1
|
Rahaman MH, Thygesen SJ, Maxwell MJ, Kim H, Mudai P, Nanson JD, Jia X, Vajjhala PR, Hedger A, Vetter I, Haselhorst T, Robertson AAB, Dymock B, Ve T, Mobli M, Stacey KJ, Kobe B. o-Vanillin binds covalently to MAL/TIRAP Lys-210 but independently inhibits TLR2. J Enzyme Inhib Med Chem 2024; 39:2313055. [PMID: 38416868 PMCID: PMC10903754 DOI: 10.1080/14756366.2024.2313055] [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: 08/11/2023] [Accepted: 01/28/2024] [Indexed: 03/01/2024] Open
Abstract
Toll-like receptor (TLR) innate immunity signalling protects against pathogens, but excessive or prolonged signalling contributes to a range of inflammatory conditions. Structural information on the TLR cytoplasmic TIR (Toll/interleukin-1 receptor) domains and the downstream adaptor proteins can help us develop inhibitors targeting this pathway. The small molecule o-vanillin has previously been reported as an inhibitor of TLR2 signalling. To study its mechanism of action, we tested its binding to the TIR domain of the TLR adaptor MAL/TIRAP (MALTIR). We show that o-vanillin binds to MALTIR and inhibits its higher-order assembly in vitro. Using NMR approaches, we show that o-vanillin forms a covalent bond with lysine 210 of MAL. We confirm in mouse and human cells that o-vanillin inhibits TLR2 but not TLR4 signalling, independently of MAL, suggesting it may covalently modify TLR2 signalling complexes directly. Reactive aldehyde-containing small molecules such as o-vanillin may target multiple proteins in the cell.
Collapse
Affiliation(s)
- Md. Habibur Rahaman
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Sara J. Thygesen
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Michael J. Maxwell
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Hyoyoung Kim
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Prerna Mudai
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Jeffrey D. Nanson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Xinying Jia
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Parimala R. Vajjhala
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
| | - Andrew Hedger
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Irina Vetter
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
- School of Pharmacy, University of Queensland, Brisbane, Australia
| | | | - Avril A. B. Robertson
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| | - Brian Dymock
- Queensland Emory Drug Discovery Initiative, University of Queensland, Brisbane, Australia
| | - Thomas Ve
- Institute for Glycomics, Griffith University, Southport, Australia
| | - Mehdi Mobli
- Centre for Advanced Imaging, University of Queensland, Brisbane, Australia
| | - Katryn J. Stacey
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
| | - Bostjan Kobe
- School of Chemistry and Molecular Biosciences, University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, University of Queensland, Brisbane, Australia
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Australia
| |
Collapse
|
2
|
van Tuijl J, van Heck JI, Bahrar H, Broeders W, Wijma J, ten Have YM, Giera M, Zweers-van Essen H, Rodwell L, Joosten LA, Netea MG, Afman LA, Bekkering S, Riksen NP. Single high-fat challenge and trained innate immunity: A randomized controlled cross-over trial. iScience 2024; 27:111103. [PMID: 39493874 PMCID: PMC11530819 DOI: 10.1016/j.isci.2024.111103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 05/11/2024] [Accepted: 10/01/2024] [Indexed: 11/05/2024] Open
Abstract
Brief exposure of monocytes to atherogenic molecules, such as oxidized lipoproteins, triggers a persistent pro-inflammatory phenotype, named trained immunity. In mice, transient high-fat diet leads to trained immunity, which aggravates atherogenesis. We hypothesized that a single high-fat challenge in humans induces trained immunity. In a randomized controlled cross-over study, 14 healthy individuals received a high-fat or reference shake, and blood was drawn before and after 1, 2, 4, 6, 24, and 72 h. Incubation of donor monocytes with the post-high-fat-shake serum induced trained immunity, regulated via Toll-like receptor 4. This was not mediated via triglyceride-rich lipoproteins, C12, 14, and 16, or metabolic endotoxemia. In vivo, however, the high-fat challenge did not affect monocyte phenotype and function. We conclude that a high-fat challenge leads to alterations in the serum composition that have the potential to induce trained immunity in vitro. However, this does not translate into a (persistent) hyperinflammatory monocyte phenotype in vivo.
Collapse
Affiliation(s)
- Julia van Tuijl
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Julia I.P. van Heck
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Harsh Bahrar
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Wieteke Broeders
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Johan Wijma
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Yvonne M. ten Have
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| | - Martin Giera
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZA, the Netherlands
| | - Heidi Zweers-van Essen
- Department of Gastroenterology and Hepatology-Dietetics, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Laura Rodwell
- Section Biostatics, Department of Health Evidence, Radboud University Medical Center, Nijmegen 6525 GA, the Netherlands
| | - Leo A.B. Joosten
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department of Medical Genetics, Iuliu Haţieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
| | - Mihai G. Netea
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Department for Immunology & Metabolism, Life and Medical Sciences Institute (LIMES), University of Bonn, 53115 Bonn, Germany
| | - Lydia A. Afman
- Nutrition, Metabolism and Genomics Group, Division of Human Nutrition and Health, Wageningen University and Research, Wageningen 6700 HB, the Netherlands
| | - Siroon Bekkering
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
- Murdoch Children’s Research Institute, Royal Children’s Hospital, Melbourne, VIC 3052, Australia
| | - Niels P. Riksen
- Department of Internal Medicine, Radboud University Medical Center Nijmegen, Nijmegen 6525 GA, the Netherlands
| |
Collapse
|
3
|
Hoekstra M, Snip OSC, Janusz P, Bernabé Kleijn MNA, Truitt ER, Sullivan BD, Schmidt TA, Jay GD, Van Eck M. Recombinant human proteoglycan 4 lowers inflammation and atherosclerosis susceptibility in female low-density lipoprotein receptor knockout mice. J Physiol 2024; 602:1939-1951. [PMID: 38606903 DOI: 10.1113/jp286354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Recombinant human proteoglycan 4 (rhPRG4) is a macromolecular mucin-like glycoprotein that is classically studied as a lubricant within eyes and joints. Given that endogenously produced PRG4 is present within atherosclerotic lesions and genetic PRG4 deficiency increases atherosclerosis susceptibility in mice, in the current study we investigated the anti-atherogenic potential of chronic rhPRG4 treatment. Female low-density lipoprotein receptor knockout mice were fed an atherogenic Western-type diet for 6 weeks and injected three times per week intraperitoneally with 0.5 mg rhPRG4 or PBS as control. Treatment with rhPRG4 was associated with a small decrease in plasma-free cholesterol levels, without a change in cholesteryl ester levels. A marked increase in the number of peritoneal foam cells was detected in response to the peritoneal rhPRG4 administration, which could be attributed to elevated peritoneal leukocyte MSR1 expression levels. However, rhPRG4-treated mice exhibited significantly smaller aortic root lesions of 278 ± 21 × 103 μm2 compared with 339 ± 15 × 103 μm2 in the aortic root of control mice. The overall decreased atherosclerosis susceptibility coincided with a shift in the monocyte and macrophage polarization states towards the patrolling and anti-inflammatory M2-like phenotypes, respectively. Furthermore, rhPRG4 treatment significantly reduced macrophage gene expression levels as well as plasma protein levels of the pro-inflammatory/pro-atherogenic cytokine TNF-alpha. In conclusion, we have shown that peritoneal administration and subsequent systemic exposure to rhPRG4 beneficially impacts the inflammatory state and reduces atherosclerosis susceptibility in mice. Our findings highlight that PRG4 is not only a lubricant but also acts as an anti-inflammatory agent. KEY POINTS: Endogenously produced proteoglycan 4 is found in atherosclerotic lesions and its genetic deficiency in mice is associated with enhanced atherosclerosis susceptibility. In this study we investigated the anti-atherogenic potential of chronic treatment with recombinant human PRG4 in hypercholesterolaemic female low-density lipoprotein receptor knockout mice. We show that recombinant human PRG4 stimulates macrophage foam cell formation, but also dampens the pro-inflammatory state of monocyte/macrophages, eventually leading to a significant reduction in plasma TNF-alpha levels and a lowered atherosclerosis susceptibility. Our findings highlight that peritoneal recombinant human PRG4 treatment can execute effects both locally and systemically and suggest that it will be of interest to study whether rhPRG4 treatment is also able to inhibit the progression and/or induce regression of previously established atherosclerotic lesions.
Collapse
Affiliation(s)
- Menno Hoekstra
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| | - Olga S C Snip
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Philip Janusz
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | - Mireia N A Bernabé Kleijn
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
| | | | | | - Tannin A Schmidt
- Biomedical Engineering Department, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Gregory D Jay
- Department of Emergency Medicine, Warren Alpert Medical School and Division of Biomedical Engineering, School of Engineering, Brown University, Providence, Rhode Island, USA
| | - Miranda Van Eck
- Division of BioTherapeutics, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Division of Systems Pharmacology and Pharmacy, Leiden Academic Centre for Drug Research, Leiden University, Leiden, The Netherlands
- Pharmacy Leiden, Leiden, The Netherlands
| |
Collapse
|
4
|
Nayak G, Dimitriadis K, Pyrpyris N, Manti M, Kamperidis N, Kamperidis V, Ziakas A, Tsioufis K. Gut Microbiome and Its Role in Valvular Heart Disease: Not a "Gutted" Relationship. Life (Basel) 2024; 14:527. [PMID: 38672797 PMCID: PMC11051562 DOI: 10.3390/life14040527] [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: 03/18/2024] [Revised: 04/07/2024] [Accepted: 04/14/2024] [Indexed: 04/28/2024] Open
Abstract
The role of the gut microbiome (GM) and oral microbiome (OM) in cardiovascular disease (CVD) has been increasingly being understood in recent years. It is well known that GM is a risk factor for various CVD phenotypes, including hypertension, dyslipidemia, heart failure and atrial fibrillation. However, its role in valvular heart disease (VHD) is less well understood. Research shows that, direct, microbe-mediated and indirect, metabolite-mediated damage as a result of gut dysbiosis and environmental factors results in a subclinical, chronic, systemic inflammatory state, which promotes inflammatory cell infiltration in heart valves and subsequently, via pro-inflammatory molecules, initiates a cascade of reaction, resulting in valve calcification, fibrosis and dysfunction. This relationship between GM and VHD adds a pathophysiological link to the pathogenesis of VHD, which can be aimed therapeutically, in order to prevent or regress any risk for valvular pathologies. Therapeutic interventions include dietary modifications and lifestyle interventions, in order to influence environmental factors that can promote gut dysbiosis. Furthermore, the combination of probiotics and prebiotics, as well as fecal m transplantation and targeted treatment with inducers or inhibitors of microbial enzymes have showed promising results in animal and/or clinical studies, with the potential to reduce the inflammatory state and restore the normal gut flora in patients. This review, thus, is going to discuss the pathophysiological links behind the relationship of GM, CVD and VHD, as well as explore the recent data regarding the effect of GM-altering treatment in CVD, cardiac function and systemic inflammation.
Collapse
Affiliation(s)
- Gyanaranjan Nayak
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (G.N.); (N.P.); (K.T.)
| | - Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (G.N.); (N.P.); (K.T.)
| | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (G.N.); (N.P.); (K.T.)
| | - Magdalini Manti
- St Mark’s Hospital, Imperial College London, London HA1 3UJ, UK (N.K.)
| | | | - Vasileios Kamperidis
- First Cardiology Department, AHEPA University Hospital, Medical School, Aristotle University of Thessaloniki, 54453 Thessaloniki, Greece; (V.K.); (A.Z.)
| | - Antonios Ziakas
- First Cardiology Department, AHEPA University Hospital, Medical School, Aristotle University of Thessaloniki, 54453 Thessaloniki, Greece; (V.K.); (A.Z.)
