1
|
Bellini I, Scribano D, Ambrosi C, Chiovoloni C, Rondón S, Pronio A, Palamara AT, Pietrantoni A, Kashkanova A, Sandoghdar V, D'Amelio S, Cavallero S. Anisakis extracellular vesicles elicit immunomodulatory and potentially tumorigenic outcomes on human intestinal organoids. Parasit Vectors 2024; 17:393. [PMID: 39285481 PMCID: PMC11406850 DOI: 10.1186/s13071-024-06471-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND Anisakis spp. are zoonotic nematodes causing mild to severe acute and chronic gastrointestinal infections. Chronic anisakiasis can lead to erosive mucosal ulcers, granulomas and inflammation, potential tumorigenic triggers. How Anisakis exerts its pathogenic potential through extracellular vesicles (EVs) and whether third-stage infective larvae may favor a tumorigenic microenvironment remain unclear. METHODS Here, we investigated the parasite's tumorigenic and immunomodulatory capabilities using comparative transcriptomics, qRT-PCR and protein analysis with multiplex ELISA on human intestinal organoids exposed to Anisakis EVs. Moreover, EVs were characterized in terms of shape, size and concentration using classic TEM, SEM and NTA analyses and advanced interferometric NTA. RESULTS Anisakis EVs showed classic shape features and a median average diameter of around 100 nm, according to NTA and iNTA. Moreover, a refractive index of 5-20% of non-water content suggested their effective biological cargo. After treatment of human intestinal organoids with Anisakis EVs, an overall parasitic strategy based on mitigation of the immune and inflammatory response was observed. Anisakis EVs impacted gene expression of main cytokines, cell cycle regulation and protein products. Seven key genes related to cell cycle regulation and apoptosis were differentially expressed in organoids exposed to EVs. In particular, the downregulation of EPHB2 and LEFTY1 and upregulation of NUPR1 genes known to be associated with colorectal cancer were observed, suggesting their involvement in tumorigenic microenvironment. A statistically significant reduction in specific mediators of inflammation and cell-cycle regulation from the polarized epithelium as IL-33R, CD40 and CEACAM1 from the apical chambers and IL-1B, GM-CSF, IL-15 and IL-23 from both chambers were observed. CONCLUSIONS The results here obtained unravel intestinal epithelium response to Anisakis EVs, impacting host's anthelminthic strategies and revealing for the first time to our knowledge the host-parasite interactions in the niche environment of an emerging accidental zoonosis. Use of an innovative EV characterization approach may also be useful for study of other helminth EVs, since the knowledge in this field is very limited.
Collapse
Affiliation(s)
- Ilaria Bellini
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Daniela Scribano
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Cecilia Ambrosi
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Open University, IRCCS, Rome, Italy
- Laboratory of Microbiology of Chronic-Neurodegenerative Diseases, San Raffaele Open University, IRCCS, Rome, Italy
| | - Claudia Chiovoloni
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Silvia Rondón
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Annamaria Pronio
- Digestive Endoscopy Unit, Department of General Surgery and Surgical Specialties "Paride Stefanini", Sapienza University of Rome, Azienda Policlinico Umberto I, Rome, Italy
| | - Anna Teresa Palamara
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | | | - Anna Kashkanova
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Vahid Sandoghdar
- Max Planck Institute for the Science of Light, Erlangen, Germany
| | - Stefano D'Amelio
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Serena Cavallero
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Laboratory Affiliated to Pasteur Institute, Fondazione Cenci Bolognetti, Piazzale Aldo Moro 5, 00185, Rome, Italy.
| |
Collapse
|
2
|
Duranova H, Kuzelova L, Borotova P, Simora V, Fialkova V. Human Umbilical Vein Endothelial Cells as a Versatile Cellular Model System in Diverse Experimental Paradigms: An Ultrastructural Perspective. MICROSCOPY AND MICROANALYSIS : THE OFFICIAL JOURNAL OF MICROSCOPY SOCIETY OF AMERICA, MICROBEAM ANALYSIS SOCIETY, MICROSCOPICAL SOCIETY OF CANADA 2024; 30:419-439. [PMID: 38817111 DOI: 10.1093/mam/ozae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/15/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024]
Abstract
Human umbilical vein endothelial cells (HUVECs) are primary cells isolated from the vein of an umbilical cord, extensively used in cardiovascular studies and medical research. These cells, retaining the characteristics of endothelial cells in vivo, serve as a valuable cellular model system for understanding vascular biology, endothelial dysfunction, pathophysiology of diseases such as atherosclerosis, and responses to different drugs or treatments. Transmission electron microscopy (TEM) has been a cornerstone in revealing the detailed architecture of multiple cellular model systems including HUVECs, allowing researchers to visualize subcellular organelles, membrane structures, and cytoskeletal elements. Among them, the endoplasmic reticulum, Golgi apparatus, mitochondria, and nucleus can be meticulously examined to recognize alterations indicative of cellular responses to various stimuli. Importantly, Weibel-Palade bodies are characteristic secretory organelles found in HUVECs, which can be easily distinguished in the TEM. These distinctive structures also dynamically react to different factors through regulated exocytosis, resulting in complete or selective release of their contents. This detailed review summarizes the ultrastructural features of HUVECs and highlights the utility of TEM as a pivotal tool for analyzing HUVECs in diverse research frameworks, contributing valuable insights into the comprehension of HUVEC behavior and enriching our knowledge into the complexity of vascular biology.
Collapse
Affiliation(s)
- Hana Duranova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Lenka Kuzelova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
- Faculty of Biotechnology and Food Sciences, Institute of Biotechnology, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Petra Borotova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Veronika Simora
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| | - Veronika Fialkova
- AgroBioTech Research Centre, Slovak University of Agriculture, Tr. A. Hlinku 2, 949 76 Nitra, Slovak Republic
| |
Collapse
|
3
|
Posadas-Sánchez R, Velázquez-Sánchez F, Reyes-Barrera J, Cardoso-Saldaña G, Velázquez-Argueta F, Antonio-Villa NE, Fragoso JM, Vargas-Alarcón G. MCP-1 rs1024611 Polymorphism, MCP-1 Concentrations, and Premature Coronary Artery Disease: Results of the Genetics of Atherosclerotic Disease (GEA) Mexican Study. Biomedicines 2024; 12:1292. [PMID: 38927499 PMCID: PMC11201762 DOI: 10.3390/biomedicines12061292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 05/24/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
Monocyte chemoattractant protein-1 (MCP-1) participates in the initiation and progression of atherosclerosis. In vitro studies have reported that the MCP-1 rs1024611 polymorphism is associated with increased MCP-1 concentrations. The study aimed to define whether MCP-1 concentrations are associated with premature coronary artery disease (pCAD) and to establish whether variations in the rs1024611 polymorphism increase MCP-1 concentrations. MCP-1 rs1024611 polymorphism was determined in 972 pCAD patients and 1070 control individuals by real-time PCR. MCP-1 concentrations were determined by the Bio-Plex system. In the total population, men had higher MCP-1 concentrations when compared to women (p < 0.001). When stratified by rs1024611 genotypes, higher MCP-1 concentrations were observed in AA individuals compared to GG subjects (p = 0.023). When performing the analysis considering sex, the differences remained significant in women (AA vs. GG, p = 0.028 and GA vs. GG, p = 0.008). MCP-1 concentrations were similar in pCAD patients and controls (p = 0.782). However, the independent analysis of the studied groups showed that in patients with the AA genotype, MCP-1 concentrations were significantly higher when compared to patients with the GG genotype (p = 0.009). Considering that the AA genotype increases MCP-1 concentration, we evaluated whether, in AA genotype carriers, MCP-1 concentrations were associated with pCAD. The results showed that for every ten pg/mL increase in MCP-1 concentration, the risk of presenting pCAD increases by 2.7% in AA genotype individuals. Individuals with the MCP-1 rs1024611 AA genotype present an increase in MCP-1 concentration. In those individuals, increased MCP-1 concentrations increase the risk of presented pCAD.
