1
|
Wu L, Zhu Y, Qin Y, Yuan H, Zhang L, Lu T, Chen Q, Hu A. Conditional Knockout of IL-1R1 in Endothelial Cells Attenuates Seizures and Neurodegeneration via Inhibiting Neuroinflammation Mediated by Nrf2/HO-1/NLRP3 Signaling in Status Epilepticus Model. Mol Neurobiol 2024; 61:4289-4303. [PMID: 38087170 PMCID: PMC11236925 DOI: 10.1007/s12035-023-03842-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 11/29/2023] [Indexed: 07/11/2024]
Abstract
Studies on the bench and at bedside have demonstrated that the process of epileptogenesis is involved in neuroinflammatory responses. As the receptor of proinflammatory cytokine IL-1β, IL-1β type 1 receptor (IL-1R1) is reported to express abundantly in the endothelial cells in epileptic brains, which is deemed to be implicated in the epileptogenic process. However, whether and how endothelial IL-1R1 modulates neuroinflammatory responses in the pathological process of epileptic seizures and/or status epilepticus (SE) remains obscure. Here, we indicated endothelial IL-1R1 is involved in neuroinflammation, facilitating epilepsy progress via Nrf2/HO-1/NLRP3. In vitro, we observed upregulation of inflammatory cytokines in co-culture model under IL-1β challenge, as well as in BV2 cells after stimulation with conditional medium (CM) from IL-1β-stimulated bEnd.3 cells. In vivo, mice with conditional knockout of endothelial IL-1R1 (IL-1R1-CKO) were generated by hybrid IL-1R1flox/flox mice with Tek-Cre mice. IL-1R1-CKO reduced seizure susceptibility in kainic acid (KA)-induced SE model. In addition, IL-1R1-CKO KA mice exhibited lessened hippocampal neuroinflammation, mitigated neuronal damage, and decreased abnormal neurogenesis. In cognitive behavioral tests, IL-1R1-CKO KA mice presented improvement in learning and memory. Furthermore, we also indicated blockage of endothelial IL-1R1 downregulated the expressions of Nrf2/HO-1/NLRP3 pathway-related proteins. Nrf2-siRNA reversed the downregulation of HO-1, NLRP3, caspase-1, and IL-1β. These results demonstrated CKO of endothelial IL-1R1 reduces seizure susceptibility and attenuates SE-related neurobehavioral damage by suppressing hippocampal neuroinflammation via Nrf2/HO-1/NLRP3.
Collapse
Affiliation(s)
- Lianlian Wu
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Yuhua Zhu
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
- Institute of Cellular and Molecular Biology, School of Life Science, Jiangsu Normal University, Xuzhou, 221004, Jiangsu, People's Republic of China
| | - Ying Qin
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Honghua Yuan
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Lingzhi Zhang
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Tianyuan Lu
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China
| | - Quangang Chen
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| | - Ankang Hu
- Experimental Animal Center, Xuzhou Medical University, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
2
|
Shi J, Wu X, Qi H, Xu X, Hong S. Application and discoveries of metabolomics and proteomics in the study of female infertility. Front Endocrinol (Lausanne) 2024; 14:1315099. [PMID: 38274228 PMCID: PMC10810415 DOI: 10.3389/fendo.2023.1315099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Female infertility is defined as the absence of clinical pregnancy after 12 months of regular unprotected sexual intercourse. Methods This study employed metabolomics and proteomics approaches to investigate the relationship between metabolites and proteins and female infertility. The study used metabolomics and proteomics data from the UK Biobank to identify metabolites and proteins linked to infertility. Results The results showed that GRAM domain-containing protein 1C and metabolites fibrinogen cleavage peptides ADpSGEGDFXAEGGGVR and 3-Hydroxybutyrate had a positive correlation with infertility, whereas proteins such as Interleukin-3 receptor subunit alpha, Thrombospondin type-1 domain-containing protein 1, Intestinal-type alkaline phosphatase, and platelet and endothelial cell adhesion molecule 1 exhibited a negative correlation. These findings provide new clues and targets for infertility diagnosis and treatment. However, further research is required to validate these results and gain a deeper understanding of the specific roles of these metabolites and proteins in infertility pathogenesis. Discussion In conclusion, metabolomics and proteomics techniques have significant application value in the study of infertility, allowing for a better understanding of the biological mechanisms underlying infertility and providing new insights and strategies for its diagnosis and treatment. These research findings provide a crucial biological mechanistic basis for early infertility screening, prevention, and treatment.
