1
|
Barone L, Cucchiara M, Palano MT, Bassani B, Gallazzi M, Rossi F, Raspanti M, Zecca PA, De Antoni G, Pagiatakis C, Papait R, Bernardini G, Bruno A, Gornati R. Dental pulp mesenchymal stem cell (DPSCs)-derived soluble factors, produced under hypoxic conditions, support angiogenesis via endothelial cell activation and generation of M2-like macrophages. J Biomed Sci 2024; 31:99. [PMID: 39491013 PMCID: PMC11533415 DOI: 10.1186/s12929-024-01087-6] [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: 04/12/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Cell therapy has emerged as a revolutionary tool to repair damaged tissues by restoration of an adequate vasculature. Dental Pulp stem cells (DPSC), due to their easy biological access, ex vivo properties, and ability to support angiogenesis have been largely explored in regenerative medicine. METHODS Here, we tested the capability of Dental Pulp Stem Cell-Conditioned medium (DPSC-CM), produced in normoxic (DPSC-CM Normox) or hypoxic (DPSC-CM Hypox) conditions, to support angiogenesis via their soluble factors. CMs were characterized by a secretome protein array, then used for in vivo and in vitro experiments. In in vivo experiments, DPSC-CMs were associated to an Ultimatrix sponge and injected in nude mice. After excision, Ultimatrix were assayed by immunohistochemistry, electron microscopy and flow cytometry, to evaluate the presence of endothelial, stromal, and immune cells. For in vitro procedures, DPSC-CMs were used on human umbilical-vein endothelial cells (HUVECs), to test their effects on cell adhesion, migration, tube formation, and on their capability to recruit human CD14+ monocytes. RESULTS We found that DPSC-CM Hypox exert stronger pro-angiogenic activities, compared with DPSC-CM Normox, by increasing the frequency of CD31+ endothelial cells, the number of vessels and hemoglobin content in the Ultimatrix sponges. We observed that Utimatrix sponges associated with DPSC-CM Hypox or DPSC-CM Normox shared similar capability to recruit CD45- stromal cells, CD45+ leukocytes, F4/80+ macrophages, CD80+ M1-macrophages and CD206+ M2-macropages. We also observed that DPSC-CM Hypox and DPSC-CM Normox have similar capabilities to support HUVEC adhesion, migration, induction of a pro-angiogenic gene signature and the generation of capillary-like structures, together with the ability to recruit human CD14+ monocytes. CONCLUSIONS Our results provide evidence that DPSCs-CM, produced under hypoxic conditions, can be proposed as a tool able to support angiogenesis via macrophage polarization, suggesting its use to overcome the issues and restrictions associated with the use of staminal cells.
Collapse
Affiliation(s)
- Ludovica Barone
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Martina Cucchiara
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Matteo Gallazzi
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Federica Rossi
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Mario Raspanti
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| | - Piero Antonio Zecca
- Department of Medicine and Technological Innovation, University of Insubria, 21100, Varese, Italy
| | - Gianluca De Antoni
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy
| | - Christina Pagiatakis
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Roberto Papait
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Giovanni Bernardini
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy
| | - Antonino Bruno
- Laboratory of Immunology and General Pathology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy.
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138, Milan, Italy.
| | - Rosalba Gornati
- Laboratory of Cell Biology, Department of Biotechnology and Life Sciences, University of Insubria, 21100, Varese, Italy.
| |
Collapse
|
2
|
Hamadi N, Al-Salam S, Beegam S, Zaaba NE, Elzaki O, Nemmar A. Impact of prolonged exposure to occasional and regular waterpipe smoke on cardiac injury, oxidative stress and mitochondrial dysfunction in male mice. Front Physiol 2024; 15:1286366. [PMID: 38370014 PMCID: PMC10869456 DOI: 10.3389/fphys.2024.1286366] [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: 08/31/2023] [Accepted: 01/17/2024] [Indexed: 02/20/2024] Open
Abstract
Regular waterpipe smoking (Reg-WPS) is well recognized for its deleterious effect on the heart. However, there is a paucity of experimental studies on the impact of occasional waterpipe smoking (Occ-WPS), also known as nondaily smoking, versus Reg-WPS on cardiac homeostasis, and the mechanisms underlying these effects. Hence, we aimed, in the present study, to investigate the effect of Occ-WPS (30 min/day, 1 day/week) versus Reg-WPS (30 min/day, 5 days/week) for 6 months on systolic blood pressure (SBP), cardiac injury, oxidative markers, chemokines, proinflammatory cytokines, DNA damage and mitochondrial function compared with air (control) exposed mice. Our results show that SBP was increased following exposure to either Occ-WPS or Reg-WPS compared with air-exposed mice. Moreover, we found that only Reg-WPS induced a significant elevation in the levels of troponin I, brain natriuretic peptide, lactate dehydrogenase, and creatine phosphokinase. However, the atrial natriuretic peptide (ANP) was significantly increased in both Occ-WPS and Reg-WPS groups. Compared with air-exposed mice, the levels of lipid peroxidation, reduced glutathione and monocyte chemoattractant protein-1 were only significantly augmented in the Reg-WPS. However, catalase, superoxide dismutase, and CXCL1 were significantly increased in both Occ-WPS and Reg-WPS. The concentrations of the adhesion molecules E-selectin, vascular cell adhesion molecule-1, and intercellular adhesion molecule-1 were solely elevated in the heart of mice exposed to Reg-WPS. Similarly, the concentrations of interleukin-1β and tumor necrosis factor α were only significantly augmented in the Reg-WPS. However, both Occ-WPS and Reg-WPS triggered significant augmentation in the levels of IL17 and DNA damage compared to the control groups. Furthermore, while Occ-WPS induced a slight but statistically insignificant elevation in the concentrations of mammalian targets of rapamycin and nuclear factor erythroid-derived 2-like 2 (Nrf2) expression, Reg-WPS exposure increased their levels substantially, in addition to p53 and mitochondrial complexes II & III, and IV activities compared with air-exposed mice. In conclusion, our findings show that while the long-term Occ-WPS exposure induced an elevation of SBP, ANP, antioxidant enzymes, IL17, CXCL1, and cardiac DNA damage, Reg-WPS exposure was consistently associated with the elevation of SBP and occurrence of cardiac damage, inflammation, oxidative stress, DNA damage and mitochondrial dysfunction.
Collapse
Affiliation(s)
- Naserddine Hamadi
- Department of Life and Environmental Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi, United Arab Emirates
| | - Suhail Al-Salam
- Department of Pathology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| | - Abderrahim Nemmar
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain, United Arab Emirates
| |
Collapse
|
3
|
Kim KH, Hwang Y, Kang SS. Regulatory Effect of Spray-Dried Lactiplantibacillus plantarum K79 on the Activation of Vasodilatory Factors and Inflammatory Responses. Food Sci Anim Resour 2024; 44:216-224. [PMID: 38229862 PMCID: PMC10789557 DOI: 10.5851/kosfa.2023.e78] [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: 11/14/2023] [Revised: 11/29/2023] [Accepted: 11/30/2023] [Indexed: 01/18/2024] Open
Abstract
The reduction of nitric oxide (NO) bioavailability in the endothelium induces endothelial dysfunction, contributing to the development of hypertension. Although Lactobacillus consumption decreases blood pressure, intracellular signaling pathways related to hypertension have not been well elucidated. Thus, this study examined the effect of spray-dried Lactiplantibacillus plantarum K79 (LpK79) on NO production, intracellular signaling pathways, and inflammatory responses related to vascular function and hypertension. NO production was assessed in human umbilical vein endothelial cells (HUVECs) treated with LpK79. Endothelial NO synthase (eNOS) and intracellular signaling molecules were determined using Western blot analysis. LpK79 dose-dependently increased NO production and activated eNOS via the phosphoinositide 3-kinase/Akt signaling pathway HUVECs. Moreover, LpK79 mitigated the activation of crucial factors pivotal for vascular contraction in smooth muscle cells, such as phospholipase Cγ, myosin phosphatase target subunit 1, and Rho-associated kinase 2. When HUVECs were treated with LpL79 in the presence of Escherichia coli lipopolysaccharide (LPS), LpK79 effectively suppressed mRNA and protein expression of pro-inflammatory mediators induced by E. coli LPS. These results suggest that LpK79 provided a beneficial effect on the regulation of vascular endothelial function.
Collapse
Affiliation(s)
- Ki Hwan Kim
- Department of Food Science and
Biotechnology, College of Life Science and Biotechnology, Dongguk
University, Goyang 10326, Korea
| | | | - Seok-Seong Kang
- Department of Food Science and
Biotechnology, College of Life Science and Biotechnology, Dongguk
University, Goyang 10326, Korea
| |
Collapse
|
4
|
Wiejak J, Murphy FA, Maffia P, Yarwood SJ. Vascular smooth muscle cells enhance immune/vascular interplay in a 3-cell model of vascular inflammation. Sci Rep 2023; 13:15889. [PMID: 37741880 PMCID: PMC10517978 DOI: 10.1038/s41598-023-43221-8] [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: 07/11/2023] [Accepted: 09/21/2023] [Indexed: 09/25/2023] Open
Abstract
Atherosclerosis is a serious cardiovascular disease that is characterised by the development of atheroma, which are lipid-laden plaques that build up within arterial walls due to chronic inflammatory processes. These lesions are fundamentally attributed to a complex cellular crosstalk between vascular smooth muscle cells (VSMCs), vascular endothelial cells (VECs) and central immune cells, such as macrophages (Mɸs), which promote vascular inflammation. The presence of VSMCs exerts both positive and negative effects during atheroma development, which can be attributed to their phenotypic plasticity. Understanding the interactions between these key cell types during the development of vascular inflammation and atheroma will enhance the scope for new therapeutic interventions. This study aims to determine the importance of VSMCs for shaping the extracellular cytokine/chemokine profile and transcriptional responses of VECs (human coronary artery endothelial cells; HCAECs) to activated lipopolysaccharide (LPS)-stimulated THP1 Mɸs, in a 3-cell model of human vascular inflammation. It is evident that within the presence of VSMCs, enhanced cytokine production was associated with up-regulation of genes associated with vascular inflammation t. Results demonstrate that the presence of VSMCs in co-culture experiments enhanced cytokine production (including CXCL1/GROα, IL-6, IL-8 and CCL2/MCP1) and inflammatory gene expression (including genes involved in JAK/STAT, Jun and NFκB signalling) in HCAECs co-cultured with LPS-stimulated THP1 Mɸs. Our results highlight the importance of VSMCs in immune/endothelial cell interplay and indicate that 3-cell, rather than 2-cell co-culture, may be more appropriate for the study of cellular crosstalk between immune and vascular compartments in response to inflammatory and atherogenic stimuli.
Collapse
Affiliation(s)
- Jolanta Wiejak
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK
| | - Fiona A Murphy
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, G4 0RE, UK
| | - Pasquale Maffia
- School of Infection & Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
- School of Cardiovascular & Metabolic Health, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, G12 8TA, UK
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131, Naples, Italy
| | - Stephen J Yarwood
- Institute of Biological Chemistry, Biophysics and Bioengineering, Heriot-Watt University, Edinburgh, EH14 4AS, UK.
| |
Collapse
|
5
|
Son H, Choi HS, Baek SE, Kim YH, Hur J, Han JH, Moon JH, Lee GS, Park SG, Woo CH, Eo SK, Yoon S, Kim BS, Lee D, Kim K. Shear stress induces monocyte/macrophage-mediated inflammation by upregulating cell-surface expression of heat shock proteins. Biomed Pharmacother 2023; 161:114566. [PMID: 36963359 DOI: 10.1016/j.biopha.2023.114566] [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: 01/25/2023] [Revised: 03/15/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023] Open
Abstract
The loss of endothelial cells is associated with the accumulation of monocytes/macrophages underneath the surface of the arteries, where cells are prone to mechanical stimulation, such as shear stress. However, the impact of mechanical stimuli on monocytic cells remains unclear. To assess whether mechanical stress affects monocytic cell function, we examined the expression of inflammatory molecules and surface proteins, whose levels changed following shear stress in human THP-1 cells. Shear stress increased the inflammatory chemokine CCL2, which enhanced the migration of monocytic cells and tumor necrosis factor (TNF)-α and interleukin (IL)- 1β at transcriptional and protein levels. We identified that the surface levels of heat shock protein 70 (HSP70), HSP90, and HSP105 increased using mass spectrometry-based proteomics, which was confirmed by western blot analysis, flow cytometry, and immunofluorescence. Treatment with HSP70/HSP105 and HSP90 inhibitors suppressed the expression and secretion of CCL2 and monocytic cell migration, suggesting an association between HSPs and inflammatory responses. We also demonstrated the coexistence and colocalization of increased HSP90 immunoreactivity and CD68 positive cells in atherosclerotic plaques of ApoE deficient mice fed a high-fat diet and human femoral artery endarterectomy specimens. These results suggest that monocytes/macrophages affected by shear stress polarize to a pro-inflammatory phenotype and increase surface protein levels involved in inflammatory responses. The regulation of the abovementioned HSPs upregulated on the monocytes/macrophages surface may serve as a novel therapeutic target for inflammation due to shear stress.