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration General Hospital, 115 27 Athens, Greece; (G.N.); (N.P.); (K.T.)
| |
Collapse
|
5
|
Kwon DH, Hwang J, You H, Kim NY, Lee GY, Han SN. Effects of an in vitro vitamin D treatment on the inflammatory responses in visceral adipose tissue from Ldlr-/- mice. Nutr Res Pract 2024; 18:19-32. [PMID: 38352213 PMCID: PMC10861343 DOI: 10.4162/nrp.2024.18.1.19] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/26/2023] [Accepted: 11/16/2023] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Atherosclerosis is associated with increased inflammation in the visceral adipose tissue (VAT). Vitamin D has been reported to modulate the inflammatory responses of stromal vascular cells (SVCs) and adipocytes in adipose tissue, but the role of vitamin D in atherosclerosis biology is unclear. This study examined the effects of in vitro 1,25-dihydroxyvitamin D3 (1,25[OH]2D3) treatment on the inflammatory responses of SVCs and adipocytes from atherosclerotic mice. MATERIALS/METHODS C57BL/6J (B6) mice were divided randomly into 2 groups and fed a 10% kcal fat control diet (control group, CON) or 41% kcal fat, 0.21% cholesterol (high fat + cholesterol, HFC) diet (obese group, OB), and B6.129S7-Ldlrtm1Her/J (Ldlr-/-) mice were fed a HFC diet (obese with atherosclerosis group, OBA) for 16 weeks. SVCs and adipocytes isolated from VAT were pre-incubated with 1,25(OH)2D3 for 24 h and stimulated with lipopolysaccarides for the next 24 h. Proinflammatory cytokine production by adipocytes and SVCs, the immune cell population in SVCs, and the expression of the genes involved in the inflammatory signaling pathway in SVCs were determined. RESULTS The numbers of total macrophages and SVCs per mouse were higher in OB and OBA groups than the CON group. The in vitro 1,25(OH)2D3 treatment significantly reduced macrophages/SVCs (%) in the OBA group. Consistent with this change, the production of interleukin-6 and monocyte chemoattractant protein 1 (MCP-1) by SVCs from the OBA group was decreased by 1,25(OH)2D3 treatment. The 1,25(OH)2D3 treatment significantly reduced the toll-like receptor 4 and dual-specificity protein phosphatase 1 (also known as mitogen-activated protein kinase phosphatase 1) mRNA levels in SVCs and MCP-1 production by adipocytes from all 3 groups. CONCLUSIONS These findings suggest that vitamin D can attribute to the inhibition of the inflammatory response in VAT from atherosclerotic mice by reducing proinflammatory cytokine production.
Collapse
Affiliation(s)
- Deok Hoon Kwon
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| | - Jungwon Hwang
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| | - Hyeyoung You
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| | - Na Young Kim
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| | - Ga Young Lee
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| | - Sung Nim Han
- Department of Food and Nutrition, College of Human Ecology, Seoul National University, Seoul 08826, Korea
- Research Institute of Human Ecology, College of Human Ecology, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
6
|
Huang H, Sun Z, Xu J, Wang L, Zhao J, Li J, Zhang S, Yuan F, Liu M, Fang Z. Yang-Xin-Shu-Mai granule alleviates atherosclerosis by regulating macrophage polarization via the TLR9/MyD88/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2024; 318:116868. [PMID: 37454749 DOI: 10.1016/j.jep.2023.116868] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 06/04/2023] [Accepted: 06/27/2023] [Indexed: 07/18/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Previous studies have found that Yang-Xin-Shu-Mai granule (YXSMG) has certain advantages in the treatment of stable coronary heart disease. However, YXSMG can inhibit the progression of atherosclerotic plaque and stabilize vulnerable plaque needs to be further explored and studied. This research, mass spectrometry analysis, network pharmacology, in vivo and in vitro experimental studies were conducted to explore the mechanism of YXSMG on atherosclerosis. AIM OF THE STUDY To decipher the mechanism of atherosclerotic plaque, stabilization for YXSMG by analysis of its active ingredients and biological network and activity in whole animal and at cellular and molecular levels. METHODS The active components of YXSMG were determined using high performance liquid chromatography-mass spectrometry/mass spectrometry (HPLC-MS/MS) analysis. The 'Disease-Compound-Target-Pathway' network diagram was constructed using network pharmacology, and the stability of binding between core targets and core compounds was analyzed with molecular docking. After intervention with YXSMG, the pathology of aortic plaque, inflammation in the surrounding tissue, expression of TLR9/MyD88/NF-κB pathway protein in plaque and M1/M2 polarization of plaque macrophages were evaluated in vivo in apolipoprotein E-deficient (ApoE-/-) mice fed with high-fat diet. To verify whether it suppressed inflammation by inhibiting Toll-like receptor 9 (TLR9) reprogramming of macrophage polarization, we used RAW264.7 macrophages treated with specific TLR9 agonist (ODN1826) and inhibitor (ODN2088). RESULTS Five active compounds were identified in YXSMG: catechin, formononetin, tanshinone IIA, cryptotanshinone and glycitein. Network pharmacology studies revealed TLR9 as one of the core targets of YXSMG intervention in atherosclerosis. Computer simulation of molecular docking showed that TLR9 could interact with the core compound to form a stable complex. In vivo experiments confirmed that YXSMG could significantly inhibit atherosclerotic plaque, reduce levels of blood lipids and inflammatory factors, downregulate TLR9/MyD88/NF-κB pathway protein and inhibit aortic sinus macrophages polarization to M1, but promote their polarization to M2 to inhibit inflammation. In vitro experiments revealed that YXSMG could downregulate expression of TLR9 gene and protein in ODN1826-activated RAW264.7 macrophages. ODN2088 had a synergistic effect with YXSMG on the TLR9/MyD88/NF-κB signaling pathway, and reprogrammed macrophages polarization from M1 to M2 by inhibiting TLR9, thus reducing immuno-inflammatory response. CONCLUSION YXSMG can reduce the level of blood lipid and improve the size of atherosclerotic plaque and inflammatory infiltration in ApoE-/- mice fed with high fat. It is concluded that YXSMG can improve the mechanism of atherosclerotic plaque by inhibiting TLR9/MyD88/NF-κB pathway reprogramming macrophage M1/M2 polarization and reducing arterial inflammation.
Collapse
Affiliation(s)
- Hong Huang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| | - Zeqi Sun
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| | - Junyao Xu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Linjie Wang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Jing Zhao
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Jie Li
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Siqi Zhang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China; Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Fang Yuan
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China
| | - Ming Liu
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China; Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| | - Zhuyuan Fang
- Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China; Institute of Hypertension, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210029, PR China.
| |
Collapse
|
7
|
Avagimyan A, Pogosova N, Kakturskiy L, Sheibani M, Urazova O, Trofimenko A, Navarsdyan G, Jndoyan Z, Abgaryan K, Fogacci F, Galli M, Agati L, Kobalava Z, Shafie D, Marzilli M, Gogiashvili L, Sarrafzadegan N. HIV-Related Atherosclerosis: State-of-the-Art-Review. Curr Probl Cardiol 2023; 48:101783. [PMID: 37172874 DOI: 10.1016/j.cpcardiol.2023.101783] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Accepted: 05/07/2023] [Indexed: 05/15/2023]
Abstract
The infection caused by the Human Immunodeficiency Virus (HIV) has spread rapidly across the globe, assuming the characteristics of an epidemic in some regions. Thanks to the introduction of antiretroviral therapy into routine clinical practice, there was a considerable breakthrough in the treatment of HIV, that is now HIV is potentially well-controlled even in low-income countries. To date, HIV infection has moved from the group of life-threatening conditions to the group of chronic and well controlled ones and the quality of life and life expectancy of HIV+ people, with an undetectable viral load is closer to that of an HIV- people. However, unsolved issues still persist. For example: people living with HIV are more prone to the age-related diseases, especially atherosclerosis. For this reason, a better understanding of the mechanisms of HIV-associated destabilization of vascular homeostasis seems to be an urgent duty, that may lead to the development of new protocols, bringing the possibilities of pathogenetic therapies to a new level. The purpose of the article was to evaluate the pathological aspects of HIV-induced atherosclerosis.
Collapse
Affiliation(s)
- Ashot Avagimyan
- Assistant Professor, Anatomical Pathology and Clinical Morphology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia.
| | - Nana Pogosova
- Professor, Deputy of General Director for Science and Preventive Cardiology, National Medical Research Centre of Cardiology after E. Chazov, Moscow, Russia
| | - Lev Kakturskiy
- Professor, Scientific Director, Research Institute of Human Morphology FSBI «Petrovskiy NRCS, Moscow, Russia
| | - Mohammad Sheibani
- Department of Pharmacology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Razi Drug Research Centre, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Olga Urazova
- Professor, Head of Pathophysiology Department, Siberian State Medical University, Tomsk, Russia
| | - Artem Trofimenko
- Associate Professor, Pathophysiology Department, Kuban State Medical University, Krasnodar, Russia
| | - Grizelda Navarsdyan
- Professor, Pathophysiology Department, Yerevan State Medical University after M. Heratsi, Yerevan, Armenia
| | - Zinaida Jndoyan
- Professor, Head of Internal Diseases Propedeutics Department, Yerevan State Medical University after M. Heratsi, Armenia
| | - Kristina Abgaryan
- Associate Professor, Medical Microbiology Department, Yerevan State Medical University after M.Heratsi, Armenia
| | - Federica Fogacci
- Research Fellow, Atherosclerosis and Metabolic Disorders Research Unit, University of Bologna, Bologna, Italy
| | - Mattia Galli
- Maria Cecilia Hospital, GVM Care & Research, Cotignola, Italy
| | - Luciano Agati
- Professor of Cardiology Department, Head of Cardiology Unit Azienda Policlinico Umberto II, Sapienza University, Rome, Italy
| | - Zhanna Kobalava
- Professor, Head of Internal Disease, Cardiology and Clinical Pharmacology Department, Peoples' Friendship University of Russia (RUDN), Moscow, Russia
| | - Davood Shafie
- Isfahan Cardiovascular Research Institute, Isfahan, Iran
| | - Mario Marzilli
- Professor, Head of Cardiovascular Medicine Division, University of Pisa, Pisa, Italy
| | - Liana Gogiashvili
- Professor, Head of Experimental and Clinical Pathology Department, Al. Natishvili Institute of Experimental Morphology, I. Javakhishvili Tbilisi State University, Tbilisi, Georgia
| | - Nizal Sarrafzadegan
- Professor, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
8
|
Xuan X, Zhang J, Fan J, Zhang S. Research progress of Traditional Chinese Medicine (TCM) in targeting inflammation and lipid metabolism disorder for arteriosclerosis intervention: A review. Medicine (Baltimore) 2023; 102:e33748. [PMID: 37144986 PMCID: PMC10158879 DOI: 10.1097/md.0000000000033748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 04/21/2023] [Indexed: 05/06/2023] Open
Abstract
Atherosclerosis (AS) is a chronic disease caused by inflammation and lipid deposition. Immune cells are extensively activated in the lesions, producing excessive pro-inflammatory cytokines, which accompany the entire pathological process of AS. In addition, the accumulation of lipid-mediated lipoproteins under the arterial intima is a crucial event in the development of AS, leading to vascular inflammation. Improving lipid metabolism disorders and inhibiting inflammatory reactions are the primary treatment methods currently used in medical practice to delay AS progression. With the development of traditional Chinese medicine (TCM), more mechanisms of action of the monomer of TCM, Chinese patent medicine, and compound prescription have been studied and explored. Research has shown that some Chinese medicines can participate in treating AS by targeting and improving lipid metabolism disorders and inhibiting inflammatory reactions. This review explores the research on Chinese herbal monomers, compound Chinese medicines, and formulae that improve lipid metabolism disorders and inhibit inflammatory reactions to provide new supplements for treating AS.