Collapse
Affiliation(s)
- Junhua Shi
- Nursing Department, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xingjie Wu
- Department of Obstetrics, Hangzhou Medical College Affiliated Lin’an People’s Hospital, The First People’s Hospital of Hangzhou Lin’an District, Hangzhou, Zhejiang, China
| | - Haiou Qi
- Nursing Department, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xin Xu
- Nursing Department, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Shihao Hong
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University; Key Laboratory of Reproductive Dysfunction Management of Zhejiang Province, Hangzhou, China
| |
Collapse
|
3
|
Piccin A, Magzoub I, Hervig T. The 'scintilla' starting vaso-occlusion in sickle cell disease. Br J Haematol 2023; 201:379-380. [PMID: 36647924 DOI: 10.1111/bjh.18648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Accepted: 01/02/2023] [Indexed: 01/18/2023]
Affiliation(s)
- Andrea Piccin
- Northern Ireland Blood Transfusion Service (NIBTS), Belfast, UK.,Department of Haematology (V), University of Medicine, Innsbruck, Austria.,Department of Industrial Engineering, University of Trento, Trento, Italy
| | | | - Tor Hervig
- Irish Blood Transfusion Service, Dublin, Ireland
| |
Collapse
|
4
|
Sundaresan S, John S, Paneerselvam G, Andiapppan R, Christopher G, Selvam GS. Gallic acid attenuates cadmium mediated cardiac hypertrophic remodelling through upregulation of Nrf2 and PECAM-1signalling in rats. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2021; 87:103701. [PMID: 34237468 DOI: 10.1016/j.etap.2021.103701] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 07/01/2021] [Accepted: 07/03/2021] [Indexed: 06/13/2023]
Abstract
Gallic acid (GA) is an abundant natural polyphenolic compound found in vegetable and fruits that reduces the cardiac disease risk factor. This study aims to evaluate GA's role on cadmium (Cd) induced cardiac remodelling in experimental rats. Male Wistar rats were exposed to Cd (15 ppm) in drinking water and administered with GA orally (15 mg/kg/d) for 60 days. The results showed that GA regulated the lipid profile and reduced the LDL to 57 % compared with Cd treated rats. GA inhibited cardiac marker enzymes activity of CK-NAC (to 72.7 %) and CK-MB (to 100.3 %). Moreover, GA attenuated lipid peroxidation and enhanced the cardiac glutathione S transferase (GST) activity (89.2 %), glutathione peroxidase (GPx) (87 %), superoxide dismutase (SOD) (88.4 %) and catalase (CAT) activity (86.5 %). Histopathological examination showed that GA impaired the ventricular hypertrophy and fibrotic proliferation induced by Cd in rats. The combination of GA + Cd, decreased the gene expression of ANP (1-fold), BNP (0.5-fold) and β- MHC (0.9-fold). Furthermore, GA significantly reduced the expression of profibrotic (TGF-β) and proinflammatory (MCP-1) gene in Cd intoxicated rats. GA upregulated the expression of Nrf2 (2-fold), HO-1 (3-fold), and PECAM-1 (0.6-fold), which augments the detoxifying enzyme activity and cellular immunity in Cd intoxicated rats. The increased protein expression of Nrf2, PECAM-1 and decreased AKT-1 levels confirmed the mechanical action of GA during the hypertrophic condition. Thus, our results suggest that GA could act as a potential therapeutic agent regulating Nrf2 and PECAM-1 signalling pathways, thereby ameliorating Cd-induced pathological cardiac remodelling.
Collapse
Affiliation(s)
- Sasikumar Sundaresan
- Department of Biochemistry, Molecular Cardiology Unit, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | - Samu John
- Rajiv Gandhi Center for Biotechnology, Thycaud, Poojapura, P.O, Thiruvananthapuram, Kerala, India
| | - Gomathi Paneerselvam
- Department of Biochemistry, Molecular Cardiology Unit, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India
| | | | | | - Govindan Sadasivam Selvam
- Department of Biochemistry, Molecular Cardiology Unit, School of Biological Sciences, Madurai Kamaraj University, Madurai, Tamil Nadu, India.
| |
Collapse
|
5
|
Lu D, Le Y, Ding J, Dou X, Mao W, Zhu J. CLIC1 Inhibition Protects Against Cellular Senescence and Endothelial Dysfunction Via the Nrf2/HO-1 Pathway. Cell Biochem Biophys 2021; 79:239-252. [PMID: 33432550 DOI: 10.1007/s12013-020-00959-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/26/2020] [Indexed: 02/02/2023]
Abstract
Chloride intracellular channel 1 (CLIC1) is a sensor of oxidative stress in endothelial cells (EC). However, the mechanism by which CLIC1 mediate the regulation of endothelial dysfunction has not been established. In this study, overexpressed CLIC1 impaired the ability of the vascular cells to resist oxidative damage and promoted cellular senescence. Besides, suppressed CLIC1 protected against cellular senescence and dysfunction in Human Umbilical Vein Endothelial Cells (HUVECs) through the Nrf2/HO-1 pathway. We also found that ROS-activated CLIC1-induced oxidative stress in HUVECs. Nrf2 nuclear translocation was inhibited by CLIC1 overexpression, but was enhanced by IAA94 (CLICs inhibitor) treatment or knockdown of CLIC1. The Nrf2/HO-1 pathway plays a critical role in the anti-oxidative effect of suppressing CLIC1. And inhibition of CLIC1 decreases oxidative stress injury by downregulating the levels of ROS, MDA, and the expression of EC effectors (ICAM1 and VCAM1) protein expression and promotes the activity of superoxide dismutase (SOD). The AMPK-mediated signaling pathway activates Nrf2 through Nrf2 phosphorylation and nuclear translocation, which is also regulated by CLIC1. Moreover, the activation of CLIC1 contributes to H2O2-induced mitochondrial dysfunction and activation of mitochondrial fission. Therefore, elucidation of the mechanisms by which CLIC1 is involved in these pivotal pathways may uncover its therapeutic potential in alleviating ECs oxidative stress and age-related cardiovascular disease development.