Collapse
Affiliation(s)
- Hyojae Son
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee-Seon Choi
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Seung Eun Baek
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Yun-Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Department of Bioinformatics, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jin Hur
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jung-Hwa Han
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Jeong Hee Moon
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Ga Seul Lee
- Core Research Facility & Analysis Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Sung Goo Park
- Disease Target Structure Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea.
| | - Chang-Hoon Woo
- Department of Pharmacology, Yeungnam University College of Medicine, Daegu 49415, Republic of Korea
| | - Seong-Kug Eo
- College of Veterinary Medicine and Bio-Safety Research Institute, Jeonbuk National University, Iksan, Republic of Korea
| | - Sik Yoon
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Dongjun Lee
- Department of Convergence Medicine, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Transplantation Research Center, Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan 50612, Republic of Korea.
| | - Koanhoi Kim
- Department of Pharmacology, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea.
| |
Collapse
|
6
|
Velásquez IM, Malarstig A, Baldassarre D, Borne Y, de Faire U, Engström G, Eriksson P, Giral P, Humphries SE, Kurl S, Leander K, Lind L, Lindén A, Orsini N, Pirro M, Silveira A, Smit AJ, Tremoli E, Veglia F, Strawbridge RJ, Gigante B. Causal analysis of plasma IL-8 on carotid intima media thickness, a measure of subclinical atherosclerosis. Curr Res Transl Med 2023; 71:103374. [PMID: 36493747 DOI: 10.1016/j.retram.2022.103374] [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: 03/25/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND We investigated the causality of IL-8 on carotid intima-media thickness (c-IMT), a measure of sub-clinical atherosclerosis. METHODS The IMPROVE is a multicenter European study (n = 3,711). The association of plasma IL-8 with c-IMT (mm) was estimated by quantile regression. Genotyping was performed using the Illumina CardioMetabo and Immuno chips. Replication was attempted in three independent studies and a meta-analysis was performed using a random model. RESULTS In IMPROVE, each unit increase in plasma IL-8 was associated with an increase in median c-IMT measures (all p<0·03) in multivariable analyses. Linear regression identified rs117518778 and rs8057084 as associated with IL-8 levels and with measures of c-IMT. The two SNPs were combined in an IL-8-increasing genetic risk that showed causality of IL-8 on c-IMT in IMPROVE and in the UK Biobank (n = 22,179). The effect of IL-8 on c-IMT measures was confirmed in PIVUS (n = 1,016) and MDCCC (n = 6,103). The association of rs8057084 with c-IMT was confirmed in PIVUS and UK Biobank with a pooled estimate effect (β) of -0·006 with 95%CI (-0·008- -0·003). CONCLUSION Our results indicate that genetic variants associated with plasma IL-8 also associate with c-IMT. However, we cannot infer causality of this association, as these variants lie outside of the IL8 locus.
Collapse
Affiliation(s)
- Ilais Moreno Velásquez
- Gorgas Memorial Institute for Health Studies, Panama City, Panama; Max Delbrück Center for Molecular Medicine in the Helmholtz-Association, Molecular Epidemiology Research Group, Berlin, Germany
| | - Anders Malarstig
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Emerging Science and Innovation, Pfizer Worldwide Research, Development and Medical, Stockholm, Sweden
| | - Damiano Baldassarre
- Department of Medical Biotechnology and Translational Medicine, Università di Milano, Milan, Italy; Centro Cardiologico Monzino, IRCCS, Milan, Italy
| | - Yan Borne
- Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| | - Ulf de Faire
- Cardiovascular and Nutritional Epidemiology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Gunnar Engström
- Department of Clinical Sciences, Malmö, Lund University, Malmö, Sweden
| | - Per Eriksson
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Karolinska University Hospital Solna, Stockholm, Sweden
| | - Philippe Giral
- Sorbonne Université, INSERM UMR1166, Cardiovascular prevention unit, AP-HP, Groupe Hôpitalier Pitié-Salpetriere, Paris, France
| | - Steve E Humphries
- Cardiovascular Genetics, Institute Cardiovascular Science, University College London, United Kingdom
| | - Sudhir Kurl
- Institute of Public Health and Clinical Nutrition, University of Eastern Finland, Kuopio Campus, Finland
| | - Karin Leander
- Cardiovascular and Nutritional Epidemiology Unit, Institute of Environmental Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Lars Lind
- Department of Medical Sciences, Clinical Epidemiology, Uppsala University, Uppsala, Sweden
| | - Anders Lindén
- Unit for Lung and Airway Research, Institute of Environmental Medicine, Stockholm, Sweden; Karolinska Severe COPD Center, Department of Respiratory Medicine and Allergy, Karolinska University Hospital, Stockholm, Sweden
| | - Nicola Orsini
- Department of Global Public Health, Karolinska Institutet, Stockholm, Sweden
| | - Matteo Pirro
- Internal Medicine, Angiology and Arteriosclerosis Diseases, Department of Medicine, University of Perugia, Perugia, Italy
| | - Angela Silveira
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Karolinska University Hospital Solna, Stockholm, Sweden
| | - Andries J Smit
- Department of Medicine, University Medical Center Groningen, Groningen & Isala Clinics Zwolle, Department of Medicine, the Netherlands
| | | | - Fabrizio Veglia
- Centro Cardiologico Monzino, IRCCS, Milan, Italy; Maria Cecilia Hospital, Cotignola, RA, Italy
| | - Rona J Strawbridge
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Institute of Health and Wellbeing, University of Glasgow, Glasgow, United Kingdom; Health Data Research, United Kingdom
| | - Bruna Gigante
- Division of Cardiovascular Medicine, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden; Division of Cardiovascular Medicine, Department of Clinical Sciences, Danderyd University Hospital, Stockholm, Sweden.
| |
Collapse
|
7
|
The Potential Importance of CXCL1 in the Physiological State and in Noncancer Diseases of the Cardiovascular System, Respiratory System and Skin. Int J Mol Sci 2022; 24:ijms24010205. [PMID: 36613652 PMCID: PMC9820720 DOI: 10.3390/ijms24010205] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/11/2022] [Accepted: 12/12/2022] [Indexed: 12/24/2022] Open
Abstract
In this paper, we present a literature review of the role of CXC motif chemokine ligand 1 (CXCL1) in physiology, and in selected major non-cancer diseases of the cardiovascular system, respiratory system and skin. CXCL1, a cytokine belonging to the CXC sub-family of chemokines with CXC motif chemokine receptor 2 (CXCR2) as its main receptor, causes the migration and infiltration of neutrophils to the sites of high expression. This implicates CXCL1 in many adverse conditions associated with inflammation and the accumulation of neutrophils. The aim of this study was to describe the significance of CXCL1 in selected diseases of the cardiovascular system (atherosclerosis, atrial fibrillation, chronic ischemic heart disease, hypertension, sepsis including sepsis-associated encephalopathy and sepsis-associated acute kidney injury), the respiratory system (asthma, chronic obstructive pulmonary disease (COPD), chronic rhinosinusitis, coronavirus disease 2019 (COVID-19), influenza, lung transplantation and ischemic-reperfusion injury and tuberculosis) and the skin (wound healing, psoriasis, sunburn and xeroderma pigmentosum). Additionally, the significance of CXCL1 is described in vascular physiology, such as the effects of CXCL1 on angiogenesis and arteriogenesis.
Collapse
|
8
|
Lee IS, Ko SJ, Lee YN, Lee G, Rahman MH, Kim B. The Effect of Laminaria japonica on Metabolic Syndrome: A Systematic Review of Its Efficacy and Mechanism of Action. Nutrients 2022; 14:3046. [PMID: 35893900 PMCID: PMC9370431 DOI: 10.3390/nu14153046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/21/2022] [Indexed: 12/10/2022] Open
Abstract
Metabolic syndrome (MetS) is a medical condition characterized by abdominal obesity, insulin resistance, high blood pressure, and hyperlipidemia. An increase in the incidence of MetS provokes an escalation in health care costs and a downturn in quality of life. However, there is currently no cure for MetS, and the absence of immediate treatment for MetS has prompted the development of novel therapies. In accordance with recent studies, the brown seaweed Laminaria japonica (LJP) has anti-inflammatory and antioxidant properties, and so forth. LJP contains bioactive compounds used as food globally, and it has been used as a medicine in East Asian countries. We conducted a systematic review to examine whether LJP could potentially be a useful therapeutic drug for MetS. The following databases were searched from initiation to September 2021: PubMed, Web of Science, EMBASE, and Cochrane Central Register of Controlled Trials Library. Clinical trials and in vivo studies evaluating the effects of LJP on MetS were included. LJP reduces the oxidative stress-related lipid mechanisms, inflammatory cytokines and macrophage-related chemokines, muscle cell proliferation, and migration. Bioactive-glucosidase inhibitors reduce diabetic complications, a therapeutic target in obesity and type 2 diabetes. In obesity, LJP increases AMP-activated protein kinase and decreases acetyl-CoA carboxylase. Based on our findings, we suggest that LJP could treat MetS, as it has pharmacological effects on MetS.
Collapse
Affiliation(s)
- In-Seon Lee
- Department of Meridians and Acupoints, College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea;
- Acupuncture & Meridian Science Research Center, Kyung Hee University, Seoul 02447, Korea
| | - Seok-Jae Ko
- Department of Gastroenterology, College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea;
| | - Yu Na Lee
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| | - Gahyun Lee
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| | - Md. Hasanur Rahman
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| | - Bonglee Kim
- College of Korean Medicine, Kyung Hee University, Seoul 05253, Korea; (Y.N.L.); (G.L.); (M.H.R.)
| |
Collapse
|
9
|
Predictive Value of Monocyte Chemoattractant Protein-1 in the Development of Diastolic Dysfunction in Patients with Psoriatic Arthritis. DISEASE MARKERS 2022; 2022:4433313. [PMID: 35692875 PMCID: PMC9187441 DOI: 10.1155/2022/4433313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/24/2022] [Indexed: 01/19/2023]
Abstract
We aimed to evaluate the diagnostic accuracy of the proinflammatory monocyte chemotactic protein-1 (MCP-1) in the diagnosis of asymptomatic diastolic dysfunction (DD) in patients with psoriatic arthritis (PsA). The disease activity in psoriatic arthritis (DAPSA) was determined using clinical and laboratory parameters, and echocardiography was performed to estimate DD. Serum MCP-1 concentrations were elevated in PsA patients with DD diagnosed with ultrasound (median (25th percentile, 75th percentile): 366.6 pg/mL (283, 407.1 pg/mL) vs. 277.5 pg/mL (223.5, 319.1 pg/mL) in controls;
). PsA patients with serum MCP-1 concentration higher than the cut-off value of 347.6 pg/mL had a 7.74-fold higher chance of developing DD than PsA patients with lower serum MCP-1 concentrations (controls), with a specificity of 86.36% and sensitivity of 55%, as verified using ultrasound. The group with MCP-1 concentrations above the cut-off value also showed a higher late peak diastolic mitral inflow velocity, A-wave value (
), E/E
ratio (
), and a lower E/A ratio (
), peak systolic left atrial reservoir strain, SA value (
), early peak diastolic displacement of the mitral septal annulus, E
wave value (
), than controls. Systolic blood pressure (
), LDL cholesterol concentration (
), glucose concentration (
), and DAPSA (
) increased in the PsA group with higher MCP-1 concentrations, although there were no differences in comorbidities and therapy between the groups compared. Thus, the serum MCP-1 concentration was a significant and independent prognostic indicator for asymptomatic DD in PsA patients (
,
). The DAPSA score in PsA patients might indicate the need for echocardiography and adjustment of anti-inflammatory treatment in terms of DD prevention.
Collapse
|
10
|
Moradi S, Fallahi J, Tanideh N, Dara M, Aliabadi BE, Nafar S, Asadi-Yousefabad SL, Tabei SMB, Razban V. Genetically modified bone marrow mesenchymal stem cells and dental pulp mesenchymal stem cells by HIF-1alpha overexpression, differs in survival and angiogenic effects after in animal model of hind limb ischemia. GENE REPORTS 2021. [DOI: 10.1016/j.genrep.2021.101187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
11
|
Biodata Mining of Differentially Expressed Genes between Acute Myocardial Infarction and Unstable Angina Based on Integrated Bioinformatics. BIOMED RESEARCH INTERNATIONAL 2021; 2021:5584681. [PMID: 34568491 PMCID: PMC8456013 DOI: 10.1155/2021/5584681] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 06/10/2021] [Accepted: 08/26/2021] [Indexed: 12/14/2022]
Abstract
Acute coronary syndrome (ACS) is a complex syndrome of clinical symptoms. In order to accurately diagnose the type of disease in ACS patients, this study is aimed at exploring the differentially expressed genes (DEGs) and biological pathways between acute myocardial infarction (AMI) and unstable angina (UA). The GSE29111 and GSE60993 datasets containing microarray data from AMI and UA patients were downloaded from the Gene Expression Omnibus (GEO) database. DEG analysis of these 2 datasets is performed using the “limma” package in R software. DEGs were also analyzed using protein-protein interaction (PPI), Molecular Complex Detection (MCODE) algorithm, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis. Correlation analysis and “cytoHubba” were used to analyze the hub genes. A total of 286 DEGs were obtained from GSE29111 and GSE60993, including 132 upregulated genes and 154 downregulated genes. Subsequent comprehensive analysis identified 20 key genes that may be related to the occurrence and development of AMI and UA and were involved in the inflammatory response, interaction of neuroactive ligand-receptor, calcium signaling pathway, inflammatory mediator regulation of TRP channels, viral protein interaction with cytokine and cytokine receptor, human cytomegalovirus infection, and cytokine-cytokine receptor interaction pathway. The integrated bioinformatical analysis could improve our understanding of DEGs between AMI and UA. The results of this study might provide a new perspective and reference for the early diagnosis and treatment of ACS.
Collapse
|
12
|
Sudhakar P, Verstockt B, Cremer J, Verstockt S, Sabino J, Ferrante M, Vermeire S. Understanding the Molecular Drivers of Disease Heterogeneity in Crohn's Disease Using Multi-omic Data Integration and Network Analysis. Inflamm Bowel Dis 2021; 27:870-886. [PMID: 33313682 PMCID: PMC8128416 DOI: 10.1093/ibd/izaa281] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Indexed: 12/12/2022]
Abstract
Crohn's disease (CD), a form of inflammatory bowel disease (IBD), is characterized by heterogeneity along multiple clinical axes, which in turn impacts disease progression and treatment modalities. Using advanced data integration approaches and systems biology tools, we studied the contribution of CD susceptibility variants and gene expression in distinct peripheral immune cell subsets (CD14+ monocytes and CD4+ T cells) to relevant clinical traits. Our analyses revealed that most clinical traits capturing CD heterogeneity could be associated with CD14+ and CD4+ gene expression rather than disease susceptibility variants. By disentangling the sources of variation, we identified molecular features that could potentially be driving the heterogeneity of various clinical traits of CD patients. Further downstream analyses identified contextual hub proteins such as genes encoding barrier functions, antimicrobial peptides, chemokines, and their receptors, which are either targeted by drugs used in CD or other inflammatory diseases or are relevant to the biological functions implicated in disease pathology. These hubs could be used as cell type-specific targets to treat specific subtypes of CD patients in a more individualized approach based on the underlying biology driving their disease subtypes. Our study highlights the importance of data integration and systems approaches to investigate complex and heterogeneous diseases such as IBD.