Collapse
Affiliation(s)
- Rosalinda Posadas-Sánchez
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (R.P.-S.); (F.V.-S.); (J.R.-B.); (G.C.-S.); (F.V.-A.); (N.E.A.-V.)
| | - Fernando Velázquez-Sánchez
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (R.P.-S.); (F.V.-S.); (J.R.-B.); (G.C.-S.); (F.V.-A.); (N.E.A.-V.)
| | - Juan Reyes-Barrera
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (R.P.-S.); (F.V.-S.); (J.R.-B.); (G.C.-S.); (F.V.-A.); (N.E.A.-V.)
| | - Guillermo Cardoso-Saldaña
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (R.P.-S.); (F.V.-S.); (J.R.-B.); (G.C.-S.); (F.V.-A.); (N.E.A.-V.)
| | - Frida Velázquez-Argueta
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (R.P.-S.); (F.V.-S.); (J.R.-B.); (G.C.-S.); (F.V.-A.); (N.E.A.-V.)
| | - Neftali Eduardo Antonio-Villa
- Department of Endocrinology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico; (R.P.-S.); (F.V.-S.); (J.R.-B.); (G.C.-S.); (F.V.-A.); (N.E.A.-V.)
| | - José Manuel Fragoso
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
| | - Gilberto Vargas-Alarcón
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico;
- Research Direction, Instituto Nacional de Cardiología Ignacio Chávez, Mexico City 14080, Mexico
| |
Collapse
|
4
|
Forbes S, Bettenay S, Meertens NM, Wildermuth BE, Wildermuth K, Mueller RS. Diascopy and histopathological evaluation of nonblanching erythematous dermatoses in dogs. Vet Dermatol 2024; 35:255-262. [PMID: 38111018 DOI: 10.1111/vde.13230] [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: 02/12/2023] [Revised: 09/19/2023] [Accepted: 12/06/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND Diascopy is a point-of-care diagnostic test used to differentiate skin erythema due to vascular dilation from haemorrhage. In the veterinary literature, only a handful of diseases have been described to be associated with a negative (nonblanching) diascopy result, and histological investigation of haemorrhage has been inconsistent. OBJECTIVES Retrospective study to undertake a histopathological investigation of canine, nonblanching erythematous dermatoses for the presence or absence of haemorrhage and vascular changes. MATERIALS AND METHODS Skin biopsies from dogs presented with moderate-to-severe nonblanching erythema were evaluated histologically. Additionally, clinical data about each patient were analysed. RESULTS Twenty cases were identified with nonblanching erythema. Diagnoses included vasculopathy (n = 6), canine eosinophilic dermatitis (n = 3), cutaneous epitheliotropic T-cell lymphoma (n = 2), and one case each of sterile granuloma and pyogranuloma syndrome, German shepherd dog pyoderma, multiple mast cell tumours, haemangiosarcoma, exfoliative cutaneous lupus erythematosus, canine leishmaniosis with sebaceous adenitis, sebaceous adenitis with concurrent dermatophytosis, calcinosis cutis and canine atopic dermatitis with insect-bite reaction. One or more vascular changes were present in all 20 cases and included perivascular oedema, endothelial swelling and neutrophilic infiltration of vessel walls. Haemorrhage was identified in 17 of 20 cases (85%). Three cases without dermal haemorrhage were calcinosis cutis, sebaceous adenitis with dermatophytosis and canine atopic dermatitis with insect-bite reaction. CONCLUSIONS AND CLINICAL RELEVANCE Negative diascopy was associated with haemorrhage and vascular pathological findings in the majority of cases, yet not all. Haemorrhage was identified histologically in all diseases previously reported as nonblanching as well as in a few additional diseases.
Collapse
Affiliation(s)
| | - Sonya Bettenay
- Centre for Clinical Veterinary Medicine, University of Munich, Munich, Germany
| | | | | | | | - Ralf S Mueller
- Centre for Clinical Veterinary Medicine, University of Munich, Munich, Germany
| |
Collapse
|
5
|
Rengarajan A, Goldblatt HE, Beebe DJ, Virumbrales-Muñoz M, Boeldt DS. Immune cells and inflammatory mediators cause endothelial dysfunction in a vascular microphysiological system. LAB ON A CHIP 2024; 24:1808-1820. [PMID: 38363157 PMCID: PMC11022267 DOI: 10.1039/d3lc00824j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Functional assessment of endothelium serves as an important indicator of vascular health and is compromised in vascular disorders including hypertension, atherosclerosis, and preeclampsia. Endothelial dysfunction in these cases is linked to dysregulation of the immune system involving both changes to immune cells and increased secretion of inflammatory cytokines. Herein, we utilize a well-established microfluidic device to generate a 3-dimensional vascular microphysiological system (MPS) consisting of a tubular blood vessel lined with human umbilical vein endothelial cells (HUVECs) to evaluate endothelial function measured via endothelial permeability and Ca2+ signaling. We evaluated the effect of a mixture of factors associated with inflammation and cardiovascular disease (TNFα, VEGF-A, IL-6 at 10 ng ml-1 each) on vascular MPS and inferred that inflammatory mediators contribute to endothelial dysfunction by disrupting the endothelial barrier over a 48 hour treatment and by diminishing coordinated Ca2+ activity over a 1 hour treatment. We also evaluated the effect of peripheral blood mononuclear cells (PBMCs) on endothelial permeability and Ca2+ signaling in the HUVEC MPS. HUVECs were co-cultured with PBMCs either directly wherein PBMCs passed through the lumen or indirectly with PBMCs embedded in the supporting collagen hydrogel. We revealed that phytohemagglutinin (PHA)-M activated PBMCs cause endothelial dysfunction in MPS both through increased permeability and decreased coordinated Ca2+ activity compared to non-activated PBMCs. Our MPS has potential applications in modeling cardiovascular disorders and screening for potential treatments using measures of endothelial function.
Collapse
Affiliation(s)
- Aishwarya Rengarajan
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, USA.
- Perinatal Research Laboratories, UnityPoint Health-Meriter Hospital, 202 South Park St. 7E, Madison, WI, 53715, USA
| | - Hannah E Goldblatt
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, USA.