Collapse
Affiliation(s)
- Dezhao Lu
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Yifei Le
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Jiali Ding
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Xiaobing Dou
- College of Life Science, Zhejiang Chinese Medical University, 310053, Hangzhou, China
| | - Wei Mao
- Cardiovascular department, The First Affiliated Hospital of Zhejiang Chinese Medicine University, 310006, Hangzhou, China.
| | - Ji Zhu
- Clinical Laboratory, The Third Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
6
|
Bonacina F, Da Dalt L, Catapano AL, Norata GD. Metabolic adaptations of cells at the vascular-immune interface during atherosclerosis. Mol Aspects Med 2020; 77:100918. [PMID: 33032828 PMCID: PMC7534736 DOI: 10.1016/j.mam.2020.100918] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/28/2020] [Accepted: 09/28/2020] [Indexed: 12/20/2022]
Abstract
Metabolic reprogramming is a physiological cellular adaptation to intracellular and extracellular stimuli that couples to cell polarization and function in multiple cellular subsets. Pathological conditions associated to nutrients overload, such as dyslipidaemia, may disturb cellular metabolic homeostasis and, in turn, affect cellular response and activation, thus contributing to disease progression. At the vascular/immune interface, the site of atherosclerotic plaque development, many of these changes occur. Here, an intimate interaction between endothelial cells (ECs), vascular smooth muscle cells (VSMCs) and immune cells, mainly monocytes/macrophages and lymphocytes, dictates physiological versus pathological response. Furthermore, atherogenic stimuli trigger metabolic adaptations both at systemic and cellular level that affect the EC layer barrier integrity, VSMC proliferation and migration, monocyte infiltration, macrophage polarization, lymphocyte T and B activation. Rewiring cellular metabolism by repurposing “metabolic drugs” might represent a pharmacological approach to modulate cell activation at the vascular immune interface thus contributing to control the immunometabolic response in the context of cardiovascular diseases.
Collapse
Affiliation(s)
- F Bonacina
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - L Da Dalt
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy.
| | - A L Catapano
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCSS Multimedica, Milan, Italy.
| | - G D Norata
- Department of Excellence of Pharmacological and Biomolecular Sciences, Università degli Studi di Milano, Milan, Italy; IRCCS, Ospedale Bassini, Cinisello Balsamo, Italy.
| |
Collapse
|
7
|
Oxidative Stress and Thrombosis during Aging: The Roles of Oxidative Stress in RBCs in Venous Thrombosis. Int J Mol Sci 2020; 21:ijms21124259. [PMID: 32549393 PMCID: PMC7352981 DOI: 10.3390/ijms21124259] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/10/2020] [Accepted: 06/11/2020] [Indexed: 01/17/2023] Open
Abstract
Mid-life stage adults are at higher risk of developing venous thrombosis (VT)/thromboembolism (VT/E). Aging is characterized by an overproduction of reactive oxygen species (ROS), which could evoke a series of physiological changes involved in thrombosis. Here, we focus on the critical role of ROS within the red blood cell (RBC) in initiating venous thrombosis during aging. Growing evidence has shifted our interest in the role of unjustifiably unvalued RBCs in blood coagulation. RBCs can be a major source of oxidative stress during aging, since RBC redox homeostasis is generally compromised due to the discrepancy between prooxidants and antioxidants. As a result, ROS accumulate within the RBC due to the constant endogenous hemoglobin (Hb) autoxidation and NADPH oxidase activation, and the uptake of extracellular ROS released by other cells in the circulation. The elevated RBC ROS level affects the RBC membrane structure and function, causing loss of membrane integrity, and decreased deformability. These changes impair RBC function in hemostasis and thrombosis, favoring a hypercoagulable state through enhanced RBC aggregation, RBC binding to endothelial cells affecting nitric oxide availability, RBC-induced platelet activation consequently modulating their activity, RBC interaction with and activation of coagulation factors, increased RBC phosphatidylserine exposure and release of microvesicles, accelerated aging and hemolysis. Thus, RBC oxidative stress during aging typifies an ultimate mechanism in system failure, which can affect major processes involved in the development of venous thrombosis in a variety of ways. The reevaluated concept of the critical role of RBC ROS in the activation of thrombotic events during aging will help identify potential targets for novel strategies to prevent/reduce the risk for VT/E or VT/E recurrences in mid-life stage adults.
