pathophysiology of headache.
Collapse
|
39
|
DONG WENPENG, WANG XIANYUE, BI SHENGHUI, PAN ZHIGUO, LIU SHENXI, YU HAO, LU HUA, LIN XI, WANG XIAOWU, MA TAO, ZHANG WEIDA. Inhibitory effects of resveratrol on foam cell formation are mediated through monocyte chemotactic protein-1 and lipid metabolism-related proteins. Int J Mol Med 2014; 33:1161-8. [DOI: 10.3892/ijmm.2014.1680] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Accepted: 02/03/2014] [Indexed: 11/06/2022] Open
|
40
|
Feng J, Li A, Deng J, Yang Y, Dang L, Ye Y, Li Y, Zhang W. miR-21 attenuates lipopolysaccharide-induced lipid accumulation and inflammatory response: potential role in cerebrovascular disease. Lipids Health Dis 2014; 13:27. [PMID: 24502419 PMCID: PMC3922422 DOI: 10.1186/1476-511x-13-27] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2013] [Accepted: 01/02/2014] [Indexed: 12/23/2022] Open
Abstract
Background Atherosclerosis constitutes the leading contributor to morbidity and mortality in cardiovascular and cerebrovascular diseases. Lipid deposition and inflammatory response are the crucial triggers for the development of atherosclerosis. Recently, microRNAs (miRNAs) have drawn more attention due to their prominent function on inflammatory process and lipid accumulation in cardiovascular and cerebrovascular disease. Here, we investigated the involvement of miR-21 in lipopolysaccharide (LPS)-induced lipid accumulation and inflammatory response in macrophages. Methods After stimulation with the indicated times and doses of LPS, miR-21 mRNA levels were analyzed by Quantitative real-time PCR. Following transfection with miR-21 or anti-miR-21 inhibitor, lipid deposition and foam cell formation was detected by high-performance liquid chromatography (HPLC) and Oil-red O staining. Furthermore, the inflammatory cytokines interleukin 6 (IL-6) and interleukin 10 (IL-10) were evaluated by Enzyme-linked immunosorbent assay (ELISA) assay. The underlying molecular mechanism was also investigated. Results In this study, LPS induced miR-21 expression in macrophages in a time- and dose-dependent manner. Further analysis confirmed that overexpression of miR-21 by transfection with miR-21 mimics notably attenuated lipid accumulation and lipid-laden foam cell formation in LPS-stimulated macrophages, which was reversely up-regulated when silencing miR-21 expression via anti-miR-21 inhibitor transfection, indicating a reverse regulator of miR-21 in LPS-induced foam cell formation. Further mechanism assays suggested that miR-21 regulated lipid accumulation by Toll-like receptor 4 (TLR4) and nuclear factor-κB (NF-κB) pathway as pretreatment with anti-TLR4 antibody or a specific inhibitor of NF-κB (PDTC) strikingly dampened miR-21 silence-induced lipid deposition. Additionally, overexpression of miR-21 significantly abrogated the inflammatory cytokines secretion of IL-6 and increased IL-10 levels, the corresponding changes were also observed when silencing miR-21 expression, which was impeded by preconditioning with TLR4 antibody or PDTC. Conclusions Taken together, these results corroborated that miR-21 could negatively regulate LPS-induced lipid accumulation and inflammatory responses in macrophages by the TLR4-NF-κB pathway. Accordingly, our research will provide a prominent insight into how miR-21 reversely abrogates bacterial infection-induced pathological processes of atherosclerosis, indicating a promising therapeutic prospect for the prevention and treatment of atherosclerosis by miR-21 overexpression.
Collapse
Affiliation(s)
- Jun Feng
- Department of Cerebral vessels, First Affiliated Hospital of Medical College, Xi'an Jiaotong University, Xi'an 710061, China.
| | | | | | | | | | | | | | | |
Collapse
|