- Perinatal Research Laboratories, UnityPoint Health-Meriter Hospital, 202 South Park St. 7E, Madison, WI, 53715, USA
| | - David J Beebe
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
- University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53705, USA
- Department of Biomedical Engineering, Wisconsin Institutes for Medical Research, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
| | - María Virumbrales-Muñoz
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, USA.
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, WI, 53705, USA
- University of Wisconsin Carbone Cancer Center, Wisconsin Institutes for Medical Research, 1111 Highland Ave, Madison, WI, 53705, USA
| | - Derek S Boeldt
- Department of Obstetrics & Gynecology, University of Wisconsin-Madison, School of Medicine and Public Health, USA.
- Perinatal Research Laboratories, UnityPoint Health-Meriter Hospital, 202 South Park St. 7E, Madison, WI, 53715, USA
| |
Collapse
|
6
|
Bréchot N, Rutault A, Marangon I, Germain S. Blood endothelium transition and phenotypic plasticity: A key regulator of integrity/permeability in response to ischemia. Semin Cell Dev Biol 2024; 155:16-22. [PMID: 37479554 DOI: 10.1016/j.semcdb.2023.07.004] [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: 04/20/2023] [Revised: 07/05/2023] [Accepted: 07/09/2023] [Indexed: 07/23/2023]
Abstract
In the human body, the 1013 blood endothelial cells (ECs) which cover a surface of 500-700 m2 (Mai et al., 2013) are key players of tissue homeostasis, remodeling and regeneration. Blood vessel ECs play a major role in the regulation of metabolic and gaz exchanges, cell trafficking, blood coagulation, vascular tone, blood flow and fluid extravasation (also referred to as blood vascular permeability). ECs are heterogeneous in various capillary beds and have the exquisite capacity to cope with environmental changes by regulating their gene expression. Ischemia has major detrimental effects on the endothelium and ischemia-induced regulation of vascular integrity is of paramount importance for human health, as small amounts of fluid accumulation in the interstitium may be responsible for major effects on organ functions and patients outcome. In this review, we will here focus on the stimuli and the molecular mechanisms that control blood endothelium maintenance and phenotypic plasticity/transition involved in controlling blood capillary leakage that might open new avenues for therapeutic applications.
Collapse
Affiliation(s)
- Nicolas Bréchot
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France; Intensive Care Medicine Department, Université de Paris Cité, Hôpital européen Georges-Pompidou, AP-HP, AP-HP.CUP, 75015 Paris, France.
| | - Alexandre Rutault
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Iris Marangon
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France
| | - Stéphane Germain
- Center for Interdisciplinary Research in Biology, College de France, Centre national de la recherche scientifique, Institut national de la santé et de la recherche médicale, Université PSL, Paris, France.
| |
Collapse
|
7
|
Zhao L, Ma D, Wang L, Su X, Feng L, Zhu L, Chen Y, Hao Y, Wang X, Feng J. Metabolic changes with the occurrence of atherosclerotic plaques and the effects of statins. Front Immunol 2023; 14:1301051. [PMID: 38143759 PMCID: PMC10739339 DOI: 10.3389/fimmu.2023.1301051] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 11/23/2023] [Indexed: 12/26/2023] Open
Abstract
Atherosclerosis is a common cardiovascular disease caused by the abnormal expression of multiple factors and genes influenced by both environmental and genetic factors. The primary manifestation of atherosclerosis is plaque formation, which occurs when inflammatory cells consume excess lipids, affecting their retention and modification within the arterial intima. This triggers endothelial cell (EC) activation, immune cell infiltration, vascular smooth muscle cell (VSMC) proliferation and migration, foam cell formation, lipid streaks, and fibrous plaque development. These processes can lead to vascular wall sclerosis, lumen stenosis, and thrombosis. Immune cells, ECs, and VSMCs in atherosclerotic plaques undergo significant metabolic changes and inflammatory responses. The interaction of cytokines and chemokines secreted by these cells leads to the onset, progression, and regression of atherosclerosis. The regulation of cell- or cytokine-based immune responses is a novel therapeutic approach for atherosclerosis. Statins are currently the primary pharmacological agents utilised for managing unstable plaques owing to their ability to enhance endothelial function, regulate VSMC proliferation and apoptosis by reducing cholesterol levels, and mitigate the expression and activity of inflammatory cytokines. In this review, we provide an overview of the metabolic changes associated with atherosclerosis, describe the effects of inflammatory responses on atherosclerotic plaques, and discuss the mechanisms through which statins contribute to plaque stabilisation. Additionally, we examine the role of statins in combination with other drugs in the management of atherosclerosis.
Collapse
Affiliation(s)
| | - Di Ma
- Bethune First Hospital, Jilin University, Changchun, China
| | - LiJuan Wang
- Bethune First Hospital, Jilin University, Changchun, China
| | | | | | | | | | | | | | | |
Collapse
|
8
|
George M, Lang M, Gali CC, Babalola JA, Tam-Amersdorfer C, Stracke A, Strobl H, Zimmermann R, Panzenboeck U, Wadsack C. Liver X Receptor Activation Attenuates Oxysterol-Induced Inflammatory Responses in Fetoplacental Endothelial Cells. Cells 2023; 12:cells12081186. [PMID: 37190095 DOI: 10.3390/cells12081186] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Revised: 04/06/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Oxysterols are oxidized cholesterol derivatives whose systemic levels are found elevated in pregnancy disorders such as gestational diabetes mellitus (GDM). Oxysterols act through various cellular receptors and serve as a key metabolic signal, coordinating inflammation. GDM is a condition of low-grade chronic inflammation accompanied by altered inflammatory profiles in the mother, placenta and fetus. Higher levels of two oxysterols, namely 7-ketocholesterol (7-ketoC) and 7β-hydroxycholesterol (7β-OHC), were observed in fetoplacental endothelial cells (fpEC) and cord blood of GDM offspring. In this study, we tested the effects of 7-ketoC and 7β-OHC on inflammation and investigated the underlying mechanisms involved. Primary fpEC in culture treated with 7-ketoC or 7β-OHC, induced the activation of mitogen-activated protein kinase (MAPK) and nuclear factor kappa B (NFκB) signaling, which resulted in the expression of pro-inflammatory cytokines (IL-6, IL-8) and intercellular cell adhesion molecule-1 (ICAM-1). Liver-X receptor (LXR) activation is known to repress inflammation. Treatment with LXR synthetic agonist T0901317 dampened oxysterol-induced inflammatory responses. Probucol, an inhibitor of LXR target gene ATP-binding cassette transporter A-1 (ABCA-1), antagonized the protective effects of T0901317, suggesting a potential involvement of ABCA-1 in LXR-mediated repression of inflammatory signaling in fpEC. TLR-4 inhibitor Tak-242 attenuated pro-inflammatory signaling induced by oxysterols downstream of the TLR-4 inflammatory signaling cascade. Taken together, our findings suggest that 7-ketoC and 7β-OHC contribute to placental inflammation through the activation of TLR-4. Pharmacologic activation of LXR in fpEC decelerates its shift to a pro-inflammatory phenotype in the presence of oxysterols.