Collapse
Affiliation(s)
- Xiaoyu Xuan
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jingyi Zhang
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Jilin Fan
- First School of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shiliang Zhang
- Department of Cardiology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
9
|
Abstract
Systemic inflammation has been suggested to have a pivotal role in atherothrombosis, but the factors that trigger systemic inflammation have not been fully elucidated. Lipopolysaccharide (LPS) is a component of the membrane of Gram-negative bacteria present in the gut that can translocate into the systemic circulation, causing non-septic, low-grade endotoxaemia. Gut dysbiosis is a major determinant of low-grade endotoxaemia via dysfunction of the intestinal barrier scaffold, which is a prerequisite for LPS translocation into the systemic circulation. Experimental studies have demonstrated that LPS is present in atherosclerotic arteries but not in normal arteries. In atherosclerotic plaques, LPS promotes a pro-inflammatory status that can lead to plaque instability and thrombus formation. Low-grade endotoxaemia affects several cell types, including leukocytes, platelets and endothelial cells, leading to inflammation and clot formation. Low-grade endotoxaemia has been described in patients at risk of or with overt cardiovascular disease, in whom low-grade endotoxaemia was associated with atherosclerotic burden and its clinical sequelae. In this Review, we describe the mechanisms favouring the development of low-grade endotoxaemia, focusing on gut dysbiosis and changes in gut permeability; the plausible biological mechanisms linking low-grade endotoxaemia and atherothrombosis; the clinical studies suggesting that low-grade endotoxaemia is a risk factor for cardiovascular events; and the potential therapeutic tools to improve gut permeability and eventually eliminate low-grade endotoxaemia.
Collapse
|
10
|
Parnigoni A, Viola M, Karousou E, Rovera S, Giaroni C, Passi A, Vigetti D. ROLE OF HYALURONAN IN PATHOPHYSIOLOGY OF VASCULAR1 ENDOTHELIAL AND SMOOTH MUSCLE CELLS. Am J Physiol Cell Physiol 2022; 323:C505-C519. [PMID: 35759431 DOI: 10.1152/ajpcell.00061.2022] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
One of the main components of the extracellular matrix (ECM) of the blood vessel is hyaluronic acid or hyaluronan (HA). It is a ubiquitous polysaccharide belonging to the family of glycosaminoglycans, but, differently from other proteoglycan-associated glycosaminoglycans, it is synthesized on the plasma membrane by a family of three HA synthases (HAS). HA can be released as a free polymer in the extracellular space or remain associated with the membrane in the pericellular space via HAS or via binding proteins. In fact, several cell surface proteins can interact with HA working as HA receptors like CD44, RHAMM, and LYVE-1. In physiological conditions, HA is localized in the glycocalyx and in the adventitia and is responsible for the loose and hydrated vascular structure favoring flexibility and allowing the stretching of vessels in response to mechanical forces. During atherogenesis, ECM undergoes dramatic alterations which have a crucial role in lipoprotein retention and in triggering multiple signaling cascades that wake up cells from their quiescent status. HA becomes highly present in the media and neointima favoring smooth muscle cells dedifferentiation, migration, and proliferation that strongly contribute to vessel wall thickening. Further, HA is able to modulate immune cell recruitment both within the vessel wall and on the endothelial cell layer. This review is focused on the effects of HA on vascular cell behavior.
Collapse
Affiliation(s)
- Arianna Parnigoni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Manuela Viola
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Evgenia Karousou
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Simona Rovera
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Cristina Giaroni
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Alberto Passi
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| | - Davide Vigetti
- Department of Medicine and Surgery, University of Insubria, Varese, Italy
| |
Collapse
|
11
|
Kong P, Cui ZY, Huang XF, Zhang DD, Guo RJ, Han M. Inflammation and atherosclerosis: signaling pathways and therapeutic intervention. Signal Transduct Target Ther 2022; 7:131. [PMID: 35459215 PMCID: PMC9033871 DOI: 10.1038/s41392-022-00955-7] [Citation(s) in RCA: 292] [Impact Index Per Article: 146.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 02/08/2023] Open
Abstract
Atherosclerosis is a chronic inflammatory vascular disease driven by traditional and nontraditional risk factors. Genome-wide association combined with clonal lineage tracing and clinical trials have demonstrated that innate and adaptive immune responses can promote or quell atherosclerosis. Several signaling pathways, that are associated with the inflammatory response, have been implicated within atherosclerosis such as NLRP3 inflammasome, toll-like receptors, proprotein convertase subtilisin/kexin type 9, Notch and Wnt signaling pathways, which are of importance for atherosclerosis development and regression. Targeting inflammatory pathways, especially the NLRP3 inflammasome pathway and its regulated inflammatory cytokine interleukin-1β, could represent an attractive new route for the treatment of atherosclerotic diseases. Herein, we summarize the knowledge on cellular participants and key inflammatory signaling pathways in atherosclerosis, and discuss the preclinical studies targeting these key pathways for atherosclerosis, the clinical trials that are going to target some of these processes, and the effects of quelling inflammation and atherosclerosis in the clinic.
Collapse
Affiliation(s)
- Peng Kong
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Xiao-Fu Huang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Dan-Dan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Rui-Juan Guo
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
12
|
Anto L, Blesso CN. Interplay Between Diet, the Gut Microbiome, and Atherosclerosis: Role of Dysbiosis and Microbial Metabolites on Inflammation and Disordered Lipid Metabolism. J Nutr Biochem 2022; 105:108991. [DOI: 10.1016/j.jnutbio.2022.108991] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 12/21/2021] [Accepted: 02/22/2022] [Indexed: 12/16/2022]
|
13
|
Aghamiri SH, Komlakh K, Ghaffari M. The crosstalk among TLR2, TLR4 and pathogenic pathways; a treasure trove for treatment of diabetic neuropathy. Inflammopharmacology 2022; 30:51-60. [PMID: 35020096 DOI: 10.1007/s10787-021-00919-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/19/2021] [Indexed: 11/25/2022]
Abstract
Diabetes is correlated with organ failures as a consequence of microvascular diabetic complications, including neuropathy, nephropathy, and retinopathy. These difficulties come with serious clinical manifestations and high medical costs. Diabetic neuropathy (DN) is one of the most prevalent diabetes complications, affecting at least 50% of diabetic patients with long disease duration. DN has serious effects on patients' life since it interferes with their daily physical activities and causes psychological comorbidities. There are some potential risk factors for the development of neuropathic injuries. It has been shown that inflammatory mechanisms play a pivotal role in the progression of DN. Among inflammatory players, TLR2 and TLR4 have gained immense importance because of their ability in recognizing distinct molecular patterns of invading pathogens and also damage-associated molecular patterns (DAMPs) providing inflammatory context for the progression of a wide array of disorders. We, therefore, sought to explore the possible role of TLR2 and TLR4 in DN pathogenesis and if whether manipulating TLRs is likely to be successful in fighting off DN.
Collapse
Affiliation(s)
- Seyed Hossein Aghamiri
- Department of Neurology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Khalil Komlakh
- Department of Neurosurgery, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mehran Ghaffari
- Department of Neurology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Portincasa P, Bonfrate L, Khalil M, Angelis MD, Calabrese FM, D’Amato M, Wang DQH, Di Ciaula A. Intestinal Barrier and Permeability in Health, Obesity and NAFLD. Biomedicines 2021; 10:83. [PMID: 35052763 PMCID: PMC8773010 DOI: 10.3390/biomedicines10010083] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 12/20/2021] [Accepted: 12/28/2021] [Indexed: 02/07/2023] Open
Abstract
The largest surface of the human body exposed to the external environment is the gut. At this level, the intestinal barrier includes luminal microbes, the mucin layer, gastrointestinal motility and secretion, enterocytes, immune cells, gut vascular barrier, and liver barrier. A healthy intestinal barrier is characterized by the selective permeability of nutrients, metabolites, water, and bacterial products, and processes are governed by cellular, neural, immune, and hormonal factors. Disrupted gut permeability (leaky gut syndrome) can represent a predisposing or aggravating condition in obesity and the metabolically associated liver steatosis (nonalcoholic fatty liver disease, NAFLD). In what follows, we describe the morphological-functional features of the intestinal barrier, the role of major modifiers of the intestinal barrier, and discuss the recent evidence pointing to the key role of intestinal permeability in obesity/NAFLD.
Collapse
Affiliation(s)
- Piero Portincasa
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Leonilde Bonfrate
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| | - Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Via Amendola 165/a, 70126 Bari, Italy; (M.D.A.); (F.M.C.)
| | - Mauro D’Amato
- Gastrointestinal Genetics Lab, CIC bioGUNE-BRTA, 48160 Derio, Spain;
- Ikerbasque, Basque Foundation for Science, 48009 Bilbao, Spain
| | - David Q.-H. Wang
- Department of Medicine and Genetics, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Einstein-Mount Sinai Diabetes Research Center, Albert Einstein College of Medicine, New York, NY 10461, USA;
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Biomedical Sciences & Human Oncology, University of Bari Medical School, 70124 Bari, Italy; (L.B.); (M.K.); (A.D.C.)
| |
Collapse
|
15
|
Calcium Dobesilate Modulates PKCδ-NADPH Oxidase- MAPK-NF-κB Signaling Pathway to Reduce CD14, TLR4, and MMP9 Expression during Monocyte-to-Macrophage Differentiation: Potential Therapeutic Implications for Atherosclerosis. Antioxidants (Basel) 2021; 10:antiox10111798. [PMID: 34829669 PMCID: PMC8615002 DOI: 10.3390/antiox10111798] [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: 10/28/2021] [Accepted: 11/08/2021] [Indexed: 01/18/2023] Open
Abstract
Monocyte-to-macrophage differentiation results in the secretion of various inflammatory mediators and oxidative stress molecules necessary for atherosclerosis pathogenesis. Consequently, this differentiation represents a potential clinical target in atherosclerosis. Calcium dobesilate (CaD), an established vasoactive and angioprotective drug in experimental models of diabetic microvascular complications reduces oxidative stress and inhibits inflammation via diverse molecular targets; however, its effect on monocytes/macrophages is poorly understood. In this study, we investigated the anti-inflammatory mechanism of CaD during phorbol 12-myristate 13-acetate (PMA)-induced monocyte-to-macrophage differentiation in in vitro models of sepsis (LPS) and hyperglycemia, using THP-1 monocytic cell line. CaD significantly suppressed CD14, TLR4, and MMP9 expression and activity, lowering pro-inflammatory mediators, such as IL1β, TNFα, and MCP-1. The effects of CaD translated through to studies on primary human macrophages. CaD inhibited reactive oxygen species (ROS) generation, PKCδ, MAPK (ERK1/2 and p38) phosphorylation, NOX2/p47phox expression, and membrane translocation. We used hydrogen peroxide (H2O2) to mimic oxidative stress, demonstrating that CaD suppressed PKCδ activation via its ROS-scavenging properties. Taken together, we demonstrate for the first time that CaD suppresses CD14, TLR4, MMP9, and signature pro-inflammatory cytokines, in human macrophages, via the downregulation of PKCδ/NADPH oxidase/ROS/MAPK/NF-κB-dependent signaling pathways. Our data present novel mechanisms of how CaD alleviates metabolic and infectious inflammation.