Collapse
Affiliation(s)
- Padhmanand Sudhakar
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID)
| | - Bram Verstockt
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID)
- University Hospitals Leuven, Department of Gastroenterology and Hepatology
| | - Jonathan Cremer
- Department of Microbiology and Immunology, Laboratory of Clinical Immunology, Katholieke Universiteit Leuven, Leuven, Belgium
| | - Sare Verstockt
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID)
| | - João Sabino
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID)
- University Hospitals Leuven, Department of Gastroenterology and Hepatology
| | - Marc Ferrante
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID)
- University Hospitals Leuven, Department of Gastroenterology and Hepatology
| | - Séverine Vermeire
- Department of Chronic Diseases, Metabolism and Ageing, Translational Research Center for Gastrointestinal Disorders (TARGID)
- University Hospitals Leuven, Department of Gastroenterology and Hepatology
| |
Collapse
|
13
|
Roshanravan N, Shabestari AN, Alamdari NM, Ostadrahimi A, Separham A, Parvizi R, Jafarabadi MA, Ghodrat M, Akbarzadeh M, Naemi M, Ghazi MKK, Hadi A, Ghaffari S. A novel inflammatory signaling pathway in patients with slow coronary flow: NF-κB/IL-1β/nitric oxide. Cytokine 2021; 143:155511. [PMID: 33839001 DOI: 10.1016/j.cyto.2021.155511] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 02/26/2021] [Accepted: 03/19/2021] [Indexed: 11/16/2022]
Abstract
PURPOSE The slow coronary flow (SCF) was identified as delayed opacification of epicardial coronary arteries in the absence of stenotic lesion. Metabolic syndrome (MetS), oxidative stress, and inflammation may be possible known insulting factors for the pathogenesis of SCF. This investigation aimed to assess the relationship between some inflammatory markers, oxidative stress parameters and MetS components with SCF phenomenon. METHODS A total of 35 patients with SCF and 35 subjects with normal coronary flow (NCF) were included in the study. We assessed some inflammatory markers (IL-1β, IL-18, TNF-α, and NF-κB mRNA expression in peripheral blood mononuclear cells (PBMCs)). Moreover, blood samples of the participants were tested for total antioxidant capacity (TAC), glutathione peroxidase (GPX) and nitric oxide (NO) levels using enzyme-linked immunosorbent assay (ELISA). Diagnosis of MetS was based on the National Cholesterol Education Program's Adult Treatment Panel III report (ATPIII) criteria, 2005. Diagnostic criteria for coronary flow rates of all subjects were documented by thrombolysis in myocardial infarction (TIMI) frame count method. RESULTS SCF patients had significantly higher prevalence of MetS (46%, p = 0.048).We found that the level of TAC was significantly higher in the NCF group (p = 0.006). Furthermore, the NO concentration was significantly lower in SCF groups (p = 0.001). A significant incremental difference was detected in IL-1β (fold change 2.82 ± 0.31, p < 0.05) and NF-κB (fold change 4.62 ± 0.32, p < 0.05) mRNA expression in the SCF group when compared with its level in the NCF group. Furthermore, according to logistic regression analysis, there were significant associations between IL-1β, NF-κB expression levels and the incidence of SCF (p < 0.05). CONCLUSION Based on the findings of this study, the pathogenesis of the SCF phenomenon may be closely associated with metabolic syndrome and inflammation. The NF-κB/IL-1β/nitric oxide & MetS signaling pathway might be considered as potential therapeutic targets in the management of SCF patients but further researches is required to guarantee these findings.
Collapse
Affiliation(s)
- Neda Roshanravan
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Namazi Shabestari
- Department of Geriatric Medicine, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Naimeh Mesri Alamdari
- Students Research Committee, School of Health, Iran University of Medical Science, Tehran, Iran
| | - Alireza Ostadrahimi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Separham
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Rezayat Parvizi
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mahshid Ghodrat
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Moloud Akbarzadeh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell And Regenerative Medicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Naemi
- Nutrition Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Amir Hadi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
14
|
Wasiak S, Dzobo KE, Rakai BD, Kaiser Y, Versloot M, Bahjat M, Stotz SC, Fu L, Sweeney M, Johansson JO, Wong NCW, Stroes ESG, Kroon J, Kulikowski E. BET protein inhibitor apabetalone (RVX-208) suppresses pro-inflammatory hyper-activation of monocytes from patients with cardiovascular disease and type 2 diabetes. Clin Epigenetics 2020; 12:166. [PMID: 33172487 PMCID: PMC7657365 DOI: 10.1186/s13148-020-00943-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
Background Patients with cardiovascular disease (CVD) and type 2 diabetes (DM2) have a high residual risk for experiencing a major adverse cardiac event. Dysregulation of epigenetic mechanisms of gene transcription in innate immune cells contributes to CVD development but is currently not targeted by therapies. Apabetalone (RVX-208) is a small molecule inhibitor of bromodomain and extra-terminal (BET) proteins—histone acetylation readers that drive pro-inflammatory and pro-atherosclerotic gene transcription. Here, we assess the impact of apabetalone on ex vivo inflammatory responses of monocytes from DM2 + CVD patients. Results Monocytes isolated from DM2 + CVD patients and matched controls were treated ex vivo with apabetalone, interferon γ (IFNγ), IFNγ + apabetalone or vehicle and phenotyped for gene expression and protein secretion. Unstimulated DM2 + CVD monocytes had higher baseline IL-1α, IL-1β and IL-8 cytokine gene expression and Toll-like receptor (TLR) 2 surface abundance than control monocytes, indicating pro-inflammatory activation. Further, DM2 + CVD monocytes were hyper-responsive to stimulation with IFNγ, upregulating genes within cytokine and NF-κB pathways > 30% more than control monocytes (p < 0.05). Ex vivo apabetalone treatment countered cytokine secretion by DM2 + CVD monocytes at baseline (GROα and IL-8) and during IFNγ stimulation (IL-1β and TNFα). Apabetalone abolished pro-inflammatory hyper-activation by reducing TLR and cytokine gene signatures more robustly in DM2 + CVD versus control monocytes. Conclusions Monocytes isolated from DM2 + CVD patients receiving standard of care therapies are in a hyper-inflammatory state and hyperactive upon IFNγ stimulation. Apabetalone treatment diminishes this pro-inflammatory phenotype, providing mechanistic insight into how BET protein inhibition may reduce CVD risk in DM2 patients.
Collapse
Affiliation(s)
- Sylwia Wasiak
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Kim E Dzobo
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Brooke D Rakai
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Yannick Kaiser
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Miranda Versloot
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Mahnoush Bahjat
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Stephanie C Stotz
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Li Fu
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Michael Sweeney
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Jan O Johansson
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Norman C W Wong
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada
| | - Erik S G Stroes
- Department of Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Jeffrey Kroon
- Department of Experimental Vascular Medicine, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Ewelina Kulikowski
- Resverlogix Corp, 300-4820 Richard Road SW, Calgary, AB, T3E 6L1, Canada.
| |
Collapse
|
15
|
Øbro NF, Grinfeld J, Belmonte M, Irvine M, Shepherd MS, Rao TN, Karow A, Riedel LM, Harris OB, Baxter EJ, Nangalia J, Godfrey A, Harrison CN, Li J, Skoda RC, Campbell PJ, Green AR, Kent DG. Longitudinal Cytokine Profiling Identifies GRO-α and EGF as Potential Biomarkers of Disease Progression in Essential Thrombocythemia. Hemasphere 2020; 4:e371. [PMID: 32647796 PMCID: PMC7306314 DOI: 10.1097/hs9.0000000000000371] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/09/2020] [Indexed: 12/17/2022] Open
Abstract
Myeloproliferative neoplasms (MPNs) are characterized by deregulation of mature blood cell production and increased risk of myelofibrosis (MF) and leukemic transformation. Numerous driver mutations have been identified but substantial disease heterogeneity remains unexplained, implying the involvement of additional as yet unidentified factors. The inflammatory microenvironment has recently attracted attention as a crucial factor in MPN biology, in particular whether inflammatory cytokines and chemokines contribute to disease establishment or progression. Here we present a large-scale study of serum cytokine profiles in more than 400 MPN patients and identify an essential thrombocythemia (ET)-specific inflammatory cytokine signature consisting of Eotaxin, GRO-α, and EGF. Levels of 2 of these markers (GRO-α and EGF) in ET patients were associated with disease transformation in initial sample collection (GRO-α) or longitudinal sampling (EGF). In ET patients with extensive genomic profiling data (n = 183) cytokine levels added significant prognostic value for predicting transformation from ET to MF. Furthermore, CD56+CD14+ pro-inflammatory monocytes were identified as a novel source of increased GRO-α levels. These data implicate the immune cell microenvironment as a significant player in ET disease evolution and illustrate the utility of cytokines as potential biomarkers for reaching beyond genomic classification for disease stratification and monitoring.
Collapse
Affiliation(s)
- Nina F. Øbro
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Jacob Grinfeld
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Miriam Belmonte
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5NG, United Kingdom
| | - Melissa Irvine
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Mairi S. Shepherd
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Tata Nageswara Rao
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Axel Karow
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
- Department of Pediatrics and Adolescent Medicine, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lisa M. Riedel
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Oliva B. Harris
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - E. Joanna Baxter
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Jyoti Nangalia
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Anna Godfrey
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - Claire N. Harrison
- Department of Hematology, Guy's and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Juan Li
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
| | - Radek C. Skoda
- Experimental Hematology, Department of Biomedicine, University Hospital Basel and University of Basel, 4031 Basel, Switzerland
| | - Peter J. Campbell
- Wellcome Trust Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Anthony R. Green
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, Cambridge University Hospitals NHS Foundation Trust, Hills Road, Cambridge CB2 0QQ, United Kingdom
| | - David G. Kent
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Hills Road, Cambridge, CB2 0XY, United Kingdom
- Department of Hematology, University of Cambridge, CB2 0XY, United Kingdom
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5NG, United Kingdom
| |
Collapse
|
16
|
Wang XH, Yu HL, Zou WB, Mi CH, Dai GJ, Zhang T, Zhang GX, Xie KZ, Wang JY. Study of the Relationship between Polymorphisms in the IL-8 Gene Promoter Region and Coccidiosis Resistance Index in Jinghai Yellow Chickens. Genes (Basel) 2020; 11:genes11050476. [PMID: 32349370 PMCID: PMC7291339 DOI: 10.3390/genes11050476] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 11/16/2022] Open
Abstract
Interleukin 8 (IL-8) participates in the immune response and has the function of inducing neutrophils to release lysosomal enzymes and eliminate pathogens. This study was to investigate the effect of single nucleotide mutations in the IL-8 gene promoter region on the coccidiosis resistance index. In this study, 180 infected Eimeria tenella (E. tenella) Jinghai yellow chickens were used as experimental samples. DNA sequencing technology was used to detect single nucleotide polymorphisms (SNPs) in the IL-8 gene promoter region. The association between these SNPs and coccidiosis resistance indexes (including superoxide dismutase (SOD), malondialdehyde (MDA), glutathione peroxidase (GSH-PX), catalase (CAT), nitric oxide (NO), interleukin-1β (IL-1β), interleukin-2 (IL-2), interleukin-6 (IL-6), IL-8, and interferon-γ (IFN-γ)) were analyzed. Three SNPs (T-550C, G-398T, and T-360C) were detected. Significant associations were found between each genotype at the T-550C site with NO (p-value = 0.006) and IL-8 (p-value = 0.034) indexes. Significant associations were found between each genotype at the G-398T site with SOD (p-value = 0.042), CAT (p-value = 0.049), NO (p-value = 0.008), and IL-2 (p-value = 0.044) indexes. Significant associations were found between each genotype at the T-360C site with SOD (p-value = 0.007), NO (p-value = 0.046), IL-2 (p-value = 0.041), IL-8 (p-value = 0.039), and IFN-γ (p-value = 0.042) indexes. Haplotype analysis showed that multiple indexes of the H1H3 haplotype combination were significantly higher than other haplotype combinations. Therefore, mutation of the IL-8 gene promoter region has a significant regulatory effect on the coccidiosis resistance index, with a change in transcription factor binding potentially altering IL-8 gene expression, thereby further affecting the IL-8 level in plasma. However, the specific mechanism needs further study.
Collapse
Affiliation(s)
- Xiao-Hui Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| | - Hai-Liang Yu
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| | - Wen-Bin Zou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| | - Chang-Hao Mi
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| | - Guo-Jun Dai
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
- Correspondence: ; Tel.: +86-139-5275-0903
| | - Tao Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| | - Gen-Xi Zhang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| | - Kai-Zhou Xie
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| | - Jin-Yu Wang
- College of Animal Science and Technology, Yangzhou University, Yangzhou, Jiangsu 225009, China; (X.-H.W.); (H.-L.Y.); (W.-B.Z.); (C.-H.M.); (T.Z.); (G.-X.Z.); (K.-Z.X.); (J.-Y.W.)
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou, Jiangsu 225009, China
| |
Collapse
|
17
|
Scally C, Abbas H, Ahearn T, Srinivasan J, Mezincescu A, Rudd A, Spath N, Yucel-Finn A, Yuecel R, Oldroyd K, Dospinescu C, Horgan G, Broadhurst P, Henning A, Newby DE, Semple S, Wilson HM, Dawson DK. Myocardial and Systemic Inflammation in Acute Stress-Induced (Takotsubo) Cardiomyopathy. Circulation 2019; 139:1581-1592. [PMID: 30586731 DOI: 10.1161/circulationaha.118.037975] [Citation(s) in RCA: 191] [Impact Index Per Article: 31.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND Acute stress-induced (takotsubo) cardiomyopathy can result in a heart failure phenotype with a prognosis comparable with that of myocardial infarction. In this study, we hypothesized that inflammation is central to the pathophysiology and natural history of takotsubo cardiomyopathy. METHODS In a multicenter study, we prospectively recruited 55 patients with takotsubo cardiomyopathy and 51 age-, sex-, and comorbidity-matched control subjects. During the index event and at the 5-month follow-up, patients with takotsubo cardiomyopathy underwent multiparametric cardiac magnetic resonance imaging, including ultrasmall superparamagnetic particles of iron oxide (USPIO) enhancement for detection of inflammatory macrophages in the myocardium. Blood monocyte subpopulations and serum cytokines were assessed as measures of systemic inflammation. Matched control subjects underwent investigation at a single time point. RESULTS Subjects were predominantly middle-aged (64±14 years) women (90%). Compared with control subjects, patients with takotsubo cardiomyopathy had greater USPIO enhancement (expressed as the difference between pre-USPIO and post-USPIO T2*) in both ballooning (14.3±0.6 milliseconds versus 10.5±0.9 milliseconds; P<0.001) and nonballooning (12.9±0.6 milliseconds versus 10.5±0.9 milliseconds; P=0.02) left ventricular myocardial segments. Serum interleukin-6 (23.1±4.5 pg/mL versus 6.5±5.8 pg/mL; P<0.001) and chemokine (C-X-C motif) ligand 1 (1903±168 pg/mL versus 1272±177 pg/mL; P=0.01) concentrations and classic CD14++CD16- monocytes (90±0.5% versus 87±0.9%; P=0.01) were also increased whereas intermediate CD14++CD16+ (5.4±0.3% versus 6.9±0.6%; P=0.01) and nonclassic CD14+CD16++ (2.7±0.3% versus 4.2±0.5%; P=0.006) monocytes were reduced in patients with takotsubo cardiomyopathy. At 5 months, USPIO enhancement was no longer detectable in the left ventricular myocardium, although persistent elevations in serum interleukin-6 concentrations ( P=0.009) and reductions in intermediate CD14++CD16+ monocytes (5.6±0.4% versus 6.9±0.6%; P=0.01) remained. CONCLUSIONS We demonstrate for the first time that takotsubo cardiomyopathy is characterized by a myocardial macrophage inflammatory infiltrate, changes in the distribution of monocyte subsets, and an increase in systemic proinflammatory cytokines. Many of these changes persisted for at least 5 months, suggesting a low-grade chronic inflammatory state. CLINICAL TRIAL REGISTRATION URL: https://www.clinicaltrials.gov . Unique identifier: NCT02897739.