Collapse
Affiliation(s)
- Meekha George
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
| | - Magdalena Lang
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | | | | | - Carmen Tam-Amersdorfer
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | - Anika Stracke
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | - Herbert Strobl
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | - Robert Zimmermann
- Institute for Molecular Biosciences, University of Graz, 8010 Graz, Austria
| | - Ute Panzenboeck
- Otto-Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Immunology, Medical University of Graz, 8010 Graz, Austria
| | - Christian Wadsack
- Department of Obstetrics and Gynecology, Medical University of Graz, 8036 Graz, Austria
- BioTech-Med, 8010 Graz, Austria
| |
Collapse
|
9
|
Yadav V, Sharma S, Kumar A, Singh S, Ravichandiran V. Serratiopeptidase Attenuates Lipopolysaccharide-Induced Vascular Inflammation by Inhibiting the Expression of Monocyte Chemoattractant Protein-1. Curr Issues Mol Biol 2023; 45:2201-2212. [PMID: 36975512 PMCID: PMC10047379 DOI: 10.3390/cimb45030142] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 11/29/2022] [Accepted: 02/01/2023] [Indexed: 03/29/2023] Open
Abstract
Lipopolysaccharide (LPS) has potent pro-inflammatory properties and acts on many cell types including vascular endothelial cells. The secretion of the cytokines MCP-1 (CCL2), interleukins, and the elevation of oxidative stress by LPS-activated vascular endothelial cells contribute substantially to the pathogenesis of vascular inflammation. However, the mechanism involving LPS-induced MCP-1, interleukins, and oxidative stress together is not well demonstrated. Serratiopeptidase (SRP) has been widely used for its anti-inflammatory effects. In this research study, our intention is to establish a potential drug candidate for vascular inflammation in cardiovascular disorder conditions. We used BALB/c mice because this is the most successful model of vascular inflammation, suggested and validated by previous research findings. Our present investigation examined the involvement of SRP in vascular inflammation caused by lipopolysaccharides (LPSs) in a BALB/c mice model. We analyzed the inflammation and changes in the aorta by H&E staining. SOD, MDA, and GPx levels were determined as per the instructions of the kit protocols. ELISA was used to measure the levels of interleukins, whereas immunohistochemistry was carried out for the evaluation of MCP-1 expression. SRP treatment significantly suppressed vascular inflammation in BALB/c mice. Mechanistic studies demonstrated that SRP significantly inhibited the LPS-induced production of proinflammatory cytokines such as IL-2, IL-1, IL-6, and TNF-α in aortic tissue. Furthermore, it also inhibited LPS-induced oxidative stress in the aortas of mice, whereas the expression and activity of monocyte chemoattractant protein-1 (MCP-1) decreased after SRP treatment. In conclusion, SRP has the ability to reduce LPS-induced vascular inflammation and damage by modulating MCP-1.
Collapse
Affiliation(s)
- Vikas Yadav
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, Bihar, India
| | - Satyam Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, Bihar, India
| | - Ashutosh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata 700054, West Bengal, India
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Sahibzada Ajit Singh Nagar 160062, Punjab, India
| | - Sanjiv Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, Bihar, India
| | - V Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hajipur 844102, Bihar, India
| |
Collapse
|
10
|
The complex role of inflammation and gliotransmitters in Parkinson's disease. Neurobiol Dis 2023; 176:105940. [PMID: 36470499 PMCID: PMC10372760 DOI: 10.1016/j.nbd.2022.105940] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 11/28/2022] [Accepted: 12/01/2022] [Indexed: 12/09/2022] Open
Abstract
Our understanding of the role of innate and adaptive immune cell function in brain health and how it goes awry during aging and neurodegenerative diseases is still in its infancy. Inflammation and immunological dysfunction are common components of Parkinson's disease (PD), both in terms of motor and non-motor components of PD. In recent decades, the antiquated notion that the central nervous system (CNS) in disease states is an immune-privileged organ, has been debunked. The immune landscape in the CNS influences peripheral systems, and peripheral immunological changes can alter the CNS in health and disease. Identifying immune and inflammatory pathways that compromise neuronal health and survival is critical in designing innovative and effective strategies to limit their untoward effects on neuronal health.
Collapse
|
11
|
Adams JA, Uryash A, Lopez JR. Non-Invasive Pulsatile Shear Stress Modifies Endothelial Activation; A Narrative Review. Biomedicines 2022; 10:biomedicines10123050. [PMID: 36551807 PMCID: PMC9775985 DOI: 10.3390/biomedicines10123050] [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: 10/17/2022] [Revised: 11/18/2022] [Accepted: 11/21/2022] [Indexed: 11/29/2022] Open
Abstract
The monolayer of cells that line both the heart and the entire vasculature is the endothelial cell (EC). These cells respond to external and internal signals, producing a wide array of primary or secondary messengers involved in coagulation, vascular tone, inflammation, and cell-to-cell signaling. Endothelial cell activation is the process by which EC changes from a quiescent cell phenotype, which maintains cellular integrity, antithrombotic, and anti-inflammatory properties, to a phenotype that is prothrombotic, pro-inflammatory, and permeable, in addition to repair and leukocyte trafficking at the site of injury or infection. Pathological activation of EC leads to increased vascular permeability, thrombosis, and an uncontrolled inflammatory response that leads to endothelial dysfunction. This pathological activation can be observed during ischemia reperfusion injury (IRI) and sepsis. Shear stress (SS) and pulsatile shear stress (PSS) are produced by mechanical frictional forces of blood flow and contraction of the heart, respectively, and are well-known mechanical signals that affect EC function, morphology, and gene expression. PSS promotes EC homeostasis and cardiovascular health. The archetype of inducing PSS is exercise (i.e., jogging, which introduces pulsations to the body as a function of the foot striking the pavement), or mechanical devices which induce external pulsations to the body (Enhanced External Pulsation (EECP), Whole-body vibration (WBV), and Whole-body periodic acceleration (WBPA aka pGz)). The purpose of this narrative review is to focus on the aforementioned noninvasive methods to increase PSS, review how each of these modify specific diseases that have been shown to induce endothelial activation and microcirculatory dysfunction (Ischemia reperfusion injury-myocardial infarction and cardiac arrest and resuscitation), sepsis, and lipopolysaccharide-induced sepsis syndrome (LPS)), and review current evidence and insight into how each may modify endothelial activation and how these may be beneficial in the acute and chronic setting of endothelial activation and microvascular dysfunction.