Collapse
|
16
|
Goretzki A, Lin Y, Schülke S. Immune metabolism in allergies, does it matter?-A review of immune metabolic basics and adaptations associated with the activation of innate immune cells in allergy. Allergy 2021; 76:3314-3331. [PMID: 33811351 DOI: 10.1111/all.14843] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 03/11/2021] [Accepted: 03/28/2021] [Indexed: 12/11/2022]
Abstract
Type I allergies are pathological, type 2 inflammatory immune responses against otherwise harmless environmental allergens that arise from complex interactions between different types of immune cells. Activated immune cells undergo extensive changes in phenotype and function to fulfill their effector functions. Hereby, activation, differentiation, proliferation, migration, and mounting of effector responses require metabolic reprogramming. While the metabolic changes associated with activation of dendritic cells, macrophages, and T cells are extensively studied, data about the metabolic phenotypes of the other cell types critically involved in allergic responses (epithelial cells, eosinophils, basophils, mast cells, and ILC2s) are rather limited. This review briefly covers the basics of cellular energy metabolism and its connection to immune cell function. In addition, it summarizes the current state of knowledge in terms of dendritic cell and macrophage metabolism and subsequently focuses on the metabolic changes associated with activation of epithelial cells, eosinophils, basophils, mast cells, as well as ILC2s in allergy. Interestingly, the innate key cell types in allergic inflammation were reported to change their metabolic phenotype during activation, shifting to either glycolysis (epithelial cells, M1 macrophages, DCs, eosinophils, basophils, acutely activated mast cells), oxidative phosphorylation (M2 macrophages, longer term activated mast cells), or fatty acid oxidation (ILC2s). Therefore, immune metabolism is of relevance in allergic diseases and its connection to immune cell effector function needs to be considered to better understand induction and maintenance of allergic responses. Further progress in this field will likely improve both our understanding of disease pathology and enable new treatment targets/strategies.
Collapse
Affiliation(s)
| | - Yen‐Ju Lin
- Molecular Allergology Paul‐Ehrlich‐Institut Langen Germany
| | - Stefan Schülke
- Molecular Allergology Paul‐Ehrlich‐Institut Langen Germany
| |
Collapse
|
17
|
Talepoor AG, Rastegari B, Kalani M, Doroudchi M. Decrease in the inflammatory cytokines of LPS-stimulated PBMCs of patients with atherosclerosis by a TLR-4 antagonist in the co-culture with HUVECs. Int Immunopharmacol 2021; 101:108295. [PMID: 34735917 DOI: 10.1016/j.intimp.2021.108295] [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: 08/05/2021] [Revised: 09/22/2021] [Accepted: 10/19/2021] [Indexed: 11/29/2022]
Abstract
Toll-like receptors (TLRs) are among the players of inflammation during atherosclerosis. We assessed the effects of Eritoran, a TLR-4 antagonist, on lipopolysaccharide (LPS)-induced cytokines production by Peripheral Blood Mononuclear Cells (PBMCs) of patients with high-stenosis (HS) (n = 6) and healthy controls (HCs) (n = 6) co-cultured with Human Umbilical Vein Endothelial Cells (HUVECs). LPS stimulation significantly increased the levels of IL-6 (P = 0.007 and P = 0.005), TNF-α (P = 0.006 and P = 0.005), IL-2 (P = 0.007 and P = 0.002), IFN-γ (P = 0.006 and P = 0.003), IL-17A (P = 0.004 and P = 0.003), IL-17F (P = 0.005 and P = 0.003), IL-5 (P = 0.007 and P = 0.005), IL-13 (P = 0.006 and P = 0.005), IL-9 (P = 0.005 and P = 0.005) and IL-21 (P = 0.007 and P = 0.005) in HUVECs co-cultured with HC and HS PBMCs as compared with un-stimulated co-culture condition, respectively. Eritoran treatment (50 μg/mL and 100 μg/mL) significantly reduced the levels of LPS-induced IL-6 (P = 0.007 and P = 0.006; P = 0.007 and P = 0.007), TNF-α (P = 0.005 and P = 0.003; P = 0.007 and P = 0.005), IL-2 (P = 0.007 and P = 0.005; P = 0.005 and P = 0.004), IFN-γ (P = 0.007 and P = 0.005; P = 0.005 and P = 0.004), IL-17A (P = 0.005 and P = 0.002; P = 0.005 and P = 0.002), IL-17F (P = 0.006 and P = 0.006; P = 0.005 and P = 0.005), IL-5 (P = 0.007 and P = 0.006; P = 0.007 and P = 0.007), IL-9 (P = 0.005 and P = 0.005; P = 0.005 and P = 0.005) and IL-21 (P = 0.007 and P = 0.007; P = 0.005 and P = 0.005) in stimulated HUVECs co-cultured with HC and HS PBMCs, compared to un-treated condition, respectively. Our results demonstrate that attenuating effect of Eritoran on the inflammatory responses to LPS is higher in PBMCs of patients with high stenosis, suggesting its potential role in ameliorating inflammatory conditions in atherosclerosis.
Collapse
Affiliation(s)
- Atefe Ghamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Banafsheh Rastegari
- Diagnostic Laboratory of Sciences and Technology Research Center, Paramedical School, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehdi Kalani
- Clinical Microbiology Research Center, Shiraz University of Medical Sciences, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
18
|
Usuda H, Okamoto T, Wada K. Leaky Gut: Effect of Dietary Fiber and Fats on Microbiome and Intestinal Barrier. Int J Mol Sci 2021; 22:ijms22147613. [PMID: 34299233 PMCID: PMC8305009 DOI: 10.3390/ijms22147613] [Citation(s) in RCA: 105] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 07/10/2021] [Accepted: 07/13/2021] [Indexed: 12/12/2022] Open
Abstract
Intestinal tract is the boundary that prevents harmful molecules from invading into the mucosal tissue, followed by systemic circulation. Intestinal permeability is an index for intestinal barrier integrity. Intestinal permeability has been shown to increase in various diseases-not only intestinal inflammatory diseases, but also systemic diseases, including diabetes, chronic kidney dysfunction, cancer, and cardiovascular diseases. Chronic increase of intestinal permeability is termed 'leaky gut' which is observed in the patients and animal models of these diseases. This state often correlates with the disease state. In addition, recent studies have revealed that gut microbiota affects intestinal and systemic heath conditions via their metabolite, especially short-chain fatty acids and lipopolysaccharides, which can trigger leaky gut. The etiology of leaky gut is still unknown; however, recent studies have uncovered exogenous factors that can modulate intestinal permeability. Nutrients are closely related to intestinal health and permeability that are actively investigated as a hot topic of scientific research. Here, we will review the effect of nutrients on intestinal permeability and microbiome for a better understanding of leaky gut and a possible mechanism of increase in intestinal permeability.
Collapse
Affiliation(s)
- Haruki Usuda
- Correspondence: (H.U.); (T.O.); Tel.: +81-853-20-3067 (H.U.)
| | | | | |
Collapse
|
19
|
Mohammad S, Thiemermann C. Role of Metabolic Endotoxemia in Systemic Inflammation and Potential Interventions. Front Immunol 2021; 11:594150. [PMID: 33505393 PMCID: PMC7829348 DOI: 10.3389/fimmu.2020.594150] [Citation(s) in RCA: 198] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/23/2020] [Indexed: 12/12/2022] Open
Abstract
Diet-induced metabolic endotoxemia is an important factor in the development of many chronic diseases in animals and man. The gut epithelium is an efficient barrier that prevents the absorption of liposaccharide (LPS). Structural changes to the intestinal epithelium in response to dietary alterations allow LPS to enter the bloodstream, resulting in an increase in the plasma levels of LPS (termed metabolic endotoxemia). LPS activates Toll-like receptor-4 (TLR4) leading to the production of numerous pro-inflammatory cytokines and, hence, low-grade systemic inflammation. Thus, metabolic endotoxemia can lead to several chronic inflammatory conditions. Obesity, diabetes, and non-alcoholic fatty liver disease (NAFLD) can also cause an increase in gut permeability and potential pharmacological and dietary interventions could be used to reduce the chronic low-grade inflammation associated with endotoxemia.
Collapse
Affiliation(s)
- Shireen Mohammad
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
20
|
Microbiota-Mediated Immune Regulation in Atherosclerosis. Molecules 2021; 26:molecules26010179. [PMID: 33401401 PMCID: PMC7795654 DOI: 10.3390/molecules26010179] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 12/29/2020] [Accepted: 12/29/2020] [Indexed: 12/19/2022] Open
Abstract
There is a high level of interest in identifying metabolites of endogenously produced or dietary compounds generated by the gastrointestinal (GI) tract microbiota, and determining the functions of these metabolites in health and disease. There is a wealth of compelling evidence that the microbiota is linked with many complex chronic inflammatory diseases, including atherosclerosis. Macrophages are key target immune cells in atherosclerosis. A hallmark of atherosclerosis is the accumulation of pro-inflammatory macrophages in coronary arteries that respond to pro-atherogenic stimuli and failure of digesting lipids that contribute to foam cell formation in atherosclerotic plaques. This review illustrates the role of tryptophan-derived microbiota metabolites as an aryl hydrocarbon receptor (AhR) ligand that has immunomodulatory properties. Also, microbiota-dependent trimethylamine-N-oxide (TMAO) metabolite production is associated with a deleterious effect that promotes atherosclerosis, and metabolite indoxyl sulfate has been shown to exacerbate atherosclerosis. Our objective in this review is to discuss the role of microbiota-derived metabolites in atherosclerosis, specifically the consequences of microbiota-induced effects of innate immunity in response to atherogenic stimuli, and how specific beneficial/detrimental metabolites impact the development of atherosclerosis by regulating chronic endotoxemic and lipotoxic inflammation.
Collapse
|
21
|
Farooqui AA. Contribution of gut microbiota and multiple organ failure in the pathogenesis of COVID-19 infection. GUT MICROBIOTA IN NEUROLOGIC AND VISCERAL DISEASES 2021. [PMCID: PMC7974322 DOI: 10.1016/b978-0-12-821039-0.00014-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
COVID-19, which originated in Wuhan, China, has spread rapidly all over the world. An increasing number of COVID-19 cases are caused by human-to-human transmission via respiratory droplets, coughs, and sneezes. The symptoms of COVID-19 patients are heterogeneous, ranging from mild upper respiratory symptoms (fever, languidness, unstable walking, dry cough, fatigue, and respiratory symptoms) to severe pneumonitis and even acute respiratory distress syndrome (ARDS) or death. COVID-19 invades human respiratory epithelial cells by binding with angiotensin-converting enzyme 2 (ACE2) receptors on human cell surfaces. Death in COVID-19 patients is caused by multiorgan function failure. In addition, systemic immune overactivation due to COVID-19 infection produces elevated expression of proinflammatory cytokines and chemokines, resulting in a so-called cytokine storm, a process that is an important factor in COVID-19 disease progression and multiple organ failure leading to death.
Collapse
|
22
|
Qiu X, Luo J, Fang L. AIBP, Angiogenesis, Hematopoiesis, and Atherogenesis. Curr Atheroscler Rep 2020; 23:1. [PMID: 33230630 DOI: 10.1007/s11883-020-00899-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2020] [Indexed: 01/04/2023]
Abstract
PURPOSE OF REVIEW The goal of this manuscript is to summarize the current understanding of the secreted APOA1 binding protein (AIBP), encoded by NAXE, in angiogenesis, hematopoiesis, and inflammation. The studies on AIBP illustrate a critical connection between lipid metabolism and the aforementioned endothelial and immune cell biology. RECENT FINDINGS AIBP dictates both developmental processes such as angiogenesis and hematopoiesis, and pathological events such as inflammation, tumorigenesis, and atherosclerosis. Although cholesterol efflux dictates AIBP-mediated lipid raft disruption in many of the cell types, recent studies document cholesterol efflux-independent mechanism involving Cdc42-mediated cytoskeleton remodeling in macrophages. AIBP disrupts lipid rafts and impairs raft-associated VEGFR2 but facilitates non-raft-associated NOTCH1 signaling. Furthermore, AIBP can induce cholesterol biosynthesis gene SREBP2 activation, which in turn transactivates NOTCH1 and supports specification of hematopoietic stem and progenitor cells (HSPCs). In addition, AIBP also binds TLR4 and represses TLR4-mediated inflammation. In this review, we summarize the latest research on AIBP, focusing on its role in cholesterol metabolism and the attendant effects on lipid raft-regulated VEGFR2 and non-raft-associated NOTCH1 activation in angiogenesis, SREBP2-upregulated NOTCH1 signaling in hematopoiesis, and TLR4 signaling in inflammation and atherogenesis. We will discuss its potential therapeutic applications in angiogenesis and inflammation due to selective targeting of activated cells.