Collapse
Affiliation(s)
- Caroline Scally
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Hassan Abbas
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Trevor Ahearn
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Janaki Srinivasan
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Alice Mezincescu
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Amelia Rudd
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Nicholas Spath
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, UK (N.S., D.E.N., S.S.)
| | - Alim Yucel-Finn
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Raif Yuecel
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Keith Oldroyd
- West of Scotland Regional Heart & Lung Centre, Glasgow, UK (K.O.)
| | - Ciprian Dospinescu
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Graham Horgan
- Biomathematics & Statistics Scotland, Aberdeen, UK (G.H.)
| | - Paul Broadhurst
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | | | - David E Newby
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, UK (N.S., D.E.N., S.S.)
| | - Scott Semple
- BHF Centre for Cardiovascular Sciences, University of Edinburgh, UK (N.S., D.E.N., S.S.)
| | - Heather M Wilson
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| | - Dana K Dawson
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, UK (C.S., H.A., T.A., J.S., A.M., A.R., A.Y.-F., R.Y., C.D., P.B., H.M.W., D.K.D.)
| |
Collapse
|
18
|
Burja B, Kuret T, Janko T, Topalović D, Živković L, Mrak-Poljšak K, Spremo-Potparević B, Žigon P, Distler O, Čučnik S, Sodin-Semrl S, Lakota K, Frank-Bertoncelj M. Olive Leaf Extract Attenuates Inflammatory Activation and DNA Damage in Human Arterial Endothelial Cells. Front Cardiovasc Med 2019; 6:56. [PMID: 31157238 PMCID: PMC6531989 DOI: 10.3389/fcvm.2019.00056] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 04/16/2019] [Indexed: 12/27/2022] Open
Abstract
Olive leaf extract (OLE) is used in traditional medicine as a food supplement and as an over-the-counter drug for a variety of its effects, including anti-inflammatory and anti-atherosclerotic ones. Mechanisms through which OLE could modulate these pathways in human vasculature remain largely unknown. Serum amyloid A (SAA) plays a causal role in atherosclerosis and cardiovascular diseases and induces pro-inflammatory and pro-adhesive responses in human coronary artery endothelial cells (HCAEC). Within this study we explored whether OLE can attenuate SAA-driven responses in HCAEC. HCAEC were treated with SAA (1,000 nM) and/or OLE (0.5 and 1 mg/ml). The expression of adhesion molecules VCAM-1 and E-selectin, matrix metalloproteinases (MMP2 and MMP9) and microRNA 146a, let-7e, and let-7g (involved in the regulation of inflammation) was determined by qPCR. The amount of secreted IL-6, IL-8, MIF, and GRO-α in cell culture supernatants was quantified by ELISA. Phosphorylation of NF-κB was assessed by Western blot and DNA damage was measured using the COMET assay. OLE decreased significantly released protein levels of IL-6 and IL-8, as well as mRNA expression of E-selectin in SAA-stimulated HCAEC and reduced MMP2 levels in unstimulated cells. Phosphorylation of NF-κB (p65) was upregulated in the presence of SAA, with OLE significantly attenuating this SAA-induced effect. OLE stabilized SAA-induced upregulation of microRNA-146a and let-7e in HCAEC, suggesting that OLE could fine-tune the SAA-driven activity of NF-κB by changing the microRNA networks in HCAEC. SAA induced DNA damage and worsened the oxidative DNA damage in HCAEC, whereas OLE protected HCAEC from SAA- and H2O2-driven DNA damage. OLE significantly attenuated certain pro-inflammatory and pro-adhesive responses and decreased DNA damage in HCAEC upon stimulation with SAA. The reversal of SAA-driven endothelial activation by OLE might contribute to its anti-inflammatory and anti-atherogenic effects in HCAEC.
Collapse
Affiliation(s)
- Blaž Burja
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia.,Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Tadeja Kuret
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia.,Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Tea Janko
- Faculty of Mathematics, Natural Science and Information Technology, University of Primorska, Koper, Slovenia
| | - Dijana Topalović
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | - Lada Živković
- Department of Pathobiology, Faculty of Pharmacy, University of Belgrade, Belgrade, Serbia
| | | | | | - Polona Žigon
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| | - Saša Čučnik
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia.,Chair of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Snezna Sodin-Semrl
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia.,Faculty of Mathematics, Natural Science and Information Technology, University of Primorska, Koper, Slovenia
| | - Katja Lakota
- Department of Rheumatology, University Medical Centre, Ljubljana, Slovenia.,Faculty of Mathematics, Natural Science and Information Technology, University of Primorska, Koper, Slovenia
| | - Mojca Frank-Bertoncelj
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
19
|
Lenzi C, Stevens J, Osborn D, Hannah MJ, Bierings R, Carter T. Synaptotagmin 5 regulates Ca 2+-dependent Weibel-Palade body exocytosis in human endothelial cells. J Cell Sci 2019; 132:jcs.221952. [PMID: 30659119 DOI: 10.1242/jcs.221952] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 01/09/2019] [Indexed: 12/11/2022] Open
Abstract
Elevations of intracellular free Ca2+ concentration ([Ca2+]i) are a potent trigger for Weibel-Palade body (WPB) exocytosis and secretion of von Willebrand factor (VWF) from endothelial cells; however, the identity of WPB-associated Ca2+-sensors involved in transducing acute increases in [Ca2+]i into granule exocytosis remains unknown. Here, we show that synaptotagmin 5 (SYT5) is expressed in human umbilical vein endothelial cells (HUVECs) and is recruited to WPBs to regulate Ca2+-driven WPB exocytosis. Western blot analysis of HUVECs identified SYT5 protein, and exogenously expressed SYT5-mEGFP localised almost exclusively to WPBs. shRNA-mediated knockdown of endogenous SYT5 (shSYT5) reduced the rate and extent of histamine-evoked WPB exocytosis and reduced secretion of the WPB cargo VWF-propeptide (VWFpp). The shSYT5-mediated reduction in histamine-evoked WPB exocytosis was prevented by expression of shRNA-resistant SYT5-mCherry. Overexpression of SYT5-EGFP increased the rate and extent of histamine-evoked WPB exocytosis, and increased secretion of VWFpp. Expression of a Ca2+-binding defective SYT5 mutant (SYT5-Asp197Ser-EGFP) mimicked depletion of endogenous SYT5. We identify SYT5 as a WPB-associated Ca2+ sensor regulating Ca2+-dependent secretion of stored mediators from vascular endothelial cells.
Collapse
Affiliation(s)
- Camille Lenzi
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW18 ORE, UK
| | | | - Daniel Osborn
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW18 ORE, UK
| | - Matthew J Hannah
- Microbiology Services Colindale, Public Health England, London, NW9 5EQ, UK
| | - Ruben Bierings
- Plasma Proteins, Sanquin Research and Landsteiner Laboratory, Academic Medical Centre, University of Amsterdam, 1006 AD Amsterdam, PO Box 9190, The Netherlands
| | - Tom Carter
- Molecular and Clinical Sciences Research Institute, St George's, University of London, London SW18 ORE, UK
| |
Collapse
|
20
|
Kitagori K, Yoshifuji H, Oku T, Ayaki T, Kuzuya A, Nakajima T, Akizuki S, Nakashima R, Murakami K, Ohmura K, Hirayama Y, Takahashi R, Mimori T. Utility of osteopontin in cerebrospinal fluid as a diagnostic marker for neuropsychiatric systemic lupus erythematosus. Lupus 2019; 28:414-422. [DOI: 10.1177/0961203319828818] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The whole protein of osteopontin (OPN full) and its cleaved form (OPN N-half) are involved in the immune response and the migration of immune cells to an inflammatory lesion. We have reported that serum OPN full and urine OPN N-half are elevated in lupus nephritis (LN). Neuropsychiatric systemic lupus erythematosus (NPSLE) is a refractory complication of SLE. To investigate whether OPN full and OPN N-half could serve as diagnostic markers for NPSLE, and to elucidate their role in NPSLE pathogenesis, the concentrations of OPN full and OPN N-half in cerebrospinal fluid (CSF) were measured in NPSLE and non-NPSLE patients. We found that the concentration of OPN full in the CSF was significantly higher in NPSLE than in non-NPSLE, and it decreased after treatment. When the cutoff value of OPN full in CSF was set to 963.4 ng/ml, the sensitivity and specificity for the diagnosis of NPSLE were 70% and 100%, respectively. The correlation analysis of OPN full, OPN N-half and various cytokines/chemokines suggested that the cytokines/chemokines could be divided into two clusters: cluster A, which contains OPN full and cluster B, which contains interleukin-6. OPN full in CSF could be a novel diagnostic marker for NPSLE.
Collapse
Affiliation(s)
- K Kitagori
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - H Yoshifuji
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - T Oku
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Candidate Discovery Science Labs, Astellas Pharma Inc, Ibaraki, Japan
| | - T Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - A Kuzuya
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - T Nakajima
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - S Akizuki
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - R Nakashima
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - K Murakami
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - K Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Y Hirayama
- Center for Innovation in Immunoregulative Technology and Therapeutics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Candidate Discovery Science Labs, Astellas Pharma Inc, Ibaraki, Japan
| | - R Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - T Mimori
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
21
|
Vroegindewey MM, Oemrawsingh RM, Kardys I, Asselbergs FW, van der Harst P, Oude Ophuis AJ, Etienne Cramer G, Maas A, Hong Kie The S, Wardeh AJ, Mouthaan H, Boersma E, Akkerhuis KM. The temporal pattern of immune and inflammatory proteins prior to a recurrent coronary event in post-acute coronary syndrome patients. Biomarkers 2018; 24:199-205. [PMID: 30514120 DOI: 10.1080/1354750x.2018.1539768] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
PURPOSE We assessed the temporal pattern of 29 immune and inflammatory proteins in post-acute coronary syndrome (ACS) patients, prior to the development of recurrent ACS. METHODS High-frequency blood sampling was performed in 844 patients admitted for ACS during one-year follow-up. We conducted a case-control study on the 45 patients who experienced reACS (cases) and two matched event-free patients (controls) per case. Olink Proteomics' immunoassay was used to obtain serum levels of the 29 proteins, expressed in an arbitrary unit on the log2-scale (Normalized Protein eXpression, NPX). Linear mixed-effects models were applied to examine the temporal pattern of the proteins, and to illustrate differences between cases and controls. RESULTS Mean age was 66 ± 12 years and 80% were men. Cases and controls had similar baseline clinical characteristics. During the first 30 days, and after multiple testing correction, cases had significantly higher serum levels of CXCL1 (difference of 1.00 NPX, p = 0.002), CD84 (difference of 0.64 NPX, p = 0.002) and TNFRSF10A (difference of 0.41 NPX, p < 0.001) than controls. After 30 days, serum levels of all 29 proteins were similar in cases and controls. In particular, no increase was observed prior to reACS. CONCLUSIONS Among 29 immune and inflammatory proteins, CXCL1, CD84 and TNFRSF10A were associated with early reACS after initial ACS-admission.
Collapse
Affiliation(s)
- Maxime M Vroegindewey
- a Department of Cardiology , Erasmus University Medical Centre , Rotterdam , The Netherlands
| | - Rohit M Oemrawsingh
- a Department of Cardiology , Erasmus University Medical Centre , Rotterdam , The Netherlands.,b Department of Cardiology , Amphia Hospital , Breda , The Netherlands
| | - Isabella Kardys
- a Department of Cardiology , Erasmus University Medical Centre , Rotterdam , The Netherlands
| | - Folkert W Asselbergs
- c Department of Cardiology Division Heart & Lungs , University Medical Centre, Utrecht University of Utrecht , Utrecht , The Netherlands.,d Durrer Centre for Cardiovascular Research Netherlands Heart Institute , Utrecht , The Netherlands.,e Faculty of Population Health Sciences , Institute of Cardiovascular Science, University College London , London , UK.,f Farr Institute of Health Informatics Research and Institute of Health Informatics, University College London , London , UK
| | | | - Anton J Oude Ophuis
- h Canisius-Wilhelmina Hospital , Nijmegen , The Netherlands.,i Working Group on Cardiovascular Research the Netherlands (WCN) , Utrecht , The Netherlands
| | - G Etienne Cramer
- j Radboud University Medical Center , Nijmegen , The Netherlands
| | - Arthur Maas
- k Gelre Hospital , Zutphen , The Netherlands
| | - S Hong Kie The
- l Treant Zorggroep, Bethesda , Hoogeveen , The Netherlands
| | | | | | - Eric Boersma
- a Department of Cardiology , Erasmus University Medical Centre , Rotterdam , The Netherlands
| | - K Martijn Akkerhuis
- a Department of Cardiology , Erasmus University Medical Centre , Rotterdam , The Netherlands
| |
Collapse
|
22
|
Li B, Sheng Z, Liu C, Qian L, Wu Y, Wu Y, Ma G, Yao Y. Kallistatin Inhibits Atherosclerotic Inflammation by Regulating Macrophage Polarization. Hum Gene Ther 2018; 30:339-351. [PMID: 30205711 DOI: 10.1089/hum.2018.084] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Kallistatin (KS) has been recognized as a plasma protein with anti-inflammatory functions. Macrophages are the primary inflammatory cells in atherosclerotic plaques. However, it is unknown whether KS plays a role in macrophage development and the pathogenesis of atherosclerosis. This study investigated the role of KS in macrophage development, a key pathological process in atherosclerosis. An atherosclerosis model was established in ApoE-/- mice via partial left carotid artery (PLCA) ligation. An adenovirus vector (Ad. HKS) containing the human KS gene was delivered via the tail vein before PLCA ligation. The mice were divided into two groups: the PLCA + Ad. HKS and PLCA + adenovirus vector (Ad. Null) groups and followed for 2 and 4 weeks. Human KS was expressed in the mice after KS gene delivery. In addition, KS significantly inhibited plaque formation and reduced inflammation in the plaques and liver 4 weeks after gene delivery. Moreover, KS gene delivery significantly increased the expression of interleukin-10 and Arginase 1, which are M2 macrophage markers, and reduced the expression of inducible nitric oxide synthase and monocyte chemotactic protein 1, which are M1 macrophage markers. Furthermore, in cultured RAW 264.7 macrophages, KS significantly stimulated M2 marker expression and differentiation and decreased M1 marker expression, as determined by flow cytometry and real-time polymerase chain reaction. These effects were blocked by Krüppel-like factor 4 small-interfering RNA oligonucleotides. These findings demonstrate that KS inhibits atherosclerotic plaque formation and regulates M1/M2 macrophage polarization via Krüppel-like factor 4 activation.