Collapse
Affiliation(s)
- Jose A. Adams
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
- Correspondence:
| | - Arkady Uryash
- Division of Neonatology, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| | - Jose R. Lopez
- Department of Research, Mount Sinai Medical Center, Miami Beach, FL 33140, USA
| |
Collapse
|
12
|
Wilcox EC, Edelman ER. Substratum interactions determine immune response to allogeneic transplants of endothelial cells. Front Immunol 2022; 13:946794. [PMID: 36003373 PMCID: PMC9393654 DOI: 10.3389/fimmu.2022.946794] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/12/2022] [Indexed: 12/30/2022] Open
Abstract
Endothelial cells (ECs) are central to vascular health but also interact with and regulate the immune system. Changes in endothelial state enable immune cells to migrate into the tissue to facilitate repair and fight infection. ECs modulate the function of immune cells through the expression of adhesion molecules, chemokines, major histocompatibility complex (MHC), and an array of co-stimulatory and inhibitor molecules. These interactions allow ECs to act as antigen presenting cells (APCs) and influence the outcome of immune recognition. This study elucidates how EC microenvironment, vascular cell biology, and immune response are not only connected but interdependent. More specifically, we explored how cell-substratum interactions influence EC antigen presentation and co-stimulation, and how these differences affect allorecognition in animal models of cell transplantation. Investigation of EC state was carried out using RNA sequencing while assessment of the allogeneic response includes measurements of immune cell cytotoxic ability, T cell proliferation, cytokine release, serum antibodies, and histological staining. Differences in substratum led to divergent EC phenotypes which in turn influenced immune response to transplanted cells, both due to the physical barrier of matrix-adhesion and differences in expression of surface markers. ECs grown in 2D on tissue culture plastic or in 3D on collagen scaffolds had significantly different basal levels of MHC expression, co-stimulatory and adhesion molecules. When treated with cytokines to mimic an inflammatory state, ECs did not converge to a single phenotype but rather responded differently based on their substratum. Generally, 3D ECs were more responsive to inflammatory stimuli than 2D ECs. These unique expression patterns measured in vitro also influence immune recognition in vivo. ECs grown in 2D were more likely to provoke a cytotoxic response while 3D ECs induced T cell proliferation. ECs are uniquely configured to sense not only local flow and mechanical forces but a range of markers related to systemic state, including immune function. ECs interact with immune cells with differing results depending on the environment in which the EC-lymphocyte interaction occurs. Therefore, understanding this relationship is essential to predicting and modifying the outcome of EC-immune interacts. We specifically examined the relationship between EC substratum and allorecognition.
Collapse
Affiliation(s)
- Elise C. Wilcox
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, United States
- *Correspondence: Elise C. Wilcox,
| | - Elazer R. Edelman
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, MA, United States
- Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
13
|
Inthawong M, Sunyakumthorn P, Wongwairot S, Anantatat T, Dunachie SJ, Im-Erbsin R, Jones JW, Mason CJ, Lugo LA, Blacksell SD, Day NPJ, Sonthayanon P, Richards AL, Paris DH. A time-course comparative clinical and immune response evaluation study between the human pathogenic Orientia tsutsugamushi strains: Karp and Gilliam in a rhesus macaque (Macaca mulatta) model. PLoS Negl Trop Dis 2022; 16:e0010611. [PMID: 35925895 PMCID: PMC9352090 DOI: 10.1371/journal.pntd.0010611] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Accepted: 06/27/2022] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Scrub typhus is a vector-borne febrile illness caused by Orientia tsutsugamushi transmitted by the bite of Trombiculid mites. O. tsutsugamushi has a high genetic diversity and is increasingly recognized to have a wider global distribution than previously assumed. METHODOLOGY/PRINCIPLE FINDINGS We evaluated the clinical outcomes and host immune responses of the two most relevant human pathogenic strains of O. tsutsugamushi; Karp (n = 4) and Gilliam (n = 4) in a time-course study over 80 days post infection (dpi) in a standardized scrub typhus non-human primate rhesus macaque model. We observed distinct features in clinical progression and immune response between the two strains; Gilliam-infected macaques developed more pronounced systemic infection characterized by an earlier onset of bacteremia, lymph node enlargement, eschar lesions and higher inflammatory markers during the acute phase of infection, when compared to the Karp strain. C-reactive protein (CRP) plasma levels, interferon gamma (IFN-γ, interleukin-1 receptor antagonist (IL-1ra), IL-15 serum concentrations, CRP/IL10- and IFN-γ/IL-10 ratios correlated positively with bacterial load in blood, implying activation of the innate immune response and preferential development of a T helper-type 1 immune response. The O. tsutsugamushi-specific immune memory responses in cells isolated from skin and lymph nodes at 80 dpi were more markedly elevated in the Gilliam-infected macaques than in the Karp-infected group. The comparative cytokine response dynamics of both strains revealed significant up-regulation of IFN-γ, tumor necrosis factor (TNF), IL-15, IL-6, IL-18, regulatory IL-1ra, IL-10, IL-8 and granulocyte-colony-stimulating factor (G-CSF). These data suggest that the clinical outcomes and host immune responses to scrub typhus could be associated with counter balancing effects of pro- and anti-inflammatory cytokine-mediated responses. Currently, no data on characterized time-course comparisons of O. tsutsugamushi strains regarding measures of disease severity and immune response is available. Our study provides evidence for the strain-specificity of host responses in scrub typhus, which supports our understanding of processes at the initial inoculation site (eschar), systemic disease progression, protective and/or pathogenic host immune mechanisms and cellular immune memory function. CONCLUSIONS/SIGNIFICANCE This study characterised an improved intradermal rhesus macaque challenge model for scrub typhus, whereby the Gilliam strain infection associated with higher disease severity in the rhesus macaque model than the previous Karp strain infection. Difficulties associated with inoculum quantitation for obligate-intracellular bacteria were overcome by using functional inoculum titrations in outbred mice. The Gilliam-based rhesus macaque model provides improved endpoint measurements and contributes towards the identification of correlates of protection for future vaccine development.
Collapse
Affiliation(s)
- Manutsanun Inthawong
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Piyanate Sunyakumthorn
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Sirima Wongwairot
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tippawan Anantatat
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas, United States of America
| | - Susanna J. Dunachie
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Rawiwan Im-Erbsin
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - James W. Jones
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Carl J. Mason
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Luis A. Lugo
- Department of Veterinary Medicine, United States Army Medical Directorate, Armed Forces Research Institute of Medical Sciences (USAMD-AFRIMS), Bangkok, Thailand
| | - Stuart D. Blacksell
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Nicholas P. J. Day
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Piengchan Sonthayanon
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Allen L. Richards
- Viral & Rickettsial Diseases Department, Naval Medical Research Center, Silver Spring, Maryland, United States of America
- Department of Preventive Medicine and Biostatistics, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Daniel H. Paris
- Mahidol-Oxford Tropical Medicine Research Unit (MORU), Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
- Department of Medicine, Swiss Tropical and Public Health Institute, Faculty of Medicine, University of Basel, Switzerland
- Department of Clinical Research, Faculty of Medicine, University of Basel, Switzerland
| |
Collapse
|
14
|
Pernot S, Evrard S, Khatib AM. The Give-and-Take Interaction Between the Tumor Microenvironment and Immune Cells Regulating Tumor Progression and Repression. Front Immunol 2022; 13:850856. [PMID: 35493456 PMCID: PMC9043524 DOI: 10.3389/fimmu.2022.850856] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 03/28/2022] [Indexed: 11/29/2022] Open
Abstract
A fundamental concern of the majority of cancer scientists is related to the identification of mechanisms involved in the evolution of neoplastic cells at the cellular and molecular level and how these processes are able to control cancer cells appearance and death. In addition to the genome contribution, such mechanisms involve reciprocal interactions between tumor cells and stromal cells within the tumor microenvironment (TME). Indeed, tumor cells survival and growth rely on dynamic properties controlling pro and anti-tumorigenic processes. The anti-tumorigenic function of the TME is mainly regulated by immune cells such as dendritic cells, natural killer cells, cytotoxic T cells and macrophages and normal fibroblasts. The pro-tumorigenic function is also mediated by other immune cells such as myeloid-derived suppressor cells, M2-tumor-associated macrophages (TAMs) and regulatory T (Treg) cells, as well as carcinoma-associated fibroblasts (CAFs), adipocytes (CAA) and endothelial cells. Several of these cells can show both, pro- and antitumorigenic activity. Here we highlight the importance of the reciprocal interactions between tumor cells and stromal cells in the self-centered behavior of cancer cells and how these complex cellular interactions control tumor progression and repression.