Collapse
Affiliation(s)
- Xueting Qiu
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA
| | - Jingmin Luo
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA
| | - Longhou Fang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA. .,Department of Obstetrics and Gynecology, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA. .,Houston Methodist Institute for Academic Medicine, Houston Methodist Research Institute, 6550 Fannin Street, Houston, TX, 77030, USA. .,Department of Cardiothoracic Surgeries, Weill Cornell Medical College, Cornell University, New York, NY, 10065, USA.
| |
Collapse
|
23
|
Qaradakhi T, Gadanec LK, McSweeney KR, Abraham JR, Apostolopoulos V, Zulli A. The Anti-Inflammatory Effect of Taurine on Cardiovascular Disease. Nutrients 2020; 12:E2847. [PMID: 32957558 PMCID: PMC7551180 DOI: 10.3390/nu12092847] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/02/2020] [Accepted: 09/12/2020] [Indexed: 12/12/2022] Open
Abstract
Taurine is a non-protein amino acid that is expressed in the majority of animal tissues. With its unique sulfonic acid makeup, taurine influences cellular functions, including osmoregulation, antioxidation, ion movement modulation, and conjugation of bile acids. Taurine exerts anti-inflammatory effects that improve diabetes and has shown benefits to the cardiovascular system, possibly by inhibition of the renin angiotensin system. The beneficial effects of taurine are reviewed.
Collapse
Affiliation(s)
- Tawar Qaradakhi
- Institute for Health and Sport, Victoria University, Melbourne, VIC 8001, Australia; (L.K.G.); (K.R.M.); (J.R.A.); (V.A.); (A.Z.)
| | | | | | | | | | | |
Collapse
|
24
|
Wong SK, Chin KY, Ima-Nirwana S. Toll-like Receptor as a Molecular Link between Metabolic Syndrome and Inflammation: A Review. Curr Drug Targets 2020; 20:1264-1280. [PMID: 30961493 DOI: 10.2174/1389450120666190405172524] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 02/07/2023]
Abstract
Metabolic Syndrome (MetS) involves a cluster of five conditions, i.e. obesity, hyperglycaemia, hypertension, hypertriglyceridemia and low High-Density Lipoprotein (HDL) cholesterol. All components of MetS share an underlying chronic inflammatory aetiology, manifested by increased levels of pro-inflammatory cytokines. The pathogenic role of inflammation in the development of MetS suggested that toll-like receptor (TLR) activation may trigger MetS. This review summarises the supporting evidence on the interactions between MetS and TLR activation, bridged by the elevation of TLR ligands during MetS. The regulatory circuits mediated by TLR activation, which modulates signal propagation, leading to the state of chronic inflammation, are also discussed. Taken together, TLR activation could be the molecular basis in the development of MetS-induced inflammation.
Collapse
Affiliation(s)
- Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
25
|
Qu D, Wang L, Huo M, Song W, Lau CW, Xu J, Xu A, Yao X, Chiu JJ, Tian XY, Huang Y. Focal TLR4 activation mediates disturbed flow-induced endothelial inflammation. Cardiovasc Res 2020; 116:226-236. [PMID: 30785200 DOI: 10.1093/cvr/cvz046] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 07/19/2018] [Accepted: 02/18/2019] [Indexed: 11/13/2022] Open
Abstract
AIMS Disturbed blood flow at arterial branches and curvatures modulates endothelial function and predisposes the region to endothelial inflammation and subsequent development of atherosclerotic lesions. Activation of the endothelial Toll-like receptors (TLRs), in particular TLR4, contributes to vascular inflammation. Therefore, we investigate whether TLR4 can sense disturbed flow (DF) to mediate the subsequent endothelial inflammation. METHODS AND RESULTS En face staining of endothelium revealed that TLR4 expression, activation, and its downstream inflammatory markers were elevated in mouse aortic arch compared with thoracic aorta, which were absent in Tlr4mut mice. Similar results were observed in the partial carotid ligation model where TLR4 signalling was activated in response to ligation-induced flow disturbance in mouse carotid arteries, and such effect was attenuated in Tlr4mut mice. DF in vitro increased TLR4 expression and activation in human endothelial cells (ECs) and promoted monocyte-EC adhesion, which were inhibited in TLR4-knockdown ECs. Among endogenous TLR4 ligands examined as candidate mediators of DF-induced TLR4 activation, fibronectin containing the extra domain A (FN-EDA) expressed by ECs was increased by DF and was revealed to directly interact with and activate TLR4. CONCLUSION Our findings demonstrate the indispensable role of TLR4 in DF-induced endothelial inflammation and pinpoint FN-EDA as the endogenous TLR4 activator in this scenario. This novel mechanism of vascular inflammation under DF condition may serve as a critical initiating step in atherogenesis.
Collapse
Affiliation(s)
- Dan Qu
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Li Wang
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Mingyu Huo
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Wencong Song
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Chi-Wai Lau
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jian Xu
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong, China
| | - Xiaoqiang Yao
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Jeng-Jiann Chiu
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Xiao Yu Tian
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Shenzhen Research Institute, Institute of Vascular Medicine and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
26
|
Li B, Xia Y, Hu B. Infection and atherosclerosis: TLR-dependent pathways. Cell Mol Life Sci 2020; 77:2751-2769. [PMID: 32002588 PMCID: PMC7223178 DOI: 10.1007/s00018-020-03453-7] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 01/02/2020] [Accepted: 01/03/2020] [Indexed: 12/15/2022]
Abstract
Atherosclerotic vascular disease (ASVD) is a chronic process, with a progressive course over many years, but it can cause acute clinical events, including acute coronary syndromes (ACS), myocardial infarction (MI) and stroke. In addition to a series of typical risk factors for atherosclerosis, like hyperlipidemia, hypertension, smoking and obesity, emerging evidence suggests that atherosclerosis is a chronic inflammatory disease, suggesting that chronic infection plays an important role in the development of atherosclerosis. Toll-like receptors (TLRs) are the most characteristic members of pattern recognition receptors (PRRs), which play an important role in innate immune mechanism. TLRs play different roles in different stages of infection of atherosclerosis-related pathogens such as Chlamydia pneumoniae (C. pneumoniae), periodontal pathogens including Porphyromonas gingivalis (P. gingivalis), Helicobacter pylori (H. pylori) and human immunodeficiency virus (HIV). Overall, activation of TLR2 and 4 seems to have a profound impact on infection-related atherosclerosis. This article reviews the role of TLRs in the process of atherosclerosis after C. pneumoniae and other infections and the current status of treatment, with a view to providing a new direction and potential therapeutic targets for the study of ASVD.
Collapse
Affiliation(s)
- Bowei Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yuanpeng Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
27
|
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: 25] [Impact Index Per Article: 6.3] [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
|
28
|
Piscitelli F, Silvestri C. Role of the Endocannabinoidome in Human and Mouse Atherosclerosis. Curr Pharm Des 2020; 25:3147-3164. [PMID: 31448709 DOI: 10.2174/1381612825666190826162735] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Accepted: 08/21/2019] [Indexed: 12/20/2022]
Abstract
The Endocannabinoid (eCB) system and its role in many physiological and pathological conditions is well described and accepted, and includes cardiovascular disorders. However, the eCB system has been expanded to an "-ome"; the endocannabinoidome (eCBome) that includes endocannabinoid-related mediators, their protein targets and metabolic enzymes, many of which significantly impact upon cardiometabolic health. These recent discoveries are here summarized with a special focus on their potential involvement in atherosclerosis. We described the role of classical components of the eCB system (eCBs, CB1 and CB2 receptors) and eCB-related lipids, their regulatory enzymes and molecular targets in atherosclerosis. Furthermore, since increasing evidence points to significant cross-talk between the eCBome and the gut microbiome and the gut microbiome and atherosclerosis, we explore the possibility that a gut microbiome - eCBome axis has potential implications in atherosclerosis.
Collapse
Affiliation(s)
- Fabiana Piscitelli
- Institute of Biomolecular Chemistry, National Council of Research, Pozzuoli (NA), Italy
| | - Cristoforo Silvestri
- Institut universitaire de cardiologie et de pneumologie de Québec (IUCPQ), 2725 Chemin Sainte-Foy, Québec, QC, G1V 4G5, Canada.,Department of Medicine, Faculty of Medicine, Laval University, Quebec City, QC, Canada
| |
Collapse
|
29
|
Chi ZC. Relationship between non-alcoholic fatty liver disease and cardiovascular disease. Shijie Huaren Xiaohua Zazhi 2020; 28:313-329. [DOI: 10.11569/wcjd.v28.i9.313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
With the in-depth study of non-alcoholic fatty liver disease (NAFLD), it has been found in recent years that NAFLD is closely related to cardiovascular disease (CVD). It has been proved that NAFLD is not only an important risk factor for CVD, but it is also an important mechanism of atherosclerosis, coronary heart disease, and hypertension in young people. This article reviews the recent progress in the understanding of the relationship between NAFLD and CVD, with an aim to improve the knowledge of CVD physicians on liver disease and provide reference for prevention and treatment of these conditions.
Collapse
Affiliation(s)
- Zhao-Chun Chi
- Department of Gastroenterology, Qingdao Municipal Hospital, Qingdao 266011, Shandong Province, China
| |
Collapse
|
30
|
Afroz R, Zhou Y, Little PJ, Xu S, Mohamed R, Stow J, Kamato D. Toll-like Receptor 4 Stimulates Gene Expression via Smad2 Linker Region Phosphorylation in Vascular Smooth Muscle Cells. ACS Pharmacol Transl Sci 2020; 3:524-534. [PMID: 32566917 DOI: 10.1021/acsptsci.9b00113] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Indexed: 02/06/2023]
Abstract
Atherosclerosis begins in the vessel wall with the retention of low density lipoproteins to modified proteoglycans with hyperelongated glycosaminoglycan (GAG) chains. Bacterial infections produce endotoxins such as lipopolysaccharide that exacerbate the outcome of atherosclerosis by generating a heightened state of inflammation. Lipopolysaccharide (LPS) via its toll-like receptor (TLR) is well-known for its role in mediating an inflammatory response in the body. Emerging evidence demonstrates that TLRs are involved in regulating vascular functions. In this study we sought to investigate the role of LPS in proteoglycan modification and GAG chain elongation, and we hypothesize that LPS will signal via Smad2 dependent pathways to regulate GAG chain elongation. The in vitro model used human aortic vascular smooth muscle cells. GAG gene expression was assessed by quantitative real-time polymerase chain reaction. Western blotting was performed using whole-cell protein lysates to assess the signaling pathway. LPS via TLR4 stimulates the expression of GAG synthesizing enzymes to an equal extent to traditional cardiovascular agonists. LPS phosphorylates the Smad2 linker region via TAK-1/MAPK dependent pathways which correlated with genes associated with GAG chain initiation and elongation. The well-characterized role of LPS in inflammation and our data on GAG gene expression demonstrates that GAG chain elongation is the earliest marker of the inflammatory cascade in atherosclerosis development.