Collapse
Affiliation(s)
- Bing Li
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| | - Zulong Sheng
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| | - Chang Liu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| | - Linglin Qian
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| | - Yuehuan Wu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| | - Yanping Wu
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| | - Genshan Ma
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| | - Yuyu Yao
- Department of Cardiology, Zhongda Hospital, Medical School of Southeast University, Nanjing, P.R. China
| |
Collapse
|
23
|
Li W, Huang H, Li L, Wang L, Li Y, Wang Y, Guo S, Li L, Wang D, He Y, Chen L. The Pathogenesis of Atherosclerosis Based on Human Signaling Networks and Stem Cell Expression Data. Int J Biol Sci 2018; 14:1678-1685. [PMID: 30416382 PMCID: PMC6216023 DOI: 10.7150/ijbs.27896] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023] Open
Abstract
Atherosclerosis is a common and complex disease, whose morbidity increased significantly. Here, an integrated approach was proposed to elucidate systematically the pathogenesis of atherosclerosis from a systems biology point of view. Two weighted human signaling networks were constructed based on atherosclerosis related gene expression data of stem cells. Then, 37 candidate Atherosclerosis-risk Modules were detected using four kinds of permutation tests. Five Atherosclerosis-risk Modules (three Absent Modules and two Emerging Modules) enriched in functions significantly associated with disease genes were identified and verified to be associated with the maintenance of normal biological process and the pathogenesis and development of atherosclerosis. Especially for Atherosclerosis-risk Emerging Module P96, it could distinguish between normal and disease samples by Supporting Vector Machine with the average expression value of the module as classification feature. These identified modules and their genes may act as potential atherosclerosis biomarkers. Our study would shed light on the signal transduction of atherosclerosis, and provide new insights to its pathogenesis from the perspective of stem cells.
Collapse
Affiliation(s)
- Wan Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Hao Huang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Lei Li
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Li Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yong Li
- Dean's Office, Harbin Medical University, Harbin, Heilongjiang, China
| | - Yahui Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Shanshan Guo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Liansheng Li
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Donghua Wang
- Department of general surgery, General Hospital of Heilongjiang Province Land Reclamation Bureau, 150088, Harbin, China
| | - Yuehan He
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| | - Lina Chen
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
24
|
Neuschäfer-Rube F, Pathe-Neuschäfer-Rube A, Hippenstiel S, Püschel GP. PGE 2 enhanced TNFα-mediated IL-8 induction in monocytic cell lines and PBMC. Cytokine 2018; 113:105-116. [PMID: 29929938 DOI: 10.1016/j.cyto.2018.06.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 06/12/2018] [Accepted: 06/13/2018] [Indexed: 12/31/2022]
Abstract
BACKGROUND & PURPOSE Recent studies suggested a role of prostaglandin E2 (PGE2) in the expression of the chemokine IL-8 by monocytes. The function of EP4 receptor for TNFα-induced IL-8 expression was studied in monocytic cell lines. EXPERIMENTAL APPROACH IL-8 mRNA and protein induction as well as IL-8 promoter activity and transcription factor activation were assessed in monocytic cell lines, primary blood mononuclear cells (PBMC) and transgenic HEK293 cells expressing the EP4 receptor. KEY RESULTS In monocytic cell lines THP-1, MonoMac and U937 PGE2 had only a marginal impact on IL-8 induction but strongly enhanced TNFα-induced IL-8 mRNA and protein synthesis. Similarly, in PBMC IL-8 mRNA induction was larger by simultaneous stimulation with TNFα and PGE2 than by either stimulus alone. The EP4 receptor subtype was the most abundant EP receptor in all three cell lines and in PBMC. Stimulation of THP-1 cells with an EP4 specific agonist enhanced TNFα-induced IL-8 mRNA and protein formation to the same extent as PGE2. In HEK293 cells expressing EP4, but not in wild type HEK293 cells lacking EP4, PGE2 enhanced TNFα-induced IL-8 protein and mRNA synthesis. In THP-1 cells, the enhancement of TNFα-mediated IL-8 mRNA induction by PGE2 was mimicked by a PKA-activator. Furthermore in these cells PGE2 induced expression of transcription factor C/EBPß, enhanced NF-κB activation by TNFα and inhibited TNFα-mediated AP-1 activation. PGE2 and TNFα synergistically activated transcription factor CREB, induced C/EBPß expression and enhanced the activity of an IL-8 promoter fragment containing -223 bp upstream of the transcription start site. CONCLUSIONS AND IMPLICATIONS These findings suggest that a combined stimulation of TNFα and PGE2/EP4 signal chains in monocytic cells leads to maximal IL-8 promoter activity, as well as IL-8 mRNA and protein induction, by activating the PKA/CREB/C/EBPß as well as NF-κB signal chains.
Collapse
Affiliation(s)
- F Neuschäfer-Rube
- Universität Potsdam, Institut für Ernährungswissenschaft, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany.
| | - A Pathe-Neuschäfer-Rube
- Universität Potsdam, Institut für Ernährungswissenschaft, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| | - S Hippenstiel
- Charité - Universitätsmedizin Berlin, Dept. of Internal Medicine/Infectious Diseases and Respiratory Medicine, Augustenburger Platz 1, 13353 Berlin, Germany
| | - G P Püschel
- Universität Potsdam, Institut für Ernährungswissenschaft, Arthur-Scheunert-Allee 114-116, 14558 Nuthetal, Germany
| |
Collapse
|
25
|
Rakic M, Persic V, Kehler T, Bastiancic AL, Rosovic I, Laskarin G, Sotosek Tokmadzic V. Possible role of circulating endothelial cells in patients after acute myocardial infarction. Med Hypotheses 2018; 117:42-46. [PMID: 30077195 DOI: 10.1016/j.mehy.2018.06.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/22/2018] [Accepted: 06/05/2018] [Indexed: 01/16/2023]
Abstract
Acute myocardial infarction (AMI) occurs as a result of insufficient myocardial perfusion leading to cell necrosis. This is most commonly due to the obstruction of the coronary artery by ruptured atherosclerotic plaque and thrombosis. Damaged ischemic and necrotic myocardial cells release pro-inflammatory substances in tissue and plasma, leading to a systemic inflammatory response. Profound systemic inflammatory response during ischemia/reperfusion injury causes disruption of endothelial glycocalyx and detachment of endothelial cells that express von Willebrant factor (vWF). We hypothesize that circulating vWF+ endothelial cells could act as antigen presenting cells which interact with T and NK cells directly, by cell to cell contact and indirectly by cytokine and chemokine secretion, leading to the immune response towards inflammation. Analyzing the frequency, phenotype and pro-inflammatory substances produced in circulating vWF positive (+) cells in patients with AMI could be beneficial to determine the severity of the pro-inflammatory response, according to the level of endothelial dysfunction in the early period of AMI. To evaluate these hypotheses, we suggest to determine frequency, phenotype, and ability of cytokine/chemokine production in circulating vWF+ endothelial cells by simultaneous surface and intracellular cell staining, and flow cytometry analysis. Secretion of pro-inflammatory cytokines and chemokines, pro-atherogenic substances and the components of glycocalyx might be measured in supernatants of magnetically separated or sorted vWF+ endothelial cells, as well as in the serum of a patient with acute AMI by enzyme linked-immunoassay tests. The interaction of increasing concentrations of isolated circulating vWF+ endothelial cells and cognate T and NK cells might be investigated by lymphocyte proliferation rate, cytotoxic mediators' expression, and cytokine production. If our hypothesis is correct, characterization of circulating vWF+ endothelial cells could grant us greater insight into their role in pathophysiology of AMI and the degree of myocardial damage.
Collapse
Affiliation(s)
- Marijana Rakic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Viktor Persic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia; Department of Medical Rehabilitation, Medical Faculty, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia
| | - Tatjana Kehler
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia
| | - Ana Lanca Bastiancic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Ivan Rosovic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Gordana Laskarin
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Hospital for Medical Rehabilitation of Hearth and Lung Diseases and Rheumatism "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia; Department of Physiology and Immunology, Medical Faculty University of Rijeka, B.Branchetta 20, 51000 Rijeka, Croatia
| | - Vlatka Sotosek Tokmadzic
- Department of Anesthesiology, Reanimatology and Intensive Care Medicine, Faculty of Medicine, University of Rijeka, Brace Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
26
|
Persic V, Bastiancic AL, Rosovic I, Raljevic D, Samsa DT, Bastiancic L, Miskulin R, Boban M, Laskarin G. Correlation between immunological-inflammatory markers and endothelial disfunction in the early stage of coronary heart disease. Med Hypotheses 2018; 115:72-76. [PMID: 29685202 DOI: 10.1016/j.mehy.2018.04.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2017] [Accepted: 04/08/2018] [Indexed: 01/06/2023]
Abstract
Classical risk factors for endothelial dysfunction (ED), such as age, gender, total cholesterol, high-density lipoprotein cholesterol, systolic blood pressure, and smoking history are utilised for the Framingham score and Systemic Coronary Risk Estimation (SCORE) for evaluation of the 10-year cardiovascular risk in routine practice. Nonetheless, pro-inflammatory mediators are deeply involved in the initiation and the progression of ED and coronary artery disease (CAD), and act additionally or independently of metabolic factors before clinical manifestations of the disease appear. C-reactive protein, a marker of intimal thickening of the myeloid-related protein 8/14 heterodimer, monocyte chemotactic protein 1, interleukin-15, the cytotoxic mediator, granulysin, and the matrix metalloproteinase 9 could be valuable, single, fast, and non-invasive laboratory tools for ED deterioration degree assessment. We propose to investigate the impact of pro-inflammatory biomarkers on ED, measured by previously established clinical methods in patients with yet undiagnosed CAD and at medium risk for an acute coronary event. It could be useful to measure and correlate the concentration of particular inflammatory markers in peripheral blood samples and the results of the Framingham and SCORE charts, multi-slice computed tomography coronary angiography, echocardiography, brachial artery flow-mediated dilatation, carotid-femoral pulse wave velocity, ankle-brachial index, carotid wall thickening, myocardial perfusion scintigraphy, and particularly, cardiac magnetic resonance imaging. The goal would be that the degree of correlation between particular inflammatory markers and the results of some methods for the assessment of ED or cardiac ischaemic imaging could be emphasised and pro-inflammatory markers positioned in the pathogenetic algorithm of CAD.
Collapse
Affiliation(s)
- Viktor Persic
- Division of Cardiology, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia; Department of Medical Rehabilitation, Medical Faculty, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia.
| | - Ana Lanca Bastiancic
- Division of Cardiology, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia.
| | - Ivan Rosovic
- Division of Cardiology, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia
| | - Damir Raljevic
- Division of Cardiology, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia
| | - Dijana Travica Samsa
- Division of Cardiology, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia
| | - Luka Bastiancic
- Department of Cardiology, Clinical Hospital Rijeka, Kresimirova 42, 51000 Rijeka, Croatia
| | - Rajko Miskulin
- Division of Cardiology, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia
| | - Marko Boban
- Division of Cardiology, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia; Department of Medical Rehabilitation, Medical Faculty, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia
| | - Gordana Laskarin
- Department of Rheumatology, Rehabilitation, and Physical Medicine, Special Hospital for Medical Rehabilitation of Heart, Lungs and Rheumatic diseases "Thalassotherapia-Opatija", 51410 Opatija, M. Tita 188, Croatia; Department of Physiology and Immunology, Medical Faculty, University of Rijeka, B. Branchetta 20, 51000 Rijeka, Croatia.
| |
Collapse
|
27
|
Jin L, Li ZF, Wang DK, Sun M, Qi W, Ma Q, Zhang L, Chu C, Chan EY, Lee SS, Wise H, To KF, Shi Y, Zhou N, Cheung WT. Molecular and functional characterization of tumor-induced factor (TIF): Hamster homolog of CXCL3 (GROγ) displays tumor suppressive activity. Cytokine 2018; 102:62-75. [DOI: 10.1016/j.cyto.2017.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2017] [Revised: 12/11/2017] [Accepted: 12/15/2017] [Indexed: 12/20/2022]
|
28
|
Rödling L, Schwedhelm I, Kraus S, Bieback K, Hansmann J, Lee-Thedieck C. 3D models of the hematopoietic stem cell niche under steady-state and active conditions. Sci Rep 2017; 7:4625. [PMID: 28676663 PMCID: PMC5496931 DOI: 10.1038/s41598-017-04808-0] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Accepted: 05/22/2017] [Indexed: 12/11/2022] Open
Abstract
Hematopoietic stem cells (HSCs) in the bone marrow are able to differentiate into all types of blood cells and supply the organism each day with billions of fresh cells. They are applied to cure hematological diseases such as leukemia. The clinical need for HSCs is high and there is a demand for being able to control and multiply HSCs in vitro. The hematopoietic system is highly proliferative and thus sensitive to anti-proliferative drugs such as chemotherapeutics. For many of these drugs suppression of the hematopoietic system is the dose-limiting toxicity. Therefore, biomimetic 3D models of the HSC niche that allow to control HSC behavior in vitro and to test drugs in a human setting are relevant for the clinics and pharmacology. Here, we describe a perfused 3D bone marrow analog that allows mimicking the HSC niche under steady-state and activated conditions that favor either HSC maintenance or differentiation, respectively, and allows for drug testing.