Collapse
Affiliation(s)
- Simon Pernot
- Reprograming Tumor Activity and Associated Microenvironment (RYTME), Bordeaux Institute of Oncology (BRIC)-Unité Mixte de Recherche (UMR) 1312 Inserm, Pessac, France
| | | | - Abdel-Majid Khatib
- Reprograming Tumor Activity and Associated Microenvironment (RYTME), Bordeaux Institute of Oncology (BRIC)-Unité Mixte de Recherche (UMR) 1312 Inserm, Pessac, France.,Institut Bergonié, Bordeaux, France
| |
Collapse
|
15
|
Van Loon E, Lamarthée B, Barba T, Claes S, Coemans M, de Loor H, Emonds MP, Koshy P, Kuypers D, Proost P, Senev A, Sprangers B, Tinel C, Thaunat O, Van Craenenbroeck AH, Schols D, Naesens M. Circulating Donor-Specific Anti-HLA Antibodies Associate With Immune Activation Independent of Kidney Transplant Histopathological Findings. Front Immunol 2022; 13:818569. [PMID: 35281018 PMCID: PMC8904423 DOI: 10.3389/fimmu.2022.818569] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 01/10/2022] [Indexed: 12/17/2022] Open
Abstract
Despite the critical role of cytokines in allograft rejection, the relation of peripheral blood cytokine profiles to clinical kidney transplant rejection has not been fully elucidated. We assessed 28 cytokines through multiplex assay in 293 blood samples from kidney transplant recipients at time of graft dysfunction. Unsupervised hierarchical clustering identified a subset of patients with increased pro-inflammatory cytokine levels. This patient subset was hallmarked by a high prevalence (75%) of donor-specific anti-human leukocyte antigen antibodies (HLA-DSA) and histological rejection (70%) and had worse graft survival compared to the group with low cytokine levels (HLA-DSA in 1.7% and rejection in 33.7%). Thirty percent of patients with high pro-inflammatory cytokine levels and HLA-DSA did not have histological rejection. Exploring the cellular origin of these cytokines, we found a corresponding expression in endothelial cells, monocytes, and natural killer cells in single-cell RNASeq data from kidney transplant biopsies. Finally, we confirmed secretion of these cytokines in HLA-DSA-mediated cross talk between endothelial cells, NK cells, and monocytes. In conclusion, blood pro-inflammatory cytokines are increased in kidney transplant patients with HLA-DSA, even in the absence of histology of rejection. These observations challenge the concept that histology is the gold standard for identification of ongoing allo-immune activation after transplantation.
Collapse
Affiliation(s)
- Elisabet Van Loon
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Baptiste Lamarthée
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Thomas Barba
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital Lyon, Hospices Civils de Lyon, Lyon, France
| | - Sandra Claes
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Maarten Coemans
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Leuven Biostatistics and Statistical Bioinformatics Centre, Department of Public Health and Primary Care, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Henriette de Loor
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Marie-Paule Emonds
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Histocompatibility and Immunogenetics Laboratory, Red Cross-Flanders, Mechelen, Belgium
| | - Priyanka Koshy
- Department of Imaging and Pathology, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Dirk Kuypers
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Paul Proost
- Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Aleksandar Senev
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Histocompatibility and Immunogenetics Laboratory, Red Cross-Flanders, Mechelen, Belgium
| | - Ben Sprangers
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium.,Laboratory of Molecular Immunology, Department of Microbiology, Immunology and Transplantation, Rega Institute for Medical Research, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Claire Tinel
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Olivier Thaunat
- Department of Transplantation, Nephrology and Clinical Immunology, Edouard Herriot Hospital Lyon, Hospices Civils de Lyon, Lyon, France
| | - Amaryllis H Van Craenenbroeck
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| | - Dominique Schols
- Department of Microbiology, Immunology and Transplantation, Laboratory of Virology and Chemotherapy, Rega Institute, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Maarten Naesens
- Department of Microbiology, Immunology and Transplantation, Nephrology and Kidney Transplantation Research Group, Katholieke Universiteit (KU) Leuven, Leuven, Belgium.,Department of Nephrology and Kidney Transplantation, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Mezu-Ndubuisi OJ, Maheshwari A. Role of the Endothelium in Neonatal Diseases. NEWBORN 2022; 1:44-57. [PMID: 35754998 PMCID: PMC9217741 DOI: 10.5005/jp-journals-11002-0025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In both fetal and neonatal physiologic and pathologic processes in most organs, endothelial cells are known to play critical roles. Although the endothelium is one of the most ubiquitous cell type in the body, the tight adherence to the blood vessel wall has made it difficult to study their diverse function and structure. In this article, we have reviewed endothelial cell origins and explored their heterogeneity in terms of structure, function, developmental changes, and their role in inflammatory and infectious diseases. We have also attempted to evaluate the untapped therapeutic potentials of endothelial cells in neonatal disease. This article comprises various peer-reviewed studies, including ours, and an extensive database literature search from EMBASE, PubMed, and Scopus.