Collapse
Affiliation(s)
- Rizwana Afroz
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Ying Zhou
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| | - Suowen Xu
- Division of Life Science and Medicine, University of Science and Technology of China, Hefei, Anhui China
| | - Raafat Mohamed
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Basic Sciences, College of Dentistry, University of Mosul, Mosul, Iraq
| | - Jennifer Stow
- Institute of Molecular Bioscience, The University of Queensland, Woolloongabba, Queensland 4102, Australia
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Queensland 4102, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Tianhe District, Guangzhou 510520, China
| |
Collapse
|
31
|
Christ A, Lauterbach M, Latz E. Western Diet and the Immune System: An Inflammatory Connection. Immunity 2020; 51:794-811. [PMID: 31747581 DOI: 10.1016/j.immuni.2019.09.020] [Citation(s) in RCA: 417] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/24/2019] [Accepted: 09/24/2019] [Indexed: 02/06/2023]
Abstract
The consumption of Western-type calorically rich diets combined with chronic overnutrition and a sedentary lifestyle in Western societies evokes a state of chronic metabolic inflammation, termed metaflammation. Metaflammation contributes to the development of many prevalent non-communicable diseases (NCDs), and these lifestyle-associated pathologies represent a rising public health problem with global epidemic dimensions. A better understanding of how modern lifestyle and Western diet (WD) activate immune cells is essential for the development of efficient preventive and therapeutic strategies for common NCDs. Here, we review the current mechanistic understanding of how the Western lifestyle can induce metaflammation, and we discuss how this knowledge can be translated to protect the public from the health burden associated with their selected lifestyle.
Collapse
Affiliation(s)
- Anette Christ
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany; Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA 01605, USA
| | - Mario Lauterbach
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, University of Bonn, Bonn 53127, Germany; Department of Infectious Diseases & Immunology, UMass Medical School, Worcester, MA 01605, USA; Center of Molecular Inflammation Research, Norwegian University of Science and Technology, Trondheim 7491, Norway; German Center for Neurodegenerative Diseases (DZNE), Bonn 53127, Germany.
| |
Collapse
|
32
|
Liu Q, Li Y, Song X, Wang J, He Z, Zhu J, Chen H, Yuan J, Zhang X, Jiang H, Zhang S, Ruan B. Both gut microbiota and cytokines act to atherosclerosis in ApoE-/- mice. Microb Pathog 2019; 138:103827. [PMID: 31682994 DOI: 10.1016/j.micpath.2019.103827] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 10/29/2019] [Accepted: 10/29/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND Several studies have suggested a role for the gut microbiome and cytokines in atherosclerosis development, but combined analyses of the changes of the gut microbiota and cytokines have not been explored previously. METHODS We treated ApoE-/- and wild-type mice with a high-fat diet for 12 weeks. The gut microbiome and cytokine composition were analyzed using 16S ribosomal DNA sequencing and RayBio Quantibody Arrays, respectively. GO and KEGG analysis were performed to rationalize the potential mechanisms involved in the process of atherosclerosis. RESULTS Gut bacterial characteristics in ApoE-/- mice were clearly separated and 21 gut bacterial clades were detected by the LEfSe analysis showing significant differences during the development of atherosclerosis. The relative abundance of Verrucomicrobia, Bacteroidaceae, Bacteroides, and Akkermansia showed significant positive correlations with serum total cholesterol, triglyceride (TG), high-density lipoprotein (HDL) and low-density lipoprotein (LDL). Additionally, the relative abundance of Ruminococcaceae was positive with the level of HDL and the abundance of Rikenellaceae showed a negative relationship with the level of TG and LDL. Thirteen differentially expressed proteins were identified with P-value < 0.05. CXCL5, FGF2, and E-Selectin were significantly negatively associated with Akkermansia and Verrucomicrobia. Additionally, CXCL5 was significantly negatively correlated with Bacteroides and Bacteroidaceae. Three "cellular component" subcategories, 24 ″molecular function" subcategories, 752 ″biological process" subcategories and 29 statistically remarkable KEGG pathway categories were identified. CONCLUSIONS Gut microbiota changes of the mice having atherosclerosis and their relationship with the inflammatory status could be one of the major etiological mechanisms underlying atherosclerosis.
Collapse
Affiliation(s)
- Qiuxia Liu
- The First Affiliated Hospital, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, College of Medicine, Zhejiang University, Hangzhou, China
| | - Yuchuan Li
- The First Affiliated Hospital, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, College of Medicine, Zhejiang University, Hangzhou, China
| | - Xue Song
- The First Affiliated Hospital, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, College of Medicine, Zhejiang University, Hangzhou, China
| | - Jing Wang
- The First Affiliated Hospital, Department of Cardiology, College of Medicine, Zhejiang University, Hangzhou, China
| | - Zebao He
- Department of Infectious Diseases, Taizhou Enze Medical Center (Group) Enze Hospital, Taizhou, China
| | - Jiansheng Zhu
- Department of Infectious Diseases, Affiliated Taizhou Hospital of Wenzhou Medical University, Linhai, China
| | - Huazhong Chen
- Department of Infectious Diseases, Affiliated Taizhou Hospital of Wenzhou Medical University, Linhai, China
| | - Jing Yuan
- The Third People's Hospital of Shenzhen, Shenzhen, China
| | - Xue Zhang
- The First Affiliated Hospital, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, College of Medicine, Zhejiang University, Hangzhou, China
| | - Haiyin Jiang
- The First Affiliated Hospital, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, College of Medicine, Zhejiang University, Hangzhou, China
| | - Sheng Zhang
- Institute of Biotechnology, Cornell University, Ithaca, NY, United States
| | - Bing Ruan
- The First Affiliated Hospital, State Key Laboratory for Diagnosis and Treatment of Infectious Disease, College of Medicine, Zhejiang University, Hangzhou, China.
| |
Collapse
|
33
|
Liu T, Liu J, Lin Y, Que B, Chang C, Zhang J, Liang Z, Gao X, Liu S, Liu L, Li D, Ji Q. IL-37 inhibits the maturation of dendritic cells through the IL-1R8-TLR4-NF-κB pathway. Biochim Biophys Acta Mol Cell Biol Lipids 2019; 1864:1338-1349. [DOI: 10.1016/j.bbalip.2019.05.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/31/2022]
|
34
|
Pattern Recognition Receptor-Mediated Chronic Inflammation in the Development and Progression of Obesity-Related Metabolic Diseases. Mediators Inflamm 2019; 2019:5271295. [PMID: 31582899 PMCID: PMC6754942 DOI: 10.1155/2019/5271295] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity-induced chronic inflammation is known to promote the development of many metabolic diseases, especially insulin resistance, type 2 diabetes mellitus, nonalcoholic fatty liver disease, and atherosclerosis. Pattern recognition receptor-mediated inflammation is an important determinant for the initiation and progression of these metabolic diseases. Here, we review the major features of the current understanding with respect to obesity-related chronic inflammation in metabolic tissues, focus on Toll-like receptors and nucleotide-binding oligomerization domain-like receptors with an emphasis on how these receptors determine metabolic disease progression, and provide a summary on the development and progress of PRR antagonists for therapeutic intervention.
Collapse
|
35
|
The Role of RP105 in Cardiovascular Disease through Regulating TLR4 and PI3K Signaling Pathways. Curr Med Sci 2019; 39:185-189. [PMID: 31016532 DOI: 10.1007/s11596-019-2017-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2018] [Revised: 12/06/2018] [Indexed: 01/02/2023]
|
36
|
Arterial stiffness induced by carotid calcification leads to cerebral gliosis mediated by oxidative stress. J Hypertens 2019; 36:286-298. [PMID: 28938336 DOI: 10.1097/hjh.0000000000001557] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
BACKGROUND Arterial stiffness is a risk factor for cognitive decline and dementia. However, its precise effects on the brain remain unexplored. Using a mouse model of carotid stiffness, we investigated its effect on glial activation and oxidative stress. METHODS Arterial stiffness was induced by the application of calcium chloride to the adventitial region of the right carotid. Superoxide anion production, NADPH activity and levels, as well as glial activation were examined with immunohistochemical and biochemical approaches, 2-week postcalcification. Antioxidant treatment was done with Tempol (1 mmol/l) administered in the drinking water during 2 weeks. RESULTS The current study revealed that arterial stiffness increases the levels of the microglial markers ionized calcium-binding adapter molecule 1 and cluster of differentiation 68 in hippocampus, and of the astrocyte marker, s100 calcium binding protein β in hippocampus and frontal cortex. The cerebral inflammatory effects of arterial stiffness were specific to the brain and not due to systemic inflammation. Treatment with Tempol prevented the increase in superoxide anion in mice with carotid stiffness and attenuated the activation of microglia and astrocytes in the hippocampus. To determine whether the increased oxidative stress derives from NADPH oxidase, superoxide anion production was assessed by incubating brain tissue in the presence of gp91ds-tat, a selective NADPH oxidase 2 inhibitor. This peptide inhibited superoxide anion production to a greater extent in the brains of mice with carotid calcification compared with controls. CONCLUSION Carotid calcification leads to cerebral gliosis mediated by oxidative stress. Correcting arterial stiffness could offer a novel paradigm to protect the brain in populations where stiffness is prominent.
Collapse
|
37
|
Alí A, Boutjdir M, Aromolaran AS. Cardiolipotoxicity, Inflammation, and Arrhythmias: Role for Interleukin-6 Molecular Mechanisms. Front Physiol 2019; 9:1866. [PMID: 30666212 PMCID: PMC6330352 DOI: 10.3389/fphys.2018.01866] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/11/2018] [Indexed: 12/12/2022] Open
Abstract
Fatty acid infiltration of the myocardium, acquired in metabolic disorders (obesity, type-2 diabetes, insulin resistance, and hyperglycemia) is critically associated with the development of lipotoxic cardiomyopathy. According to a recent Presidential Advisory from the American Heart Association published in 2017, the current average dietary intake of saturated free-fatty acid (SFFA) in the US is 11–12%, which is significantly above the recommended <10%. Increased levels of circulating SFFAs (or lipotoxicity) may represent an unappreciated link that underlies increased vulnerability to cardiac dysfunction. Thus, an important objective is to identify novel targets that will inform pharmacological and genetic interventions for cardiomyopathies acquired through excessive consumption of diets rich in SFFAs. However, the molecular mechanisms involved are poorly understood. The increasing epidemic of metabolic disorders strongly implies an undeniable and critical need to further investigate SFFA mechanisms. A rapidly emerging and promising target for modulation by lipotoxicity is cytokine secretion and activation of pro-inflammatory signaling pathways. This objective can be advanced through fundamental mechanisms of cardiac electrical remodeling. In this review, we discuss cardiac ion channel modulation by SFFAs. We further highlight the contribution of downstream signaling pathways involving toll-like receptors and pathological increases in pro-inflammatory cytokines. Our expectation is that if we understand pathological remodeling of major cardiac ion channels from a perspective of lipotoxicity and inflammation, we may be able to develop safer and more effective therapies that will be beneficial to patients.
Collapse
Affiliation(s)
- Alessandra Alí
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Molecular and Developmental Medicine, University of Siena, Siena, Italy
| | - Mohamed Boutjdir
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Medicine, New York University School of Medicine, New York, NY, United States
| | - Ademuyiwa S Aromolaran
- Cardiovascular Research Program, VA New York Harbor Healthcare System, Brooklyn, NY, United States.,Department of Medicine, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Cell Biology, State University of New York Downstate Medical Center, Brooklyn, NY, United States.,Department of Pharmacology, State University of New York Downstate Medical Center, Brooklyn, NY, United States
| |
Collapse
|
38
|
Benomar Y, Taouis M. Molecular Mechanisms Underlying Obesity-Induced Hypothalamic Inflammation and Insulin Resistance: Pivotal Role of Resistin/TLR4 Pathways. Front Endocrinol (Lausanne) 2019; 10:140. [PMID: 30906281 PMCID: PMC6418006 DOI: 10.3389/fendo.2019.00140] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Accepted: 02/15/2019] [Indexed: 12/16/2022] Open
Abstract
Low-grade inflammation and insulin resistance are among the clinical features of obesity that are thought to promote the progressive onset of type 2 diabetes. However, the underlying mechanisms linking these disorders remain not fully understood. Recent reports pointed out hypothalamic inflammation as a major step in the onset of obesity-induced insulin resistance. In light of the increasing prevalence of obesity and T2D, two worldwide public health concerns, deciphering mechanisms implicated in hypothalamic inflammation constitutes a major challenge in the field of insulin-resistance/obesity. Several clinical and experimental studies have identified resistin as a key hormone linking insulin-resistance to obesity, notably through the activation of Toll Like Receptor (TLR) 4 signaling pathways. In this review, we present an overview of the molecular mechanisms underlying obesity-induced hypothalamic inflammation and insulin resistance with peculiar focus on the role of resistin/TLR4 signaling pathway.