Collapse
|
8
|
Kummer L, Zaradzki M, Vijayan V, Arif R, Weigand MA, Immenschuh S, Wagner AH, Larmann J. Vascular Signaling in Allogenic Solid Organ Transplantation - The Role of Endothelial Cells. Front Physiol 2020; 11:443. [PMID: 32457653 PMCID: PMC7227440 DOI: 10.3389/fphys.2020.00443] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 04/09/2020] [Indexed: 12/12/2022] Open
Abstract
Graft rejection remains the major obstacle after vascularized solid organ transplantation. Endothelial cells, which form the interface between the transplanted graft and the host’s immunity, are the first target for host immune cells. During acute cellular rejection endothelial cells are directly attacked by HLA I and II-recognizing NK cells, macrophages, and T cells, and activation of the complement system leads to endothelial cell lysis. The established forms of immunosuppressive therapy provide effective treatment options, but the treatment of chronic rejection of solid organs remains challenging. Chronic rejection is mainly based on production of donor-specific antibodies that induce endothelial cell activation—a condition which phenotypically resembles chronic inflammation. Activated endothelial cells produce chemokines, and expression of adhesion molecules increases. Due to this pro-inflammatory microenvironment, leukocytes are recruited and transmigrate from the bloodstream across the endothelial monolayer into the vessel wall. This mononuclear infiltrate is a hallmark of transplant vasculopathy. Furthermore, expression profiles of different cytokines serve as clinical markers for the patient’s outcome. Besides their effects on immune cells, activated endothelial cells support the migration and proliferation of vascular smooth muscle cells. In turn, muscle cell recruitment leads to neointima formation followed by reduction in organ perfusion and eventually results in tissue injury. Activation of endothelial cells involves antibody ligation to the surface of endothelial cells. Subsequently, intracellular signaling pathways are initiated. These signaling cascades may serve as targets to prevent or treat adverse effects in antibody-activated endothelial cells. Preventive or therapeutic strategies for chronic rejection can be investigated in sophisticated mouse models of transplant vasculopathy, mimicking interactions between immune cells and endothelium.
Collapse
Affiliation(s)
- Laura Kummer
- Department of Anesthesiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Marcin Zaradzki
- Institute of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Vijith Vijayan
- Institute for Transfusion Medicine, Hannover Medical School, Hanover, Germany
| | - Rawa Arif
- Institute of Cardiac Surgery, University Hospital Heidelberg, Heidelberg, Germany
| | - Markus A Weigand
- Department of Anesthesiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, Hanover, Germany
| | - Andreas H Wagner
- Institute of Physiology and Pathophysiology, Heidelberg University, Heidelberg, Germany
| | - Jan Larmann
- Department of Anesthesiology, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
9
|
Gutmann C, Siow R, Gwozdz AM, Saha P, Smith A. Reactive Oxygen Species in Venous Thrombosis. Int J Mol Sci 2020; 21:E1918. [PMID: 32168908 PMCID: PMC7139897 DOI: 10.3390/ijms21061918] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 01/03/2023] Open
Abstract
Reactive oxygen species (ROS) have physiological roles as second messengers, but can also exert detrimental modifications on DNA, proteins and lipids if resulting from enhanced generation or reduced antioxidant defense (oxidative stress). Venous thrombus (DVT) formation and resolution are influenced by ROS through modulation of the coagulation, fibrinolysis, proteolysis and the complement system, as well as the regulation of effector cells such as platelets, endothelial cells, erythrocytes, neutrophils, mast cells, monocytes and fibroblasts. Many conditions that carry an elevated risk of venous thrombosis, such as the Antiphospholipid Syndrome, have alterations in their redox homeostasis. Dietary and pharmacological antioxidants can modulate several important processes involved in DVT formation, but their overall effect is unknown and there are no recommendations regarding their use. The development of novel antioxidant treatments that aim to abrogate the formation of DVT or promote its resolution will depend on the identification of targets that enable ROS modulation confined to their site of interest in order to prevent off-target effects on physiological redox mechanisms. Subgroups of patients with increased systemic oxidative stress might benefit from unspecific antioxidant treatment, but more clinical studies are needed to bring clarity to this issue.
Collapse
Affiliation(s)
- Clemens Gutmann
- King’s British Heart Foundation Centre, King’s College London, 125 Coldharbour Lane, London SE5 9NU, UK;
| | - Richard Siow
- Vascular Biology & Inflammation Section, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, SE1 9NH, UK;
| | - Adam M. Gwozdz
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Prakash Saha
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| | - Alberto Smith
- Academic Department of Surgery, School of Cardiovascular Medicine & Sciences, British Heart Foundation of Research Excellence, King’s College London, London SE1 7EH, UK; (A.M.G.); (P.S.)
| |
Collapse
|
10
|
Caligiuri G. Mechanotransduction, immunoregulation, and metabolic functions of CD31 in cardiovascular pathophysiology. Cardiovasc Res 2019; 115:1425-1434. [DOI: 10.1093/cvr/cvz132] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 05/02/2019] [Accepted: 05/14/2019] [Indexed: 12/13/2022] Open
Abstract
Abstract
Biomechanical changes in the heart and vessels drive rapid and dynamic regulation of blood flow, a vital process for meeting the changing metabolic needs of the peripheral tissues at any given point in time. The fluid movement of the blood exerts haemodynamic stress upon the solid elements of the cardiovascular system: the heart, vessels, and cellular components of the blood. Cardiovascular diseases can lead to prolonged mechanical stress, such as cardiac remodelling during heart failure or vascular stiffening in atherosclerosis. This can lead to a significantly reduced or increasingly turbulent blood supply, inducing a shift in cellular metabolism that, amongst other effects, can trigger the release of reactive oxygen species and initiate a self-perpetuating cycle of inflammation and oxidative stress. CD31 is the most abundant constitutive co-signalling receptor glycoprotein on endothelial cells, which line the cardiovascular system and form the first-line of cellular contact with the blood. By associating with most endothelial receptors involved in mechanosensing, CD31 regulates the response to biomechanical stimuli. In addition, by relocating in the lipid rafts of endothelial cells as well as of cells stably interacting with the endothelium, including leucocytes and platelets, CD31–CD31 trans-homophilic engagement guides and restrains platelet and immune cell accumulation and activation and at sites of damage. In this way, CD31 is at the centre of mediating mechanical, metabolic, and immunological changes within the circulation and provides a single target that may have pleiotropic beneficial effects.