Collapse
Affiliation(s)
- Olachi J Mezu-Ndubuisi
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - Akhil Maheshwari
- Global Newborn Society, Clarksville, Maryland, United States of America
| |
Collapse
|
17
|
Lorenz M, Witt E, Völker U, Stangl K, Stangl V, Hammer E. Serum starvation induces sexual dimorphisms in secreted proteins of human umbilical vein endothelial cells (HUVECs) from twin pairs. Proteomics 2022; 22:e2100168. [PMID: 35357760 DOI: 10.1002/pmic.202100168] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Revised: 03/24/2022] [Accepted: 03/24/2022] [Indexed: 11/06/2022]
Abstract
There is growing evidence for sex and gender differences in the clinical manifestation and outcomes of human diseases. Human primary endothelial cells represent a useful cardiovascular model to study sexual dimorphisms at the cellular level. Here, we analyzed sexual dimorphisms of the secretome after serum starvation using HUVECs from twin pairs of the opposite sex to minimize the impact of varying genetic background. HUVECs were starved for 5 and 16 h, respectively, and proteins of the cell culture supernatants were analyzed by tandem mass spectrometry. Altogether, 960 extracellular proteins were identified of which 683 were amendable to stringent quantification. Significant alterations were observed for 324 proteins between long-term and short-term starvation and the majority were similar in both sexes. Only 5 proteins showed significant sex-specific regulation between long- versus short-term starvation. Furthermore, 19 unique proteins with significant sexual dimorphisms at the same time points of serum starvation were observed. A larger number of proteins, e.g. tissue factor inhibitor 2 (TFPI2), displayed higher levels in the supernatants of females compared to male cells after long term serum starvation that might point to higher adaptation capacity of female cells. The overall results demonstrate that male and female cells differ in their secretome. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mario Lorenz
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Eric Witt
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald
| | - Uwe Völker
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald
| | - Karl Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Verena Stangl
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Medizinische Klinik für Kardiologie und Angiologie, Campus Mitte, Berlin, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Berlin
| | - Elke Hammer
- Interfaculty Institute for Genetics and Functional Genomics, University Medicine Greifswald, Greifswald, Germany.,DZHK (German Centre for Cardiovascular Research), partner site Greifswald
| |
Collapse
|
18
|
Liu C, Yang S. A Meta-Analysis of the Influence of Tumor Necrosis Factor- α-308 Gene Polymorphism on Liver Cirrhosis. JOURNAL OF HEALTHCARE ENGINEERING 2022; 2022:9764770. [PMID: 35345657 PMCID: PMC8957422 DOI: 10.1155/2022/9764770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 02/15/2022] [Indexed: 01/30/2023]
Abstract
Cirrhosis is an active hepatic inflammation process of the liver considered as the serious phase of different liver injuries. Epidemiological studies have evaluated the possible association between TNF-α-308G/A gene polymorphism and liver cirrhosis. In this study, we have furthered the study to assess the exact association of TNF-α-308G/A gene polymorphism with liver cirrhosis susceptibility by integrating all available data. Eligible case-control studies were carried out from the establishment of the project to September 2021. Published literature from multiple databases was retrieved, collected, and analyzed by two investigators independently. Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated for every study. Review Manager 5.2 and Stata 15.0 software were used for meta-analysis and stability was assessed by both subgroup analysis and sensitivity analysis. Begg's funnel plot and Egger's regression across the studies were also explored. We examined 22 case-control studies with 2683 cirrhosis patients and 2905 normal controls in four genetic models (GA vs. GG: OR = 0.95, 95%CI: (0.70, 1.30); AA vs. GG: OR = 1.11, 95% CI: (0.66, 1.85), GA + AA vs. GG: OR = 1.00, 95% CI: (0.73, 1.37); AA vs. GA + GG: OR = 1.07, 95%CI: (0.70,1.63)). TNF-α-308G/A gene polymorphism was relatively independent, and the results did not show a significant difference between the two groups. In the subgroup analysis by etiological classification of liver cirrhosis, cirrhosis after HCV infection was positively associated with the risk of TNF-α-308G/A polymorphism (AA vs. GG: OR = 3.02, 95% CI: (1.15, 7.88), AA vs. GA + GG: OR = 2.68, 95% CI: (1.04, 6.95)). The meta-analysis showed TNF-α-308G/A gene polymorphism might not have affected susceptibility for liver cirrhosis. Nevertheless, further and well-designed studies were needed to confirm the findings.
Collapse
Affiliation(s)
- Chang Liu
- Department of Gastroenterology, The First People's Hospital of Chong Qing Liang Jiang New Area, Chongqing 401121, China
| | - Songtao Yang
- Department of Gastroenterology, The First People's Hospital of Chong Qing Liang Jiang New Area, Chongqing 401121, China
| |
Collapse
|
19
|
Kang H, Yu H, Fan J, Cao G. Rotigotine protects against oxidized low-density lipoprotein(ox-LDL)-induced damages in human umbilical vein endothelial cells(HUVECs). Bioengineered 2021; 12:10568-10579. [PMID: 34860135 PMCID: PMC8810014 DOI: 10.1080/21655979.2021.2000224] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Rotigotine is a non-ergoline dopamine agonist that has been licensed for the treatment of Parkinson’s disease. Cardiovascular diseases are the world’s leading cause of death. Ox-LDL- induced endothelial damages are involved in the initiation and progression of cardiovascular diseases. In this study, we assessed the beneficial properties of Rotigotine on ox-LDL-induced insults to HUVECs to highlight its potential use in the treatment of cardiovascular diseases. Our findings show that Rotigotine suppresses the expressions of low-density lipoprotein receptor (LDL-R), proprotein convertase subtilisin/kexin type 9 (PCSK-9), and sterol regulatory element-binding protein (SREBP-2). It also inhibits ox-LDL-induced cholesterol accumulation in endothelial cells (ECs), improves U937 monocytes adhesion, and decreases the representation of NADPH oxidase (NOX-4) and generation of reactive oxygen species (ROS) in endothelial cells (ECs). Furthermore, Rotigotine inhibited the expressions of both vascular cellular adhesion molecule-1 (VCAM-1) and intercellular adhesion molecule-1 (ICAM-1) in HUVECs and had anti-inflammatory efficacy in ox-LDL-induced cells by inhibiting the expressions of pro-inflammatory cytokines. Notably, Rotigotine inhibits the activation of nuclear factor-kappaB (NF-κB) by preventing nuclear translocation of NF-κB p65 and reducing the luciferase activity of NF-κB reporter. We, therefore, conclude that these effects of Rotigotine on HUVECs suggest that it may play a therapeutic role in cardiovascular diseases.
Collapse
Affiliation(s)
- Hui Kang
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hui Yu
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Jingxiu Fan
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ge Cao
- Department of Cardiovascular Surgery, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
20
|
Rashed ER, Abdel-Rafei MK, Thabet NM. Roles of Simvastatin and Sildenafil in Modulation of Cranial Irradiation-Induced Bystander Multiple Organs Injury in Rats. Inflammation 2021; 44:2554-2579. [PMID: 34420155 DOI: 10.1007/s10753-021-01524-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/16/2021] [Indexed: 01/11/2023]
Abstract
In radiobiology and radiation oncology fields, the observation of a phenomenon called radiation-induced bystander effect (RIBE) has introduced the prospect of remotely located tissues' affection. This phenomenon has been broadly developed to involve the concept of RIBE, which are relevant to the radiation-induced response of a distant tissue other than the irradiated one. The current study aimed at investigating each of the RIBE of cranial irradiation on oxidative and inflammatory status in different organs such as liver, kidney, heart, lung, and spleen. Being a vital target of the cholinergic anti-inflammatory response to an inflammatory stimulus, the splenic α-7-nicotinic acetylcholine receptor (α-7nAchR) was evaluated and the hepatic contents of thioredoxin, peroxisome proliferator-activated receptor-alpha and paraoxinase-1 (Trx/PPAR-α/PON) were also assessed as indicators for the liver oxidative stress and inflammatory responses. Being reported to act as antioxidant and anti-inflammatory agents, simvastatin (SV) and/or sildenafil (SD) were investigated for their effects against RIBE on these organs. These objectives were achieved via the biochemical assessments and the histopathological tissues examinations. Five experimental groups, one sham irradiated and four irradiated groups, were exposed to cranial irradiation at dose level of 25 Gy using an experimental irradiator with a Cobalt (Co60) source, RIBE, RIBE + SV (20 mg.(kg.bw)-1 day-1), RIBE + SD (75 mg.(kg.bw)-1 day-1), and RIBE + SV + SD. Cranial irradiation induced structural, biochemical, and functional dys-regulations in non-targeted organs. RIBE-induced organs' injuries have been significantly corrected by the administration of SV and/or SD. Our results suggest the possibility of a potentiated interaction between SV and SD in the modulation of the RIBE associated with head and neck radiotherapy.