Collapse
|
39
|
Moser VA, Uchoa MF, Pike CJ. TLR4 inhibitor TAK-242 attenuates the adverse neural effects of diet-induced obesity. J Neuroinflammation 2018; 15:306. [PMID: 30396359 PMCID: PMC6217784 DOI: 10.1186/s12974-018-1340-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 10/22/2018] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Obesity exerts negative effects on brain health, including decreased neurogenesis, impaired learning and memory, and increased risk for Alzheimer's disease and related dementias. Because obesity promotes glial activation, chronic neuroinflammation, and neural injury, microglia are implicated in the deleterious effects of obesity. One pathway that is particularly important in mediating the effects of obesity in peripheral tissues is toll-like receptor 4 (TLR4) signaling. The potential contribution of TLR4 pathways in mediating adverse neural outcomes of obesity has not been well addressed. To investigate this possibility, we examined how pharmacological inhibition of TLR4 affects the peripheral and neural outcomes of diet-induced obesity. METHODS Male C57BL6/J mice were maintained on either a control or high-fat diet for 12 weeks in the presence or absence of the specific TLR4 signaling inhibitor TAK-242. Outcomes examined included metabolic indices, a range of behavioral assessments, microglial activation, systemic and neuroinflammation, and neural health endpoints. RESULTS Peripherally, TAK-242 treatment was associated with partial inhibition of inflammation in the adipose tissue but exerted no significant effects on body weight, adiposity, and a range of metabolic measures. In the brain, obese mice treated with TAK-242 exhibited a significant reduction in microglial activation, improved levels of neurogenesis, and inhibition of Alzheimer-related amyloidogenic pathways. High-fat diet and TAK-242 were associated with only very modest effects on a range of behavioral measures. CONCLUSIONS These results demonstrate a significant protective effect of TLR4 inhibition on neural consequences of obesity, findings that further define the role of microglia in obesity-mediated outcomes and identify a strategy for improving brain health in obese individuals.
Collapse
Affiliation(s)
- V. Alexandra Moser
- 0000 0001 2156 6853grid.42505.36Neuroscience Graduate Program, University of Southern California, 3641 Watt Way, HNB 120, Los Angeles, CA 90089 USA
| | - Mariana F. Uchoa
- 0000 0001 2156 6853grid.42505.36Neuroscience Graduate Program, University of Southern California, 3641 Watt Way, HNB 120, Los Angeles, CA 90089 USA
| | - Christian J. Pike
- 0000 0001 2156 6853grid.42505.36Neuroscience Graduate Program, University of Southern California, 3641 Watt Way, HNB 120, Los Angeles, CA 90089 USA ,0000 0001 2156 6853grid.42505.36Leonard Davis School of Gerontology, University of Southern California, 3715 McClintock Avenue, Los Angeles, CA 90089-0191 USA
| |
Collapse
|
40
|
Nicoli F, Paul S, Appay V. Harnessing the Induction of CD8 + T-Cell Responses Through Metabolic Regulation by Pathogen-Recognition-Receptor Triggering in Antigen Presenting Cells. Front Immunol 2018; 9:2372. [PMID: 30410483 PMCID: PMC6209652 DOI: 10.3389/fimmu.2018.02372] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/24/2018] [Indexed: 01/03/2023] Open
Abstract
Cytotoxic CD8+ T-cells are key players of the immune responses against viruses. During the priming of a CD8+ T-cell response, the activation of a naïve T-cell by a professional antigen presenting cell (APC) involves the induction of various intracellular and metabolic pathways. The modulation of these pathways at the level of APCs or T-cells offers great potential to enhance the induction of robust effector cells and the generation of long-lived memory cells. On the one hand, signaling through pathogen recognition receptors (PRRs) expressed by APCs can greatly influence T-cell priming, and the potential of several PRR ligands as adjuvants are being studied. On the other hand, the engagement of several metabolic processes, at play in APCs and T-cells upon stimulation, implies that modulating cellular metabolism can impact on priming efficacy. Here, we review recent efforts to understand the interplay between PRR mediated signaling and metabolic pathway modulation in this context, through three examples: interplay between TLR4 and fatty acid metabolism, between TLR9 and IDO, and between STING and autophagy. These initial works highlight the potential for harnessing the induction of antiviral CD8+ T-cell responses using synergistic modulation of metabolic and PRR pathways.
Collapse
Affiliation(s)
- Francesco Nicoli
- Department of Molecular Medicine, University of Padua, Padua, Italy
| | - Stéphane Paul
- GIMAP/EA3064, Université de Lyon, CIC 1408 Vaccinology, Saint-Etienne, France
| | - Victor Appay
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses, Paris, France.,International Research Center of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
41
|
Dalby MJ, Aviello G, Ross AW, Walker AW, Barrett P, Morgan PJ. Diet induced obesity is independent of metabolic endotoxemia and TLR4 signalling, but markedly increases hypothalamic expression of the acute phase protein, SerpinA3N. Sci Rep 2018; 8:15648. [PMID: 30353127 PMCID: PMC6199263 DOI: 10.1038/s41598-018-33928-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Accepted: 10/08/2018] [Indexed: 01/17/2023] Open
Abstract
Hypothalamic inflammation is thought to contribute to obesity. One potential mechanism is via gut microbiota derived bacterial lipopolysaccharide (LPS) entering into the circulation and activation of Toll-like receptor-4. This is called metabolic endotoxemia. Another potential mechanism is systemic inflammation arising from sustained exposure to high-fat diet (HFD) over more than 12 weeks. In this study we show that mice fed HFD over 8 weeks become obese and show elevated plasma LPS binding protein, yet body weight gain and adiposity is not attenuated in mice lacking Tlr4 or its co-receptor Cd14. In addition, caecal microbiota composition remained unchanged by diet. Exposure of mice to HFD over a more prolonged period (20 weeks) to drive systemic inflammation also caused obesity. RNAseq used to assess hypothalamic inflammation in these mice showed increased hypothalamic expression of Serpina3n and Socs3 in response to HFD, with few other genes altered. In situ hybridisation confirmed increased Serpina3n and Socs3 expression in the ARC and DMH at 20-weeks, but also at 8-weeks and increased SerpinA3N protein could be detected as early as 1 week on HFD. Overall these data show lack of hypothalamic inflammation in response to HFD and that metabolic endotoxemia does not link HFD to obesity.
Collapse
Affiliation(s)
- Matthew J Dalby
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Gabriella Aviello
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Alexander W Ross
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Alan W Walker
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Perry Barrett
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - Peter J Morgan
- Rowett Institute, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom.
| |
Collapse
|
42
|
Kanter JE, Goodspeed L, Wang S, Kramer F, Wietecha T, Gomes-Kjerulf D, Subramanian S, O'Brien KD, den Hartigh LJ. 10,12 Conjugated Linoleic Acid-Driven Weight Loss Is Protective against Atherosclerosis in Mice and Is Associated with Alternative Macrophage Enrichment in Perivascular Adipose Tissue. Nutrients 2018; 10:nu10101416. [PMID: 30282904 PMCID: PMC6213611 DOI: 10.3390/nu10101416] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 09/26/2018] [Accepted: 09/27/2018] [Indexed: 02/05/2023] Open
Abstract
The dietary fatty acid 10,12 conjugated linoleic acid (10,12 CLA) promotes weight loss by increasing fat oxidation, but its effects on atherosclerosis are less clear. We recently showed that weight loss induced by 10,12 CLA in an atherosclerosis-susceptible mouse model with characteristics similar to human metabolic syndrome is accompanied by accumulation of alternatively activated macrophages within subcutaneous adipose tissue. The objective of this study was to evaluate whether 10,12 CLA-mediated weight loss was associated with an atheroprotective phenotype. Male low-density lipoprotein receptor deficient (Ldlr−/−) mice were made obese with 12 weeks of a high-fat, high-sucrose diet feeding (HFHS: 36% fat, 36% sucrose, 0.15% added cholesterol), then either continued on the HFHS diet with or without caloric restriction (CR), or switched to a diet with 1% of the lard replaced by either 9,11 CLA or 10,12 CLA for 8 weeks. Atherosclerosis and lipid levels were quantified at sacrifice. Weight loss in mice following 10,12 CLA supplementation or CR as a weight-matched control group had improved cholesterol and triglyceride levels, yet only the 10,12 CLA-treated mice had improved en face and aortic sinus atherosclerosis. 10,12 CLA-supplemented mice had increased lesion macrophage content, with enrichment of surrounding perivascular adipose tissue (PVAT) alternative macrophages, which may contribute to the anti-atherosclerotic effect of 10,12 CLA.
Collapse
Affiliation(s)
- Jenny E Kanter
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
| | - Leela Goodspeed
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
| | - Shari Wang
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
| | - Farah Kramer
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
| | - Tomasz Wietecha
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
- Department of Medicine, Cardiology, University of Washington, Box 356422, 1959 Pacific Ave NE, Seattle, WA 98195, USA.
| | - Diego Gomes-Kjerulf
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
| | - Savitha Subramanian
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
| | - Kevin D O'Brien
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
- Department of Medicine, Cardiology, University of Washington, Box 356422, 1959 Pacific Ave NE, Seattle, WA 98195, USA.
| | - Laura J den Hartigh
- Department of Medicine, Division of Metabolism, Endocrinology and Nutrition, University of Washington Medicine Diabetes Institute, University of Washington, Box 358062, 750 Republican Street, Seattle, WA 98109, USA.
| |
Collapse
|
43
|
Ma J, Li H. The Role of Gut Microbiota in Atherosclerosis and Hypertension. Front Pharmacol 2018; 9:1082. [PMID: 30319417 PMCID: PMC6167910 DOI: 10.3389/fphar.2018.01082] [Citation(s) in RCA: 128] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/06/2018] [Indexed: 12/20/2022] Open
Abstract
In recent years, accumulating evidence has indicated the importance of gut microbiota in maintaining human health. Gut dysbiosis is associated with the pathogenesis of a number of metabolic diseases including obesity, type 2 diabetes mellitus (T2DM), non-alcoholic fatty liver disease (NAFLD), and cardiovascular diseases (CVDs). Indeed, CVD has become the leading cause of death worldwide, especially in developed countries. In this review, we mainly discuss the gut microbiota-involved mechanisms of CVD focusing on atherosclerosis and hypertension, two major risk factors for serious CVD. Then, we briefly discuss the prospects of gut microbiota-targeted therapeutic strategies for the treatment of CVD in the future.