Collapse
Affiliation(s)
- Giuseppina Caligiuri
- Université de Paris, Cardiovascular Immunobiology, UMRS1148, INSERM, Paris, France
- Cardiology Department and Physiology Departments, AP-HP, University Hospital Xavier Bichat, 46 Rue Henri Huchard, Paris, France
| |
Collapse
|
11
|
Crisford H, Sapey E, Stockley RA. Proteinase 3; a potential target in chronic obstructive pulmonary disease and other chronic inflammatory diseases. Respir Res 2018; 19:180. [PMID: 30236095 PMCID: PMC6149181 DOI: 10.1186/s12931-018-0883-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Accepted: 09/06/2018] [Indexed: 12/15/2022] Open
Abstract
Chronic Obstructive Pulmonary Disease (COPD) is a common, multifactorial lung disease which results in significant impairment of patients' health and a large impact on society and health care burden. It is believed to be the result of prolonged, destructive neutrophilic inflammation which results in progressive damage to lung structures. During this process, large quantities of neutrophil serine proteinases (NSPs) are released which initiate the damage and contribute towards driving a persistent inflammatory state.Neutrophil elastase has long been considered the key NSP involved in the pathophysiology of COPD. However, in recent years, a significant role for Proteinase 3 (PR3) in disease development has emerged, both in COPD and other chronic inflammatory conditions. Therefore, there is a need to investigate the importance of PR3 in disease development and hence its potential as a therapeutic target. Research into PR3 has largely been confined to its role as an autoantigen, but PR3 is involved in triggering inflammatory pathways, disrupting cellular signalling, degrading key structural proteins, and pathogen response.This review summarises what is presently known about PR3, explores its involvement particularly in the development of COPD, and indicates areas requiring further investigation.
Collapse
Affiliation(s)
- Helena Crisford
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2GW, UK.
- Institute of Inflammation and Ageing, College of Medical and Dental Sciences, Centre for Translational Inflammation Research, University of Birmingham Research Laboratories, Queen Elizabeth Hospital Birmingham, Mindelsohn Way, Birmingham, B15 2WB, UK.
| | - Elizabeth Sapey
- Institute of Inflammation and Ageing, University of Birmingham, Edgbaston, Birmingham, B15 2GW, UK
| | - Robert A Stockley
- University Hospital Birmingham NHS Foundation Trust, Edgbaston, Birmingham, B15 2GW, UK
| |
Collapse
|
12
|
Liu H, Liu Y, Li Y, Liu Z, Li L, Ding S, Wang Y, Zhang T, Li L, Shao Z, Fu R. Proteinase 3 expression on the neutrophils of patients with paroxysmal nocturnal hemoglobinuria. Exp Ther Med 2017; 15:2525-2532. [PMID: 29467851 DOI: 10.3892/etm.2017.5662] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 07/20/2017] [Indexed: 12/26/2022] Open
Abstract
Proteinase 3 (PR3) is released from neutrophils and regulates platelet activity, which is associated with cluster of differentiation (CD)177 antigen (NB1), a glycosylphosphatidylinositol-linked protein. In the present study, the effect of PR3 on thrombosis in paroxysmal nocturnal hemoglobinuria (PNH) and PNH-aplastic anemia (AA) syndrome was explored. The expression of PR3 and NB1 on CD59- neutrophils was detected by flow cytometry, immunofluorescence (IF), reverse transcription-quantitative polymerase chain reaction analysis and western blotting. Serum levels of PR3, proteinase-activated receptor 1 (PAR1) and D-Dimer were measured using ELISAs. The expression of PR3 and NB1 on the plasma membrane of CD59- neutrophils in patients with PNH/PNH-AA was significantly lower compared with their expression on CD59+ neutrophils in patients and controls (P=0.001). However, no correlation between PR3 and NB1 expression was identified. IF staining further demonstrated partially positive PR3 expression on CD59- neutrophils. The serum level of PR3 in patients was identified to be significantly decreased compared with healthy controls (P<0.0001), and significantly negatively correlated with PAR1 (r=-0.456; P=0.043) and D-Dimer (r=-0.503; P=0.028) levels. The mRNA and protein levels of PR3 on PNH clones did not change significantly compared with the control group. In conclusion, PR3 expression on the plasma membrane of neutrophils and in the serum of patients with PNH/PNH-AA decreased, which may result in increased PAR1 expression and increased clotting. The present study provides the basis for further study on platelets in PNH.