Collapse
Affiliation(s)
- Lisa Rödling
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Ivo Schwedhelm
- Institute for Tissue Engineering and Regenerative Medicine, University of Würzburg, 97070, Würzburg, Germany
| | - Saskia Kraus
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Karen Bieback
- Institute of Transfusion Medicine and Immunology Mannheim, Medical Faculty Mannheim, Heidelberg University; German Red Cross Blood Donor Service Baden-Württemberg-Hessen, 68167, Mannheim, Germany
| | - Jan Hansmann
- Institute for Tissue Engineering and Regenerative Medicine, University of Würzburg, 97070, Würzburg, Germany
| | - Cornelia Lee-Thedieck
- Karlsruhe Institute of Technology (KIT), Institute of Functional Interfaces, Hermann-von-Helmholtz-Platz 1, 76344, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
29
|
Sokolic J, Tokmadzic VS, Knezevic D, Medved I, Vukelic Damjani N, Balen S, Rakic M, Lanca Bastiancic A, Laskarin G. Endothelial dysfunction mediated by interleukin-18 in patients with ischemic heart disease undergoing coronary artery bypass grafting surgery. Med Hypotheses 2017; 104:20-24. [PMID: 28673582 DOI: 10.1016/j.mehy.2017.05.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 05/06/2017] [Indexed: 12/16/2022]
Abstract
When medication management or percutaneous coronary intervention is not successful in patients with advanced ischemic heart disease, surgical revascularisation-predominantly coronary artery bypass grafting (CABG)-is considered the gold standard. However, CABG surgery can lead to ischemia/reperfusion injury, which is characterized by a strong inflammatory response. Interleukin (IL)-18, is a strong inflammatory mediator, that is released from cardiomyocytes and can be found in the systemic circulation of patients during and immediately after CABG surgery. The existing damage of endothelial glycocalyx in patients with ischemic heart disease is further impaired concurrently during the surgery due to the anaesthesia-surgical technique used and intravascular fluid loading. This results in the increased incidence of adverse events, including myocardial infarction. IL-18 leads to the activation of lymphocyte cytotoxicity via cytotoxic mediators (Fas ligand, Tumour necrosis factor (TNF)-related apoptosis-inducing ligand, perforin, and granulysin). We hypothesize that IL-18 is released locally in the heart and the systemic circulation in patients undergoing CABG surgery and may be correlated with the level of activity of circulating lymphocytes. In turn, this may lead to lymphocyte-mediated cytotoxicity directed toward damaged and activated endothelial cells. Shear stress glycocalyx, as well as damaged and activated endothelial cells then become the main the source of pro-inflammatory cytokines, chemokines, and adhesion molecules. These attract activated lymphocytes to adhere to the endothelium or enter the subintimal layer, increasing existing or initiating the formation of new plaques, which leads to the development of myocardial infarction during or shortly after surgery. To evaluate our hypothesis, we will measure the local concentration of IL-18 in the sinus coronarius and systemic circulation. These values will then be correlated with immunological and biochemical parameters, predominantly with the concentration of degradation products of glycocalyx and cytotoxic mediators in activated lymphocytes. If our hypothesis is correct, measuring the IL-18 concentration that is responsible for glycocalyx deterioration, may become a useful tool for predicting myocardial infarction occurrence in patients undergoing CABG surgery.
Collapse
Affiliation(s)
- Jadranko Sokolic
- Clinic of Anesthesiology and Intensive Care Medicine, Clinical Hospital Center Rijeka, 51 000 Rijeka, Kresimirova 42, Croatia
| | - Vlatka Sotosek Tokmadzic
- Clinic of Anesthesiology and Intensive Care Medicine, Clinical Hospital Center Rijeka, 51 000 Rijeka, Kresimirova 42, Croatia; Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia.
| | - Danijel Knezevic
- Department of Anesthesiology, Reanimatology and Intensive Care, Faculty of Medicine, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia
| | - Igor Medved
- Department of Surgery, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Tome Strizica 3, Croatia
| | - Nada Vukelic Damjani
- Department of Transfusion Medicine, Clinical Hospital Center Rijeka, 51 000 Rijeka, Kresimirova 42, Croatia
| | - Sanja Balen
- Department of Transfusion Medicine, Clinical Hospital Center Rijeka, 51 000 Rijeka, Kresimirova 42, Croatia
| | - Marijana Rakic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Ana Lanca Bastiancic
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia
| | - Gordana Laskarin
- Division of Cardiology, Hospital for Medical Rehabilitation of the Hearth and Lung Diseases and Rheumatism "Thalassotherapia" Opatija, 51410 Opatija, M. Tita 188, Croatia; Department of Physiology and Immunology, Medical Faculty, University of Rijeka, 51000 Rijeka, B. Branchetta 20, Croatia
| |
Collapse
|
30
|
Huang J, Yang Q, He L, Huang J. Role of TLR4 and miR-155 in peripheral blood mononuclear cell-mediated inflammatory reaction in coronary slow flow and coronary arteriosclerosis patients. J Clin Lab Anal 2017; 32. [PMID: 28407320 DOI: 10.1002/jcla.22232] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 03/14/2017] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND To study the role of toll-like receptor 4 (TLR4) and MicroRNA-155 (miR-155) in the peripheral blood mononuclear cell (PBMC)-mediated inflammation in coronary slow flow (CSF) and coronary arteriosclerosis. METHODS Patients were divided into acute coronary syndrome (ACS), stable angina pectoris (SAP), CSF, and healthy control (HC) groups. The isolated PBMCs were treated with lipopolysaccharide (LPS)/and antagomiR-155. TLR4, miR-155, and the concentrations of tumor necrosis factor (TNF)-α, interleukin (IL)-6, IL-1, and IL-10 were measured. RESULTS Before LPS intervention, the TLR4, TNF-α, IL-1, and IL-6 levels were higher and the level of miR-155, IL-10 was lower in the ACS group compared with the SAP, CSF, and HC groups. After exposure to LPS, the levels of TLR4, miR-155, TNF-α, IL-1, and IL-6 were increased and the level of IL-10 was decreased in the SAP and CSF groups compared with the HC group. But when over-expressing antagomiR-155 into PBMCs from SAP and CSF groups, the miR-155 expression, and TNF-α, IL-6, and IL-1 secretion increase induced by LPS were restrained. CONCLUSIONS TLR4, miR-155, and inflammatory cytokines may be closely related to ACS. LPS can induce TLR4, miR-155 expression and inflammatory response in SAP and CSF patients. AntagomiR-155 can inhibit this inflammatory response.
Collapse
Affiliation(s)
- Jiangyan Huang
- Department of Cardiology, First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Qin Yang
- Department of Cardiology, First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Longmiao He
- Department of Cardiology, First Affiliated Hospital, Nanchang University, Nanchang, China
| | - Jun Huang
- Department of Cardiology, First Affiliated Hospital, Nanchang University, Nanchang, China
| |
Collapse
|
31
|
Fakhrudin N, Dwi Astuti E, Sulistyawati R, Santosa D, Susandarini R, Nurrochmad A, Wahyuono S. n-Hexane Insoluble Fraction of Plantago lanceolata Exerts Anti-Inflammatory Activity in Mice by Inhibiting Cyclooxygenase-2 and Reducing Chemokines Levels. Sci Pharm 2017; 85:scipharm85010012. [PMID: 28335408 PMCID: PMC5388149 DOI: 10.3390/scipharm85010012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/05/2017] [Accepted: 03/08/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammation is involved in the progression of many disorders, such as tumors, arthritis, gastritis, and atherosclerosis. Thus, the development of new agents targeting inflammation is still challenging. Medicinal plants have been used traditionally to treat various diseases including inflammation. A previous study has indicated that dichloromethane extract of P. lanceolata leaves exerts anti-inflammatory activity in an in vitro model. Here, we examined the in vivo anti-inflammatory activities of a n-hexane insoluble fraction of P. lanceolata leaves dichloromethane extract (HIFPL). We first evaluated its potency to reduce paw edema induced by carrageenan, and the expression of the proinflammatory enzyme, cyclooxygenase (COX)-2, in mice. The efficacy of HIFPL to inhibit COX-2 was also evaluated in an in vitro enzymatic assay. We further studied the effect of HIFPL on leukocytes migration in mice induced by thioglycollate. The level of chemokines facilitating the migration of leukocytes was also measured. We found that HIFPL (40, 80, 160 mg/kg) demonstrated anti-inflammatory activities in mice. The HIFPL reduced the volume of paw edema and COX-2 expression. However, HIFPL acts as an unselective COX-2 inhibitor as it inhibited COX-1 with a slightly higher potency. Interestingly, HIFPL strongly inhibited leukocyte migration by reducing the level of chemokines, Interleukine-8 (IL-8) and Monocyte chemoattractant protein-1 (MCP-1).
Collapse
Affiliation(s)
- Nanang Fakhrudin
- Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
- Center for Natural Antiinfective Research, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
| | - Eny Dwi Astuti
- Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
| | - Rini Sulistyawati
- Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
- Akademi Analis Farmasi Al Islam Yogyakarta, Gedongkiwo, Mantrijeron, Yogyakarta 55142, Indonesia.
| | - Djoko Santosa
- Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
- Center for Natural Antiinfective Research, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
| | - Ratna Susandarini
- Faculty of Biology, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
| | - Arief Nurrochmad
- Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
| | - Subagus Wahyuono
- Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
- Center for Natural Antiinfective Research, Faculty of Pharmacy, Universitas Gadjah Mada, Sekip Utara, Yogyakarta 55281, Indonesia.
| |
Collapse
|
32
|
Zheng GH, Xiong SQ, Chen HY, Mei LJ, Wang T. Association of platelet-activating factor receptor gene rs5938 (G/T) and rs313152 (T/C) polymorphisms with coronary heart disease and blood stasis syndrome in a Chinese Han population. Chin J Integr Med 2017; 23:893-900. [PMID: 28197937 DOI: 10.1007/s11655-017-2802-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Indexed: 11/27/2022]
Abstract
OBJECTIVE To explore the association of the platelet-activating factor receptor (PAFR) gene rs5938, rs313152 and rs76744145 polymorphisms with coronary heart disease (CHD) and blood stasis syndrome (BSS) of CHD in Chinese Han population. METHODS A total of 570 CHD patients (299 with BSS and 271 with non-BSS) and 317 controls were enrolled. The PAFR gene rs5938, rs313152 and rs76744145 polymorphisms were genotyped using the multiplex SNaPshot technology. The statistical analysis was conducted using a multiple variable logistic regression model. RESULTS Significant differences were detected in the genotypes frequency distributions of the rs5938 (P<0.01), but not the rs313152 (P>0.05), between the controls and CHD patients. Individuals with an rs5938 or rs313152 mutated allele had a low risk for CHD [adjusted odds ratio (aOR)=0.35, 95% confidence interval (CI): 0.23 to 0.56, P<0.01; aOR=0.65, 95% CI: 0.46 to 0.91, P<0.05, respectively]. After the CHD patients were stratified as BSS or non-BSS according to their Chinese medicine patterns, the rs5938 polymorphism mutated alleles had a significant association with a low risk for BSS of CHD (aOR=0.32, 95% CI: 0.18 to 0.57, P<0.01) and non-BSS of CHD (aOR=0.31, 95% CI: 0.17 to 0.55, P<0.01). The rs313152 polymorphism was associated with a low risk for BSS (aOR=0.51, 95% CI: 0.33 to 0.79, P<0.01), but not for non-BSS (aOR=1.22, 95% CI: 0.81 to 1.85, P<0.05). Furthermore, the interaction effect of the rs5938 and rs313152 polymorphisms for BSS of CHD was significantly based on an aOR value associated with the combination of the rs5938 GT genotype with the rs313152 TC genotype of 0.27 (95% CI: 0.1 to 0.7, P<0.01). CONCLUSION The PAFR gene rs5938 or rs313152 polymorphisms might be a potential biomarker for susceptibility to CHD, especially to BSS of CHD in Chinese Han population.
Collapse
Affiliation(s)
- Guo-Hua Zheng
- College of Health Information Technology and Management, Shanghai University of Medicine and Health Sciences, Shanghai, 201318, China.
| | - Shang-Quan Xiong
- Department of Cardiologic Medicine, The Second People's Hospital of Fujian Province, Fuzhou, 350007, China
| | - Hai-Ying Chen
- The Second Clinic of Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Li-Juan Mei
- The Second Clinic of Chinese Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350122, China
| | - Ting Wang
- Department of Cardiologic Medicine, The Second People's Hospital of Fujian Province, Fuzhou, 350007, China
| |
Collapse
|
33
|
Cimmino G, Ciuffreda LP, Ciccarelli G, Calabrò P, Ferraiolo FAV, Rivellino A, De Palma R, Golino P, Rossi F, Cirillo P, Berrino L. Upregulation of TH/IL-17 Pathway-Related Genes in Human Coronary Endothelial Cells Stimulated with Serum of Patients with Acute Coronary Syndromes. Front Cardiovasc Med 2017; 4:1. [PMID: 28224128 PMCID: PMC5293806 DOI: 10.3389/fcvm.2017.00001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/11/2017] [Indexed: 12/14/2022] Open
Abstract
Background Inflammation plays an essential role in the development and complications of atherosclerosis plaques, including acute coronary syndromes (ACS). Indeed, previous reports have shown that within the coronary circulation of ACS patients, several soluble mediators are released. Moreover, it has been demonstrated that endothelial dysfunction might play an important role in atherosclerosis as well as ACS pathophysiology. However, the mechanisms by which these soluble mediators might affect endothelial functions are still largely unknown. We have evaluated whether soluble mediators contained in serum from coronary circulation of ACS patients might promote changes of gene profile in human coronary endothelial cells (HCAECs). Methods HCAECs were stimulated in vitro for 12 h with serum obtained from the coronary sinus (CS) and the aorta (Ao) of ACS patients; stable angina (SA) patients served as controls. Gene expression profiles of stimulated cells were evaluated by microarray and real-time PCR. Results HCAECs stimulated with serum from CS of ACS patients showed a significant change (upregulation and downregulation) in gene expression profile as compared with cells stimulated with serum from CS of SA patients. Moreover, ad hoc sub analysis indicated the upregulation of Th-17/IL-17 pathway-related genes. Conclusion This study demonstrates that, in ACS patients, the chemical mediators released in the coronary circulation might be able to perturb coronary endothelial cells (ECs) modifying their gene profile. These modified ECs, through downregulation of protective gene and, mainly, through upregulation of gene able to modulate the Th-17/IL-17 pathway, might play a key role in progression of coronary atherosclerosis and in developing future acute events.