Collapse
Affiliation(s)
| | - Houkai Li
- Functional Metabolomic and Gut Microbiome Laboratory, Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
44
|
Rahman K, Fisher EA. Insights From Pre-Clinical and Clinical Studies on the Role of Innate Inflammation in Atherosclerosis Regression. Front Cardiovasc Med 2018; 5:32. [PMID: 29868610 PMCID: PMC5958627 DOI: 10.3389/fcvm.2018.00032] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 03/20/2018] [Indexed: 12/11/2022] Open
Abstract
Atherosclerosis, the underlying cause of coronary artery (CAD) and other cardiovascular diseases, is initiated by macrophage-mediated immune responses to lipoprotein and cholesterol accumulation in artery walls, which result in the formation of plaques. Unlike at other sites of inflammation, the immune response becomes maladaptive and inflammation fails to resolve. The most common treatment for reducing the risk from atherosclerosis is low density lipoprotein cholesterol (LDL-C) lowering. Studies have shown, however, that while significant lowering of LDL-C reduces the risk of heart attacks to some degree, there is still residual risk for the majority of the population. We and others have observed “residual inflammatory risk” of atherosclerosis after plasma cholesterol lowering in pre-clinical studies, and that this phenomenon is clinically relevant has been dramatically reinforced by the recent Canakinumab Anti-inflammatory Thrombosis Outcomes Study (CANTOS) trial. This review will summarize the role of the innate immune system, specifically macrophages, in atherosclerosis progression and regression, as well as the pre-clinical and clinical models that have provided significant insights into molecular pathways involved in the resolution of plaque inflammation and plaque regression. Partnered with clinical studies that can be envisioned in the post-CANTOS period, including progress in developing targeted plaque therapies, we expect that pre-clinical studies advancing on the path summarized in this review, already revealing key mechanisms, will continue to be essential contributors to achieve the goals of dampening plaque inflammation and inducing its resolution in order to maximize the therapeutic benefits of conventional risk factor modifications, such as LDL-C lowering.
Collapse
Affiliation(s)
- Karishma Rahman
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY, United States
| | - Edward A Fisher
- Department of Medicine, Division of Cardiology, New York University School of Medicine, New York, NY, United States
| |
Collapse
|
45
|
Tian D, Sha Y, Lu JM, Du XJ. MiR-370 inhibits vascular inflammation and oxidative stress triggered by oxidized low-density lipoprotein through targeting TLR4. J Cell Biochem 2018; 119:6231-6237. [PMID: 29663491 DOI: 10.1002/jcb.26851] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 03/09/2018] [Indexed: 12/21/2022]
Abstract
Atherosclerosis, as a chronic cardiovascular disease, still remains a serious threat to human health. Inflammation and oxidative stress are commonly involved in various stages of atherosclerosis development. MicroRNAs are reported to play important roles in macrophages, which can respond to inflammation and oxidative stress. In our current study, we focused on the biological roles of miR-370 in atherosclerosis. According to the previously research, miR-370 was downregulated in AS mice models. Oxidized low-density lipoprotein (Ox-LDL) is regarded as a crucial regulator of atherosclerosis and we observed that miR-370 was decreased by ox-LDL dose-dependently and time-dependently in THP-1 cells. Then, it was found that miR-370 overexpression was able to inhibit inflammation molecules including IL-6 and IL-1β. Meanwhile, ROS levels, and malondialdehyde (MDA) were also restrained by miR-370 mimics in vitro. Toll-like receptor 4 (TLR4) has been implicated in many inflammation diseases. It can serve as a target of miR-370 and TLR4 expression was greatly increased in ox-LDL-incubated THP-1 cells in a time and dose dependent manner. The negative correlation was validated using a dual-luciferase reporter assay in our study. In conclusion, our present study revealed that miR-370 can reduce inflammatory reaction and inhibit the ROS production by targeting TLR4 in THP-1 cells.
Collapse
Affiliation(s)
- Dan Tian
- Department of Emergency, Renmin Hospital of Wuhan University, Wuchang, Wuhan, China
| | - Yin Sha
- Department of Neurology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an,, China
| | - Jing-Min Lu
- Department of Neurology, Huai'an Second People's Hospital, The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an,, China
| | - Xian-Jin Du
- Department of Emergency, Renmin Hospital of Wuhan University, Wuchang, Wuhan, China
| |
Collapse
|
46
|
|
47
|
Fischer JW. Role of hyaluronan in atherosclerosis: Current knowledge and open questions. Matrix Biol 2018; 78-79:324-336. [PMID: 29510229 DOI: 10.1016/j.matbio.2018.03.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 02/20/2018] [Accepted: 03/01/2018] [Indexed: 12/21/2022]
Abstract
Hyaluronan (HA), HA synthases (HAS) and HA receptors are expressed during the progression of atherosclerotic plaques. HA is thought to promote the activated phenotype of local vascular smooth muscle cells characterized by increased migration, proliferation and matrix synthesis. Furthermore, HA may modulate the immune response by increasing macrophage retention and by promoting the polarization of Th1 cells that enhance macrophage driven inflammation as well. The pro-atherosclerotic functions of HA are opposed by the presence of HA in the glycocalyx where it critically contributes to anti-thrombotic and anti-inflammatory function of the glycocalyx. Patients with atherosclerosis often are affected by comorbidities among them diabetes mellitus type 2 and inflammatory comorbidities. Diabetes mellitus type 2 likely has close interrelations to HA synthesis in atherosclerosis because the activity and transcription of HA synthases are sensitive to the intracellular glucose metabolism, which determines the substrate availability and the posttranslational modifications of HA synthases. The pro-inflammatory comorbidities aggravate the course of atherosclerosis and will affect the expression of the genes related to HA biosynthesis, -degradation, HA-matrix assembly or signaling. One example being the induction of HAS3 by interleukin-1β and other cytokines. Furthermore complications of atherosclerosis such as the healing after myocardial infarction also involve HA responses.
Collapse
Affiliation(s)
- Jens W Fischer
- Institut für Pharmakologie und Klinische Pharmakologie, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany; CARID, Cardiovascular Research Institute Düsseldorf, University Hospital, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany.
| |
Collapse
|
48
|
Cong L, Zhang Y, Huang H, Cao J, Fu X. DFMG reverses proliferation and migration of vascular smooth muscle cells induced by co-culture with injured vascular endothelial cells via suppression of the TLR4-mediated signaling pathway. Mol Med Rep 2018; 17:5692-5699. [PMID: 29484442 PMCID: PMC5866011 DOI: 10.3892/mmr.2018.8635] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2017] [Accepted: 12/13/2017] [Indexed: 12/19/2022] Open
Abstract
7-Difluoromethoxy-5,4′-dimethoxy-genistein (DFMG) is a novel chemical compound synthesized using genistein. Previous studies have indicated that DFMG can reverse the apoptosis of vascular endothelial cells (VECs) by regulating the mitochondrial apoptosis pathway. The present study aimed to investigate the activity and molecular mechanism underlying DFMG-mediated protection of vascular smooth muscle cell (VSMCs) using a non-contact co-culture model established by using Transwell insert. Secretion of interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α) were measured by ELISA. Proliferation and migration of VSMCs were assessed using a Cell Counting kit-8 and wound healing assays, respectively. Toll-like receptor 4 (TLR4) mRNA and protein levels were detected by reverse transcription-quantitative polymerase chain reaction and western blotting analyses, respectively. In the present study, lysophosphatidylcholine (LPC) significantly increased the secretion of IL-6 and TNF-α in VECs. VECs treated with LPC markedly increased proliferation and migration of VSMCs, which were inhibited by DFMG. Transfection of either TLR4 short hairpin RNA (shRNA) or TLR4 cDNA in VECs inhibited and increased proliferation and migration of VSMCs, respectively. Furthermore, transfection of VECs with TLR4 shRNA suppressed the proliferation and migration of VSMCs induced by co-culture with injured VECs, which was further enhanced by treatment with DFMG. By contrast, transfection of VECs with TLR4 cDNA enhanced proliferation and migration of VSMCs and this effect was inhibited by treatment with DFMG. Taken together, the results of the present study demonstrated that DFMG can reverse proliferation and migration of VSMCs induced by co-culture with injured VECs via suppression of the TLR4-mediated signaling pathway.
Collapse
Affiliation(s)
- Li Cong
- Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Yong Zhang
- Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - He Huang
- Department of Pathology, Maternal and Child Health Hospital of Hunan Province, Changsha, Hunan 410008, P.R. China
| | - Jianguo Cao
- Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| | - Xiaohua Fu
- Medical College, Hunan Normal University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
49
|
Abstract
Although diet has long been known to contribute to the pathogenesis of cardiovascular disease (CVD), research over the past decade has revealed an unexpected interplay between nutrient intake, gut microbial metabolism and the host to modify the risk of developing CVD. Microbial-associated molecular patterns are sensed by host pattern recognition receptors and have been suggested to drive CVD pathogenesis. In addition, the host microbiota produces various metabolites, such as trimethylamine-N-oxide, short-chain fatty acids and secondary bile acids, that affect CVD pathogenesis. These recent advances support the notion that targeting the interactions between the host and microorganisms may hold promise for the prevention or treatment of CVD. In this Review, we summarize our current knowledge of the gut microbial mechanisms that drive CVD, with special emphasis on therapeutic interventions, and we highlight the need to establish causal links between microbial pathways and CVD pathogenesis.
Collapse
Affiliation(s)
- J Mark Brown
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, NC-10, Cleveland, Ohio 44195, USA
| | - Stanley L Hazen
- Department of Cellular and Molecular Medicine, Cleveland Clinic Lerner Research Institute, 9500 Euclid Avenue, NC-10, Cleveland, Ohio 44195, USA
| |
Collapse
|
50
|
Braunersreuther V, Burger F, Lenglet S, Pelli G, Carbone F, Fraga-Silva R, Stergiopulos N, Monaco C, Mueller C, Pagano S, Dallegri F, Mach F, Vuilleumier N, Montecucco F. Anti-apoA-1 auto-antibodies increase mouse atherosclerotic plaque vulnerability, myocardial necrosis and mortality triggering TLR2 and TLR4. Thromb Haemost 2017; 114:410-22. [PMID: 25879306 DOI: 10.1160/th14-12-1039] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 02/19/2015] [Indexed: 12/12/2022]
Abstract
SummaryAuto-antibodies to apolipoprotein A-1 (anti-apoA-1 IgG) were shown to promote inflammation and atherogenesis, possibly through innate immune receptors signalling. Here, we aimed at investigating the role of Toll-like receptors (TLR) 2 and 4 on anti-apoA-1 IgG-induced athero-sclerotic plaque vulnerability, myocardial necrosis and mortality in mice. Adult male apolipoprotein E knockout (ApoE)-/- (n=72), TLR2-/-ApoE-/- (n=36) and TLR4-/-Apo-/- (n=28) mice were intravenously injected with 50 µg/mouse of endotoxin-free polyclonal anti-apoA-1 IgG or control isotype IgG (CTL IgG) every two weeks for 16 weeks. Atherosclerotic plaque size and vulnerability were assessed by histology. Myocardial ischaemia and necrosis, respectively, were determined by electrocardiographic (ECG) changes assessed by telemetry and serum troponin I (cTnI) measurements. Impact on survival was assessed by Kaplan-Meier analyses. In ApoE-/- mice, anti-apoA-1 IgG passive immunisation enhanced histological features of athero-sclerotic plaque vulnerability (increase in neutrophil and MMP-9 and reduction in collagen content), induced a substantial cTnI elevation (p=0.001), and increased mortality rate by 23 % (LogRank, p=0.04) when compared to CTL IgG. On a subgroup of ApoE-/- mice equipped with telemetry (n=4), a significant ST-segment depression was noted in anti-apoA-1 IgG-treated mice when compared to CTL IgG recipients (p< 0.001), and an acute ST-segment elevation myocardial infarction preceding mouse death was observed in one case. The deleterious effects of anti-apoA-1 IgG on atherosclerotic plaque vulnerability, myocardial necrosis and death were partially reversed in TLR2-/-ApoE-/- and TLR4-/-ApoE-/- backgrounds. In conclusion, anti-apoA-1 auto-antibodies seem to be active mediators of atherosclerotic plaque vulnerability, myocardial necrosis, and mortality in mice through TLR2- and TLR4-mediated pathways.
Collapse
|