Collapse
Affiliation(s)
- Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yi Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yi Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Liyan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Shaoxue Ding
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Yihao Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Tian Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
13
|
Immenschuh S, Vijayan V, Janciauskiene S, Gueler F. Heme as a Target for Therapeutic Interventions. Front Pharmacol 2017; 8:146. [PMID: 28420988 PMCID: PMC5378770 DOI: 10.3389/fphar.2017.00146] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 03/07/2017] [Indexed: 12/30/2022] Open
Abstract
Heme is a complex of iron and the tetrapyrrole protoporphyrin IX with essential functions in aerobic organisms. Heme is the prosthetic group of hemoproteins such as hemoglobin and myoglobin, which are crucial for reversible oxygen binding and transport. By contrast, high levels of free heme, which may occur in various pathophysiological conditions, are toxic via pro-oxidant, pro-inflammatory and cytotoxic effects. The toxicity of heme plays a major role for the pathogenesis of prototypical hemolytic disorders including sickle cell disease and malaria. Moreover, there is increasing appreciation that detrimental effects of heme may also be critically involved in diseases, which usually are not associated with hemolysis such as severe sepsis and atherosclerosis. In mammalians homeostasis of heme and its potential toxicity are primarily controlled by two physiological systems. First, the scavenger protein hemopexin (Hx) non-covalently binds extracellular free heme with high affinity and attenuates toxicity of heme in plasma. Second, heme oxygenases (HOs), in particular the inducible HO isozyme, HO-1, can provide antioxidant cytoprotection via enzymatic degradation of intracellular heme. This review summarizes current knowledge on the pathophysiological role of heme for various diseases as demonstrated in experimental animal models and in humans. The functional significance of Hx and HOs for the regulation of heme homeostasis is highlighted. Finally, the therapeutic potential of pharmacological strategies that apply Hx and HO-1 in various clinical settings is discussed.
Collapse
Affiliation(s)
- Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical SchoolHannover, Germany
| | - Vijith Vijayan
- Institute for Transfusion Medicine, Hannover Medical SchoolHannover, Germany
| | | | - Faikah Gueler
- Department of Nephrology, Hannover Medical SchoolHannover, Germany
| |
Collapse
|
14
|
Affiliation(s)
- Ralph Kettritz
- Experimental and Clinical Research Center; A joint cooperation between the Charité and the Max-Delbrück Center for Molecular Medicine (MDC) and Department of Nephrology and Intensive Care Medicine; Charité University Health Services; Berlin Germany
| |
Collapse
|
15
|
Zilian E, Saragih H, Vijayan V, Hiller O, Figueiredo C, Aljabri A, Blasczyk R, Theilmeier G, Becker JU, Larmann J, Immenschuh S. Heme Oxygenase-1 Inhibits HLA Class I Antibody-Dependent Endothelial Cell Activation. PLoS One 2015; 10:e0145306. [PMID: 26690352 PMCID: PMC4686182 DOI: 10.1371/journal.pone.0145306] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 12/01/2015] [Indexed: 12/27/2022] Open
Abstract
Antibody-mediated rejection (AMR) is a key limiting factor for long-term graft survival in solid organ transplantation. Human leukocyte antigen (HLA) class I (HLA I) antibodies (Abs) play a major role in the pathogenesis of AMR via their interactions with HLA molecules on vascular endothelial cells (ECs). The antioxidant enzyme heme oxygenase (HO)-1 has anti-inflammatory functions in the endothelium. As complement-independent effects of HLA I Abs can activate ECs, it was the goal of the current study to investigate the role of HO-1 on activation of human ECs by HLA I Abs. In cell cultures of various primary human macro- and microvascular ECs treatment with monoclonal pan- and allele-specific HLA I Abs up-regulated the expression of inducible proinflammatory adhesion molecules and chemokines (vascular cell adhesion molecule-1 [VCAM-1], intercellular cell adhesion molecule-1 [ICAM-1], interleukin-8 [IL-8] and monocyte chemotactic protein 1 [MCP-1]). Pharmacological induction of HO-1 with cobalt-protoporphyrin IX reduced, whereas inhibition of HO-1 with either zinc-protoporphyrin IX or siRNA-mediated knockdown increased HLA I Ab-dependent up-regulation of VCAM-1. Treatment with two carbon monoxide (CO)-releasing molecules, which liberate the gaseous HO product CO, blocked HLA I Ab-dependent EC activation. Finally, in an in vitro adhesion assay exposure of ECs to HLA I Abs led to increased monocyte binding, which was counteracted by up-regulation of HO-1. In conclusion, HLA I Ab-dependent EC activation is modulated by endothelial HO-1 and targeted induction of this enzyme may be a novel therapeutic approach for the treatment of AMR in solid organ transplantation.