Collapse
Affiliation(s)
- Giovanni Cimmino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Giovanni Ciccarelli
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Paolo Calabrò
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | | | - Alessia Rivellino
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Raffaele De Palma
- Department of Clinical and Experimental Medicine, Section of Immunology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Paolo Golino
- Department of Cardio-Thoracic and Respiratory Sciences, Section of Cardiology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| | - Plinio Cirillo
- Department of Advanced Biomedical Sciences, Section of Cardiology, University of Naples, "Federico II" , Naples , Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, University of Campania "Luigi Vanvitelli" , Naples , Italy
| |
Collapse
|
34
|
Brophy ML, Dong Y, Wu H, Rahman HNA, Song K, Chen H. Eating the Dead to Keep Atherosclerosis at Bay. Front Cardiovasc Med 2017; 4:2. [PMID: 28194400 PMCID: PMC5277199 DOI: 10.3389/fcvm.2017.00002] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/12/2017] [Indexed: 12/22/2022] Open
Abstract
Atherosclerosis is the primary cause of coronary heart disease (CHD), ischemic stroke, and peripheral arterial disease. Despite effective lipid-lowering therapies and prevention programs, atherosclerosis is still the leading cause of mortality in the United States. Moreover, the prevalence of CHD in developing countries worldwide is rapidly increasing at a rate expected to overtake those of cancer and diabetes. Prominent risk factors include the hardening of arteries and high levels of cholesterol, which lead to the initiation and progression of atherosclerosis. However, cell death and efferocytosis are critical components of both atherosclerotic plaque progression and regression, yet, few currently available therapies focus on these processes. Thus, understanding the causes of cell death within the atherosclerotic plaque, the consequences of cell death, and the mechanisms of apoptotic cell clearance may enable the development of new therapies to treat cardiovascular disease. Here, we review how endoplasmic reticulum stress and cholesterol metabolism lead to cell death and inflammation, how dying cells affect plaque progression, and how autophagy and the clearance of dead cells ameliorates the inflammatory environment of the plaque. In addition, we review current research aimed at alleviating these processes and specifically targeting therapeutics to the site of the plaque.
Collapse
Affiliation(s)
- Megan L Brophy
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital, Boston, MA, USA
| | - Yunzhou Dong
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hao Wu
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - H N Ashiqur Rahman
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Kai Song
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| | - Hong Chen
- Karp Family Research Laboratories, Vascular Biology Program, Harvard Medical School, Boston Children's Hospital , Boston, MA , USA
| |
Collapse
|
35
|
Laskarin G, Persic V, Kukic SR, Massari D, Legovic A, Boban M, Miskulin R, Rogoznica M, Kehler T. Can pain intensity in osteoarthritis joint be indicator of the impairment of endothelial function? Med Hypotheses 2016; 94:15-9. [DOI: 10.1016/j.mehy.2016.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/02/2016] [Indexed: 10/21/2022]
|
36
|
Yuan B, Shi H, Zheng K, Su Z, Su H, Zhong M, He X, Zhou C, Chen H, Xiong Q, Zhang Y, Yang Z. MCP-1-mediated activation of microglia promotes white matter lesions and cognitive deficits by chronic cerebral hypoperfusion in mice. Mol Cell Neurosci 2016; 78:52-58. [PMID: 27530284 DOI: 10.1016/j.mcn.2016.08.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2016] [Revised: 07/31/2016] [Accepted: 08/10/2016] [Indexed: 11/19/2022] Open
Abstract
Microglia activation played a vital role in the pathogenesis of white matter lesions (WMLs) by chronic cerebral hypoperfusion. In addition, hypoxia induced up-regulated expression of MCP-1, promotes the activation of microglia. However, the role of MCP-1-mediated microglia activation in chronic cerebral ischemia is still unknown. To explore that, chronic cerebral hypoperfusion model was established by permanent stenosis of bilateral common carotid artery in mice. The activation of microglia and the related signal pathway p38MAPK/PKC in white matter, and working memory of mice were observed. We found that stenosis of common carotid arteries could induce MCP-1-mediated activation of microglia through p38MAPK/PKC pathway and white matter lesions. Taken together, our findings represent a novel mechanism of MCP-1 involved in activation of microglia and provide a novel therapeutical strategy for chronic cerebral hypoperfusion.
Collapse
Affiliation(s)
- Bangqing Yuan
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China; Department of Neurosurgery, The 476th Hospital of PLA, Fuzhou, Fujian 350025, China
| | - Hui Shi
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Kuang Zheng
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Zulu Su
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Hai Su
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Ming Zhong
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Xuenong He
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Changlong Zhou
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Hao Chen
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Qijiang Xiong
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Yi Zhang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China
| | - Zhao Yang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing 402160, China.
| |
Collapse
|
37
|
Decano JL, Mattson PC, Aikawa M. Macrophages in Vascular Inflammation: Origins and Functions. Curr Atheroscler Rep 2016; 18:34. [PMID: 27125207 DOI: 10.1007/s11883-016-0585-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
38
|
Singla DK, Singla R, Wang J. BMP-7 Treatment Increases M2 Macrophage Differentiation and Reduces Inflammation and Plaque Formation in Apo E-/- Mice. PLoS One 2016; 11:e0147897. [PMID: 26824441 PMCID: PMC4732822 DOI: 10.1371/journal.pone.0147897] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 01/11/2016] [Indexed: 12/30/2022] Open
Abstract
Inflammation plays a fundamental role in the inception and development of atherosclerosis (ATH). Mechanisms of inflammation include the infiltration of monocytes into the injured area and subsequent differentiation into either pro-inflammatory M1 macrophages or anti-inflammatory M2 macrophages. We have previously published data suggesting bone morphogenetic protein-7 (BMP-7) enhances M2 macrophage differentiation and anti-inflammatory cytokine secretion in vitro. In this regard, we hypothesized BMP-7 would inhibit plaque formation in an animal model of ATH through monocytic plasticity mediation. ATH was generated in male and female Apo E(-/-) mice via partial left carotid artery (PLCA) ligation and mice were divided into 3 groups: Sham, PLCA, and PLCA+BMP-7 (200 ug/kg; i.v.). Our data suggest that BMP-7 inhibits plaque formation and increases arterial systolic velocity. Furthermore, we report inhibition of monocyte infiltration and a decrease in associated pro-inflammatory cytokines (MCP-1, TNF-α, and IL-6) in the PLCA+BMP-7 mice. In contrast, our data suggest a significant (p<0.05) increase in M2 macrophage populations with consequential enhanced anti-inflammatory cytokine (IL-1RA, IL-10, and Arginase 1) expression following BMP-7 treatment. We have also observed that mechanisms promoting monocyte into M2 macrophage differentiation by BMP-7 involve the upregulation and activation of the BMP-7 receptor (BMP-7RII). In conclusion, we report that BMP-7 has the potential to mediate cellular plasticity and mitigate the inflammatory immune response, which results in decreased plaque formation and improved blood velocity.
Collapse
Affiliation(s)
- Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
- * E-mail:
| | - Reetu Singla
- Division of Metabolic and Cardiovascular Sciences, Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
| | - Jing Wang
- Division of Metabolic and Cardiovascular Sciences, Biomolecular Science Center, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States of America
| |
Collapse
|
39
|
Expression characteristics of neutrophil and mononuclear-phagocyte related genes mRNA in the stable angina pectoris and acute myocardial infarction stages of coronary artery disease. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2015; 12:279-86. [PMID: 26089853 PMCID: PMC4460172 DOI: 10.11909/j.issn.1671-5411.2015.03.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2014] [Revised: 03/20/2015] [Accepted: 04/21/2015] [Indexed: 12/24/2022]
Abstract
Objective To investigate expression differences of neutrophil and mononuclear phagocyte related gene mRNAs among acute myocardial infarction (AMI), stable angina (SA) and control groups, and then discuss their expression characteristics in the stable angina pectoris (SAP) and AMI stages of coronary artery disease (CAD). Methods Whole Human Genome Oligo Microarrays were applied to assess the differential expression characteristics of neutrophil and mononuclear phagocyte related mRNAs in patients with AMI (n = 20), SA (n = 20) and controls (n = 20). Results (1) Almost all colony-stimulating factors (CSF) and their receptors related mRNAs was up-regulated in AMI and SA groups compared with the control group, and the expression of granulocyte-macrophage colony stimulating factor receptor (GM-CSFR) and granulocyte colony stimulating factor receptor (G-CSFR) mRNAs in the AMI group was significantly up-regulated compared with the other two groups (P < 0.01). (2) The expression of mRNAs related to monocyte chemoattractant protein-1 (MCP-1), CCR2 (MCP-1 receptor) and CXCR2 (IL-8 receptor) was significantly up-regulated (P < 0.01) in AMI group compared with SA and control groups. IL-8 mRNA expression in the AMI group was clearly higher than the controls (P < 0.05). (3) All mRNAs expression related to opsonic receptors (IgG FcR and C3bR/C4bR) was significantly up-regulated in AMI group compared with SA and control group (P < 0.01), and the SA group showed an upward trend compared with controls. (4) Most pattern recognition receptor (PRR)-related mRNAs expression was up-regulated in AMI group compared with SA and control groups. Most toll-like receptor (TLR) mRNAs expression was significantly up-regulated (P < 0.01) than the SA and control groups; macrophage scavenger receptor (MSR) mRNA was significantly up-regulated in AMI group compared with the control group (P < 0.01), and the SA group showed an upward trend compared with the controls. Conclusions The expression of most neutrophil and mononuclear-macrophage function related genes mRNAs was significantly up-regulated by stages during the progression of CAD, suggesting that the adhesive, chemotactic and phagocytic functions of neutrophil and mononuclear-macrophage were strengthened in the occurrence and development of coronary atherosclerosis and AMI. This also showed a stepped upward trend as the disease progressed.
Collapse
|
40
|
Hernández D, Triñanes J, Salido E, Pitti S, Rufino M, González-Posada JM, Torres A. Artery Wall Assessment Helps Predict Kidney Transplant Outcome. PLoS One 2015; 10:e0129083. [PMID: 26066045 PMCID: PMC4466324 DOI: 10.1371/journal.pone.0129083] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 05/05/2015] [Indexed: 12/13/2022] Open
Abstract
Background Kidney transplant recipients have high cardiovascular risk, and vascular inflammation may play an important role. We explored whether the inflammatory state in the vessel wall was related to carotid intima-media thickness (c-IMT) and patient survival following kidney transplantation. Methods In this prospective observational cohort study we measured c-IMT and expression of proinflammatory cytokines and adhesion molecules in the inferior epigastric artery in 115 kidney transplant candidates. Another c-IMT measurement was done 1-year post-transplantation in 107. By stepwise multiple regression analysis we explored factors associated with baseline c-IMT and their changes over time. Multivariate Cox regression analysis was constructed to identify risk factors for mortality. Results A worse cardiovascular profile (older age, smoker, diabetic, carotid plaque, systolic blood pressure and vascular calcification) and higher VCAM-1 levels were found in patients in the highest baseline c-IMT tertile, who also had a worse survival. Factors independently related to baseline c-IMT were age (β=0.369, P<0.0001), fasting glucose (β=0.168, P=0.045), smoking (β=0.228, P=0.003) and VCAM-1 levels (β=0.244, P=0.002). Independent factors associated with c-IMT measurement 1-year post-transplantation were baseline c-IMT (β=-0.677, P<0.0001), post-transplant diabetes (β=0.225, P=0.003) and triglycerides (β=0.302, P=0.023). Vascular VCAM-1 levels were associated with increased risk of mortality in bivariate and multivariate Cox regression. Notably, nearly 50% of patients showed an increase or maintenance of high c-IMT 1 year post-transplantation and these patients experienced a higher mortality (13 versus 3.5%; P=0.021). Conclusion A worse cardiovascular profile and a higher vascular VCAM-1 protein levels at time of KT are related to subclinical atheromatosis. This could lead to a higher post-transplant mortality. Pre-transplant c IMT, post-transplant diabetes and triglycerides at 1-year post-transplantation may condition a high c-IMT measurement post-transplantation, which may decrease patient survival.
Collapse
Affiliation(s)
- Domingo Hernández
- Nephrology Department, Carlos Haya Regional University Hospital and University of Málaga (IBIMA), REDinREN, Málaga, Spain
- * E-mail:
| | - Javier Triñanes
- Research Unit, Hospital Universitario de Canarias, Tenerife, Spain
| | - Eduardo Salido
- Research Unit, Hospital Universitario de Canarias, Tenerife, Spain
| | - Sergio Pitti
- Radiology Department, Hospital Universitario de Canarias, Tenerife, Spain
| | - Margarita Rufino
- Nephrology Department, Hospital Universitario de Canarias, CIBICAN, University of La Laguna, Instituto Reina Sofía de Investigación Renal (IRSIN), Tenerife, Spain
| | - José Manuel González-Posada
- Nephrology Department, Hospital Universitario de Canarias, CIBICAN, University of La Laguna, Instituto Reina Sofía de Investigación Renal (IRSIN), Tenerife, Spain
| | - Armando Torres
- Nephrology Department, Hospital Universitario de Canarias, CIBICAN, University of La Laguna, Instituto Reina Sofía de Investigación Renal (IRSIN), Tenerife, Spain
| |
Collapse
|
41
|
Plasma levels of cytokines and chemokines and the risk of mortality in HIV-infected individuals: a case-control analysis nested in a large clinical trial. AIDS 2015; 29:847-51. [PMID: 25695873 DOI: 10.1097/qad.0000000000000618] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND All-cause mortality and serious non-AIDS events (SNAEs) in individuals with HIV-1 infection receiving antiretroviral therapy are associated with increased production of interleukin-6 which appears to be driven by monocyte/macrophage activation. Plasma levels of other cytokines or chemokines associated with immune activation might also be biomarkers of an increased risk of mortality and/or SNAEs. METHODS Baseline plasma samples from 142 participants enrolled into the Strategies for Management of Antiretroviral Therapy study, who subsequently died, and 284 matched controls, were assayed for levels of 15 cytokines and chemokines. Cytokine and chemokine levels were analysed individually and when grouped according to function (innate/proinflammatory response, cell trafficking and cell activation/proliferation) for their association with the risk of subsequent death. RESULTS Higher plasma levels of proinflammatory cytokines (interleukin-6 and tumour necrosis factor-α) were associated with an increased risk of all-cause mortality but in analyses adjusted for potential confounders, only the association with interleukin-6 persisted. Increased plasma levels of the chemokine CXCL8 were also associated with all-cause mortality independently of hepatitis C virus status but not when analyses were adjusted for all confounders. In contrast, higher plasma levels of cytokines mediating cell activation/proliferation were not associated with a higher mortality risk and exhibited a weak protective effect when analysed as a group. CONCLUSION Whereas plasma levels of interleukin-6 are the most informative biomarker of cytokine dysregulation associated with all-cause mortality in individuals with HIV-1 infection, assessment of plasma levels of CXCL8 might provide information about causes of mortality and possibly SNAEs.