Collapse
Affiliation(s)
- Eva Zilian
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Hendry Saragih
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
- Faculty of Biology, Gadjah Mada University, Yogyakarta, Indonesia
| | - Vijith Vijayan
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Oliver Hiller
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | | | - Abid Aljabri
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
| | - Gregor Theilmeier
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Jan Ulrich Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Jan Larmann
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, Hannover, Germany
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany
- * E-mail:
| |
Collapse
|
16
|
Bunse CE, Fortmeier V, Tischer S, Zilian E, Figueiredo C, Witte T, Blasczyk R, Immenschuh S, Eiz-Vesper B. Modulation of heme oxygenase-1 by metalloporphyrins increases anti-viral T cell responses. Clin Exp Immunol 2015; 179:265-76. [PMID: 25196646 DOI: 10.1111/cei.12451] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/31/2014] [Indexed: 12/14/2022] Open
Abstract
Heme oxygenase (HO)-1, the inducible isoform of HO, has immunomodulatory functions and is considered a target for therapeutic interventions. In the present study, we investigated whether modulation of HO-1 might have regulatory effects on in-vitro T cell activation. The study examined whether: (i) HO-1 induction by cobalt-protoporphyrin (CoPP) or inhibition by tin-mesoporphyrin (SnMP) can affect expansion and function of virus-specific T cells, (ii) HO-1 modulation might have a functional effect on other cell populations mediating effects on proliferating T cells [e.g. dendritic cells (DCs), regulatory T cells (T(regs)) and natural killer cells] and (iii) HO-1-modulated anti-viral T cells might be suitable for adoptive immunotherapy. Inhibition of HO-1 via SnMP in cytomegalovirus (CMV)pp65-peptide-pulsed peripheral blood mononuclear cells (PBMCs) led to increased anti-viral T cell activation and the generation of a higher proportion of effector memory T cells (CD45RA(-) CD62L(-)) with increased capability to secrete interferon (IFN)-γ and granzyme B. T(reg) depletion and SnMP exposure increased the number of anti-viral T cells 15-fold. To test the possibility that HO-1 modulation might be clinically applicable in conformity with good manufacturing practice (GMP), SnMP was tested in isolated anti-viral T cells using the cytokine secretion assay. Compared to control, SnMP treatment resulted in higher cell counts and purity without negative impact on quality and effector function [CD107a, IFN-γ and tumour necrosis factor (TNF)-α levels were stable]. These results suggest an important role of HO-1 in the modulation of adaptive immune responses. HO-1 inhibition resulted in markedly more effective generation of functionally active T cells suitable for adoptive T cell therapy.
Collapse
Affiliation(s)
- C E Bunse
- Institute for Transfusion Medicine, Hannover Medical School, Hannover, Germany; Integrated Research and Treatment Centre Transplantation - IFB-Tx, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Dorresteijn MJ, Paine A, Zilian E, Fenten MGE, Frenzel E, Janciauskiene S, Figueiredo C, Eiz-Vesper B, Blasczyk R, Dekker D, Pennings B, Scharstuhl A, Smits P, Larmann J, Theilmeier G, van der Hoeven JG, Wagener FADTG, Pickkers P, Immenschuh S. Cell-type-specific downregulation of heme oxygenase-1 by lipopolysaccharide via Bach1 in primary human mononuclear cells. Free Radic Biol Med 2015; 78:224-32. [PMID: 25463280 DOI: 10.1016/j.freeradbiomed.2014.10.579] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/24/2014] [Accepted: 10/29/2014] [Indexed: 11/18/2022]
Abstract
Heme oxygenase (HO)-1 is the inducible isoform of the heme-degrading enzyme HO, which is upregulated by multiple stress stimuli. HO-1 has major immunomodulatory and anti-inflammatory effects via its cell-type-specific functions in mononuclear cells. Contradictory findings have been reported on HO-1 regulation by the Toll-like receptor (TLR) 4 ligand lipopolysaccharide (LPS) in these cells. Therefore, we reinvestigated the effects of LPS on HO-1 gene expression in human and murine mononuclear cells in vitro and in vivo. Remarkably, LPS downregulated HO-1 in primary human peripheral blood mononuclear cells (PBMCs), CD14(+) monocytes, macrophages, dendritic cells, and granulocytes, but upregulated this enzyme in primary murine macrophages and human monocytic leukemia cell lines. Furthermore, experiments with human CD14(+) monocytes revealed that activation of other TLRs including TLR1, -2, -5, -6, -8, and -9 decreased HO-1 mRNA expression. LPS-dependent downregulation of HO-1 was specific, because expression of cyclooxygenase-2, NADP(H)-quinone oxidoreductase-1, and peroxiredoxin-1 was increased under the same experimental conditions. Notably, LPS upregulated expression of Bach1, a critical transcriptional repressor of HO-1. Moreover, knockdown of this nuclear factor enhanced basal and LPS-dependent HO-1 expression in mononuclear cells. Finally, downregulation of HO-1 in response to LPS was confirmed in PBMCs from human individuals subjected to experimental endotoxemia. In conclusion, LPS downregulates HO-1 expression in primary human mononuclear cells via a Bach1-mediated pathway. As LPS-dependent HO-1 regulation is cell-type- and species-specific, experimental findings in cell lines and animal models need careful interpretation.
Collapse
MESH Headings
- Animals
- Basic-Leucine Zipper Transcription Factors/genetics
- Basic-Leucine Zipper Transcription Factors/metabolism
- Blotting, Western
- Down-Regulation
- Endotoxemia/drug therapy
- Endotoxemia/enzymology
- Endotoxemia/pathology
- Fanconi Anemia Complementation Group Proteins/genetics
- Fanconi Anemia Complementation Group Proteins/metabolism
- Gene Expression Regulation, Enzymologic/drug effects
- Heme Oxygenase-1/genetics
- Heme Oxygenase-1/metabolism
- Humans
- Leukemia, Monocytic, Acute/drug therapy
- Leukemia, Monocytic, Acute/enzymology
- Leukemia, Monocytic, Acute/pathology
- Leukocytes, Mononuclear/cytology
- Leukocytes, Mononuclear/drug effects
- Leukocytes, Mononuclear/enzymology
- Lipopolysaccharides/pharmacology
- Macrophages/cytology
- Macrophages/drug effects
- Macrophages/enzymology
- Mice
- Monocytes/cytology
- Monocytes/drug effects
- Monocytes/enzymology
- RNA, Messenger/genetics
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
Collapse
Affiliation(s)
- Mirrin J Dorresteijn
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Department of Pharmacology and Toxicology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Ananta Paine
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Eva Zilian
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Maaike G E Fenten
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Department of Pharmacology and Toxicology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Eileen Frenzel
- Department of Internal Medicine-Respiratory Medicine, Hannover Medical School, 30635 Hannover, Germany
| | - Sabina Janciauskiene
- Department of Internal Medicine-Respiratory Medicine, Hannover Medical School, 30635 Hannover, Germany
| | - Constanca Figueiredo
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Britta Eiz-Vesper
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Rainer Blasczyk
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Douwe Dekker
- Department of Pharmacology and Toxicology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Bas Pennings
- Department of Pharmacology and Toxicology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, 6500 Nijmegen, The Netherlands
| | - Alwin Scharstuhl
- Department of Pharmacology and Toxicology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Paul Smits
- Department of Pharmacology and Toxicology, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Jan Larmann
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Gregor Theilmeier
- Department of Anesthesiology and Intensive Care Medicine, Hannover Medical School, 30625 Hannover, Germany
| | - Johannes G van der Hoeven
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Nijmegen Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Frank A D T G Wagener
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Department of Orthodontics and Craniofacial Biology, Radboud University Medical Center, 6500 Nijmegen, The Netherlands
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands; Nijmegen Center for Infectious Diseases, Radboud University Medical Center, 6500 HB Nijmegen, The Netherlands
| | - Stephan Immenschuh
- Institute for Transfusion Medicine, Hannover Medical School, 30625 Hannover, Germany.
| |
Collapse
|
18
|
Lucchesi D, Russo R, Gabriele M, Longo V, Del Prato S, Penno G, Pucci L. Grain and bean lysates improve function of endothelial progenitor cells from human peripheral blood: involvement of the endogenous antioxidant defenses. PLoS One 2014; 9:e109298. [PMID: 25329912 PMCID: PMC4201454 DOI: 10.1371/journal.pone.0109298] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/29/2014] [Indexed: 12/14/2022] Open
Abstract
Increased oxidative stress contributes to the functional impairment of endothelial progenitor cells (EPCs), the pivotal players in the servicing of the endothelial cell lining. Several evidences suggest that decreasing oxidative stress by natural compounds with antioxidant properties may improve EPCs bioactivity. Here, we investigated the effects of Lisosan G (LG), a Triticum Sativum grain powder, and Lady Joy (LJ), a bean lysate, on function of EPCs exposed to oxidative stress. Peripheral blood mononuclear cells were isolated and plated on fibronectin-coated culture dishes; adherent cells, identified as early EPCs, were pre-treated with different concentrations of LG and LJ and incubated with hydrogen peroxide (H2O2). Viability, senescence, adhesion, ROS production and antioxidant enzymes gene expression were evaluated. Lysate-mediated Nrf-2 (nuclear factor (erythroid-derived 2)-like 2)/ARE (antioxidant response element) activation, a modulator of oxidative stress, was assessed by immunocytochemistry. Lady Joy 0.35-0.7 mg/ml increases EPCs viability; pre-treatment with either LG 0.7 mg/ml and LJ 0.35-0.7 mg/ml protect EPCs viability against H2O2-induced injury. LG 0.7 and LJ 0.35-0.7 mg/ml improve EPCs adhesion; pre-treatment with either LG 0.35 and 0.7 mg/ml or LJ 0.35, 0.7 and 1.4 mg/ml preserve adhesiveness of EPCs exposed to H2O2. Senescence is attenuated in EPCs incubated with lysates 0.35 mg/ml. After exposure to H2O2, LG pre-treated cells show a lower senescence than untreated EPCs. Lysates significantly decrease H2O2-induced ROS generation. Both lysates increase glutathione peroxidase-1 and superoxide dismutase-2 (SOD-2) expression; upon H2O2 exposure, pre-treatment with LJ allows higher SOD-2 expression. Heme oxigenase-1 increases in EPCs pre-treated with LG even upon H2O2 exposure. Finally, incubation with LG 0.7 mg/ml results in Nrf-2 translocation into the nucleus both at baseline and after the oxidative challenge. Our data suggest a protective effect of lysates on EPCs exposed to oxidative stress through the involvement of antioxidant systems. Lisosan G seems to activate the Nrf-2/ARE pathways.
Collapse
Affiliation(s)
- Daniela Lucchesi
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases, University of Pisa, Pisa, Italy
| | - Rossella Russo
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| | - Morena Gabriele
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| | - Vincenzo Longo
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| | - Stefano Del Prato
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases, University of Pisa, Pisa, Italy
| | - Giuseppe Penno
- Department of Clinical and Experimental Medicine, Section of Metabolic Diseases, University of Pisa, Pisa, Italy
| | - Laura Pucci
- Institute of Agricultural Biology and Biotechnology, National Research Council, CNR, Pisa, Italy
| |
Collapse
|