Collapse
|
42
|
Sock-Jin L, Kumolosasi E, Azmi N, Bukhari SNA, Jasamai M, Fauzi NM. Effects of synthetic chalcone derivatives on oxidised palmitoyl arachidonoyl phosphorylcholine-induced proinflammatory chemokines production. RSC Adv 2015. [DOI: 10.1039/c5ra11073d] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Oxidised 1-palmitoyl-2-arachidonoyl-sn-glycero-3-phosphorylcholine (OxPAPC) induces the production of proinflammatory chemokines has been widely studied for its role in vascular inflammation.
Collapse
Affiliation(s)
- Lim Sock-Jin
- Drug and Herbal Research Centre
- Faculty of Pharmacy
- Universiti Kebangsaan Malaysia
- 50300 Kuala Lumpur
- Malaysia
| | - Endang Kumolosasi
- Drug and Herbal Research Centre
- Faculty of Pharmacy
- Universiti Kebangsaan Malaysia
- 50300 Kuala Lumpur
- Malaysia
| | - Norazrina Azmi
- Drug and Herbal Research Centre
- Faculty of Pharmacy
- Universiti Kebangsaan Malaysia
- 50300 Kuala Lumpur
- Malaysia
| | - Syed Nasir Abbas Bukhari
- Drug and Herbal Research Centre
- Faculty of Pharmacy
- Universiti Kebangsaan Malaysia
- 50300 Kuala Lumpur
- Malaysia
| | - Malina Jasamai
- Drug and Herbal Research Centre
- Faculty of Pharmacy
- Universiti Kebangsaan Malaysia
- 50300 Kuala Lumpur
- Malaysia
| | - Norsyahida Mohd Fauzi
- Drug and Herbal Research Centre
- Faculty of Pharmacy
- Universiti Kebangsaan Malaysia
- 50300 Kuala Lumpur
- Malaysia
| |
Collapse
|
43
|
Petrov V, Funderburg N, Weinberg A, Sieg S. Human β defensin-3 induces chemokines from monocytes and macrophages: diminished activity in cells from HIV-infected persons. Immunology 2014; 140:413-20. [PMID: 23829433 DOI: 10.1111/imm.12148] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 06/06/2013] [Accepted: 07/02/2013] [Indexed: 12/17/2022] Open
Abstract
Human β defensin-3 (hBD-3) is an antimicrobial peptide with diverse functionality. We investigated the capacity of hBD-3 and, for comparison, Pam3CSK4 and LL-37 to induce co-stimulatory molecules and chemokine expression in monocytes. These stimuli differentially induced CD80 and CD86 on the surface of monocytes and each stimulant induced a variety of chemokines including monocyte chemoattractant protein 1 (MCP-1), Gro-α, macrophage-derived chemokine (MDC) and macrophage inflammatory protein 1β (MIP1β), while only hBD-3 and Pam3CSK4 significantly induced the angiogenesis factor, vascular endothelial growth factor (VEGF). Human BD-3 induced similar chemokines in monocyte-derived macrophages and additionally induced expression of Regulated upon activation normal T-cell expressed and presumably secreted (RANTES) in these cells. Comparison of monocytes from HIV(+) and HIV(-) donors indicated that monocytes from HIV(+) donors were more likely to spontaneously express certain chemokines (MIP-1α, MIP-1β and MCP-1) and less able to increase expression of other molecules in response to hBD-3 (MDC, Gro-α and VEGF). Chemokine receptor expression (CCR5, CCR2 and CXCR2) was relatively normal in monocytes from HIV(+) donors compared with cells from HIV(-) donors with the exception of diminished expression of the receptor for MDC, CCR4, which was reduced in the patrolling monocyte subset (CD14(+) CD16(++) ) of HIV(+) donors. These observations implicate chemokine induction by hBD-3 as a potentially important mechanism for orchestrating cell migration into inflamed tissues. Alterations in chemokine production or their receptors in monocytes of HIV-infected persons could influence cell migration and modify the effects of hBD-3 at sites of inflammation.
Collapse
Affiliation(s)
- Velizar Petrov
- Department of Biological Sciences, Case School of Dental Medicine, Case Western Reserve University, Cleveland, OH, USA
| | | | | | | |
Collapse
|
44
|
Sprague L, Muccioli M, Pate M, Singh M, Xiong C, Ostermann A, Niese B, Li Y, Li Y, Courreges MC, Benencia F. Dendritic cells: In vitro culture in two- and three-dimensional collagen systems and expression of collagen receptors in tumors and atherosclerotic microenvironments. Exp Cell Res 2014; 323:7-27. [PMID: 24569142 DOI: 10.1016/j.yexcr.2014.01.031] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 01/25/2014] [Accepted: 01/28/2014] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DCs) are immune cells found in the peripheral tissues where they sample the organism for infections or malignancies. There they take up antigens and migrate towards immunological organs to contact and activate T lymphocytes that specifically recognize the antigen presented by these antigen presenting cells. In the steady state there are several types of resident DCs present in various different organs. For example, in the mouse, splenic DC populations characterized by the co-expression of CD11c and CD8 surface markers are specialized in cross-presentation to CD8 T cells, while CD11c/SIRP-1α DCs seem to be dedicated to activating CD4 T cells. On the other hand, DCs have also been associated with the development of various diseases such as cancer, atherosclerosis, or inflammatory conditions. In such disease, DCs can participate by inducing angiogenesis or immunosuppression (tumors), promoting autoimmune responses, or exacerbating inflammation (atherosclerosis). This change in DC biology can be prompted by signals in the microenvironment. We have previously shown that the interaction of DCs with various extracellular matrix components modifies the immune properties and angiogenic potential of these cells. Building on those studies, herewith we analyzed the angiogenic profile of murine myeloid DCs upon interaction with 2D and 3D type-I collagen environments. As determined by PCR array technology and quantitative PCR analysis we observed that interaction with these collagen environments induced the expression of particular angiogenic molecules. In addition, DCs cultured on collagen environments specifically upregulated the expression of CXCL-1 and -2 chemokines. We were also able to establish DC cultures on type-IV collagen environments, a collagen type expressed in pathological conditions such as atherosclerosis. When we examined DC populations in atherosclerotic veins of Apolipoprotein E deficient mice we observed that they expressed adhesion molecules capable of interacting with collagen. Finally, to further investigate the interaction of DCs with collagen in other pathological conditions, we determined that both murine ovarian and breast cancer cells express several collagen molecules that can contribute to shape their particular tumor microenvironment. Consistently, tumor-associated DCs were shown to express adhesion molecules capable of interacting with collagen molecules as determined by flow cytometry analysis. Of particular relevance, tumor-associated DCs expressed high levels of CD305/LAIR-1, an immunosuppressive receptor. This suggests that signaling through this molecule upon interaction with collagen produced by tumor cells might help define the poorly immunogenic status of these cells in the tumor microenvironment. Overall, these studies demonstrate that through interaction with collagen proteins, DCs can be capable of modifying the microenvironments of inflammatory disease such as cancer or atherosclerosis.
Collapse
Affiliation(s)
- Leslee Sprague
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, USA
| | - Maria Muccioli
- Molecular and Cellular Biology Program, Ohio University, USA
| | - Michelle Pate
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, USA
| | - Manindra Singh
- Molecular and Cellular Biology Program, Ohio University, USA
| | - Chengkai Xiong
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, USA
| | - Alexander Ostermann
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, USA
| | - Brandon Niese
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, USA
| | - Yihan Li
- Molecular and Cellular Biology Program, Ohio University, USA
| | - Yandi Li
- Molecular and Cellular Biology Program, Ohio University, USA
| | - Maria Cecilia Courreges
- Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, USA
| | - Fabian Benencia
- Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, USA; Molecular and Cellular Biology Program, Ohio University, USA; Diabetes Institute, Ohio University, USA; Department of Biomedical Sciences, Heritage College of Osteopathic Medicine, Ohio University, USA.
| |
Collapse
|
45
|
Mirabella L, Grasselli G, Haitsma JJ, Zhang H, Slutsky AS, Sinderby C, Beck J. Lung protection during non-invasive synchronized assist versus volume control in rabbits. Crit Care 2014; 18:R22. [PMID: 24456613 PMCID: PMC4057206 DOI: 10.1186/cc13706] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2013] [Accepted: 01/14/2014] [Indexed: 11/10/2022] Open
Abstract
Introduction Experimental work provides insight into potential lung protective strategies. The objective of this study was to evaluate markers of ventilator-induced lung injury after two different ventilation approaches: (1) a “conventional” lung-protective strategy (volume control (VC) with low tidal volume, positive end-expiratory pressure (PEEP) and paralysis), (2) a physiological approach with spontaneous breathing, permitting synchrony, variability and a liberated airway. For this, we used non-invasive Neurally Adjusted Ventilatory Assist (NIV-NAVA), with the hypothesis that liberation of upper airways and the ventilator’s integration with lung protective reflexes would be equally lung protective. Methods In this controlled and randomized in vivo laboratory study, 25 adult White New Zealand rabbits were studied, including five non-ventilated control animals. The twenty animals with aspiration-induced lung injury were randomized to ventilation with either VC (6 mL/kg, PEEP 5 cm H2O, and paralysis) or NIV-NAVA for six hours (PEEP = zero because of leaks). Markers of lung function, lung injury, vital signs and ventilator parameters were assessed. Results At the end of six hours of ventilation (n = 20), there were no significant differences between VC and NIV-NAVA for vital signs, PaO2/FiO2 ratio, lung wet-to-dry ratio and broncho-alveolar Interleukin 8 (Il-8). Plasma IL-8 was higher in VC (P <0.05). Lung injury score was lower for NIV-NAVA (P = 0.03). Dynamic lung compliance recovered after six hours in NIV-NAVA but not in VC (P <0.05). During VC, peak pressures increased from 9.2 ± 2.4 cm H2O (hour 1) to 12.3 ± 12.3 cm H2O (hour 6) (P <0.05). During NIV-NAVA, the tracheal end-expiratory pressure was similar to the end-expiratory pressure during VC. Two animals regurgitated during NIV-NAVA, without clinical consequences, and survived the protocol. Conclusions In experimental acute lung injury, NIV-NAVA is as lung-protective as VC 6 ml/kg with PEEP. Electronic supplementary material The online version of this article (doi:10.1186/cc13706) contains supplementary material, which is available to authorized users.
Collapse
|
46
|
Immunological aspects of atherosclerosis. Semin Immunopathol 2013; 36:73-91. [DOI: 10.1007/s00281-013-0402-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 10/15/2013] [Indexed: 12/21/2022]
|
47
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
48
|
Yu H, Huang X, Ma Y, Gao M, Wang O, Gao T, Shen Y, Liu X. Interleukin-8 regulates endothelial permeability by down-regulation of tight junction but not dependent on integrins induced focal adhesions. Int J Biol Sci 2013; 9:966-79. [PMID: 24155670 PMCID: PMC3805902 DOI: 10.7150/ijbs.6996] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2013] [Accepted: 08/29/2013] [Indexed: 01/02/2023] Open
Abstract
Interleukin-8 (IL-8) is a common inflammatory factor, which involves in various non-specific pathological processes of inflammation. It has been found that increased endothelial permeability accompanied with high expression of IL-8 at site of injured endothelium and atherosclerotic plaque at early stages, suggesting that IL-8 participated in regulating endothelial permeability in the developing processes of vascular disease. The purpose of this study is to investigate the regulation effects of IL-8 on the vascular endothelial permeability, and the mRNA and protein expression of tight junction components (i.e., ZO-1, Claudin-5 and Occludin). Endothelial cells were stimulated by IL-8 with the dose of 50, 100 and 200 ng/mL, and duration of 2, 4, 6, 8h, respectively. The mRNA and protein expression level of tight junction components with IL-8 under different concentration and duration was examined by RT-PCR and Western blot, respectively. Meanwhile, the integrins induced focal adhesions event with IL-8 stimulation was also investigated. The results showed that IL-8 regulated the permeability of endothelium by down-regulation of tight junction in a dose- and time-dependence manner, but was not by integrins induced focal adhesions. This finding reveals the molecular mechanism in the increase of endothelial cell permeability induced by IL-8, which is expected to provide a new idea as a therapeutic target in vascular diseases.
Collapse
Affiliation(s)
- Hongchi Yu
- Institute of Biomedical Engineering, School of Preclinical and Forensic Medicine, Sichuan University, Chengdu 610041, China
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Wang ZH, Liu XL, Zhong M, Zhang LP, Shang YY, Hu XY, Li L, Zhang Y, Deng JT, Zhang W. Pleiotropic Effects of Atorvastatin on Monocytes in Atherosclerotic Patients. J Clin Pharmacol 2013; 50:311-9. [DOI: 10.1177/0091270009340889] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
50
|
Regulation of atherogenesis by chemokines and chemokine receptors. Arch Immunol Ther Exp (Warsz) 2012; 61:1-14. [PMID: 23224338 DOI: 10.1007/s00005-012-0202-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Accepted: 11/18/2012] [Indexed: 12/24/2022]
Abstract
Atherosclerosis is a chronic inflammatory and metabolic disorder affecting large- and medium-sized arteries, and the leading cause of mortality worldwide. The pathogenesis of atherosclerosis involves accumulation of lipids and leukocytes in the intima of blood vessel walls creating plaque. How leukocytes accumulate in plaque remains poorly understood; however, chemokines acting at specific G protein-coupled receptors appear to be important. Studies using knockout mice suggest that chemokine receptor signaling may either promote or inhibit atherogenesis, depending on the receptor. These proof of concept studies have spurred efforts to develop drugs targeting the chemokine system in atherosclerosis, and several have shown beneficial effects in animal models. This study will review key discoveries in basic and translational research in this area.
Collapse
|