Collapse
Affiliation(s)
- Engy Refaat Rashed
- Drug Radiation Research Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| | - Mohamed Khairy Abdel-Rafei
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Noura Magdy Thabet
- Radiation Biology Department, National Centre for Radiation Research and Technology (NCRRT), Egyptian Atomic Energy Authority, Cairo, Egypt
| |
Collapse
|
21
|
Makarewicz-Wujec M, Henzel J, Kępka C, Kruk M, Wardziak Ł, Trochimiuk P, Parzonko A, Dzielińska Z, Demkow M, Kozłowska-Wojciechowska M. Usefulness of MCP-1 Chemokine in the Monitoring of Patients with Coronary Artery Disease Subjected to Intensive Dietary Intervention: A Pilot Study. Nutrients 2021; 13:nu13093047. [PMID: 34578925 PMCID: PMC8467171 DOI: 10.3390/nu13093047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/21/2021] [Accepted: 08/27/2021] [Indexed: 12/11/2022] Open
Abstract
Monocyte chemotactic protein-1 (MCP-1) plays an important role in the entire atherosclerotic process, from atherogenesis to destabilisation of the atherosclerotic plaque. The purpose of this study is to evaluate the effect of the dietary approaches to stop hypertension (DASH) diet in patients with coronary artery disease on the MCP-1 plasma concentration and to evaluate the potential usefulness of this chemokine as a marker of change in the volume and composition of coronary plaque. Material and method. As part of the dietary intervention to stop coronary atherosclerosis in computed tomography (DISCO-CT) study, patients were randomised to an intervention group (n = 40) in which the DASH diet was introduced, and to a control group (n = 39) with no dietary intervention. In the DASH group, dietary counselling was provided at all follow-up visits within 12 months of the follow-up period. MCP-1 plasma concentration was determined using enzyme-linked immunosorbent assay (ELISA). Coronary plaque analysis was performed using a semi-automated plaque analysis software system (QAngioCT, Medis, The Netherlands). Results. In the DASH group, MCP-1 plasma concentration significantly decreased by 34.1 pg/mL (p = 0.01), while in the control group, the change in MPC-1 was not significant. Significant inverse correlations were revealed for the change in MCP-1 plasma concentration and change in the consumption of vitamin C and dietary fibre both in the DASH (r = −0.519, p = 0.0005; r = −0.353, p = 0.025, respectively) and in the control group (r = −0.488 p = 0.001; r = −0.502, p = 0.001, respectively). In patients with the highest decrease in percent atheroma volume (PAV), a significant positive correlation was observed between the change in MCP-1 plasma concentration and changes in PAV (r = 0.428, p = 0.033) and calcified plaque component (r = 0.468, p = 0.018), while the change in noncalcified plaque component correlated inversely with change in MCP1 (r = −0.459, p = 0.021). Conclusion. Dietary intervention based on the DASH diet model reduces the MCP-1plasma concentration, mostly due to an increased intake of plant-derived, fibre-rich foods and antioxidants. The change in MCP-1 plasma concentration seems to reflect changes in the atheroma volume and proportions between the calcified and non-calcified plaque elements.
Collapse
Affiliation(s)
- Magdalena Makarewicz-Wujec
- Department of Clinical Pharmacy and Pharmaceutical Care, Medical University of Warsaw, 02-091 Warsaw, Poland;
- Correspondence: (M.M.-W.); (J.H.); Tel.: +48-225-720-985 (M.M.-W.); +48-223-434-342 (J.H.)
| | - Jan Henzel
- Department of Coronary and Structural Heart Diseases, Cardinal Stefan Wyszynski Institute of Cardiology, 04-628 Warsaw, Poland; (C.K.); (M.K.); (Ł.W.); (P.T.); (Z.D.); (M.D.)
- Correspondence: (M.M.-W.); (J.H.); Tel.: +48-225-720-985 (M.M.-W.); +48-223-434-342 (J.H.)
| | - Cezary Kępka
- Department of Coronary and Structural Heart Diseases, Cardinal Stefan Wyszynski Institute of Cardiology, 04-628 Warsaw, Poland; (C.K.); (M.K.); (Ł.W.); (P.T.); (Z.D.); (M.D.)
| | - Mariusz Kruk
- Department of Coronary and Structural Heart Diseases, Cardinal Stefan Wyszynski Institute of Cardiology, 04-628 Warsaw, Poland; (C.K.); (M.K.); (Ł.W.); (P.T.); (Z.D.); (M.D.)
| | - Łukasz Wardziak
- Department of Coronary and Structural Heart Diseases, Cardinal Stefan Wyszynski Institute of Cardiology, 04-628 Warsaw, Poland; (C.K.); (M.K.); (Ł.W.); (P.T.); (Z.D.); (M.D.)
| | - Piotr Trochimiuk
- Department of Coronary and Structural Heart Diseases, Cardinal Stefan Wyszynski Institute of Cardiology, 04-628 Warsaw, Poland; (C.K.); (M.K.); (Ł.W.); (P.T.); (Z.D.); (M.D.)
| | - Andrzej Parzonko
- Department of Pharmacognosy and Molecular Basis of Phytotherapy, Medical University of Warsaw, 02-091 Warsaw, Poland;
| | - Zofia Dzielińska
- Department of Coronary and Structural Heart Diseases, Cardinal Stefan Wyszynski Institute of Cardiology, 04-628 Warsaw, Poland; (C.K.); (M.K.); (Ł.W.); (P.T.); (Z.D.); (M.D.)
| | - Marcin Demkow
- Department of Coronary and Structural Heart Diseases, Cardinal Stefan Wyszynski Institute of Cardiology, 04-628 Warsaw, Poland; (C.K.); (M.K.); (Ł.W.); (P.T.); (Z.D.); (M.D.)
| | | |
Collapse
|
22
|
CSF Diagnostics: A Potentially Valuable Tool in Neurodegenerative and Inflammatory Disorders Involving Motor Neurons: A Review. Diagnostics (Basel) 2021; 11:diagnostics11091522. [PMID: 34573864 PMCID: PMC8470638 DOI: 10.3390/diagnostics11091522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 11/16/2022] Open
Abstract
Cerebrospinal fluid (CSF) diagnostics has emerged as a valid tool for a variety of neurological diseases. However, CSF diagnostics has been playing a subordinate role in the diagnosis of many neurological conditions. Thus, in the multitude of neuromuscular diseases in which motor neurons are affected, a CSF sample is rarely taken routinely. However, CSF diagnostics has the potential to specify the diagnosis and monitor the treatment of neuromuscular disorders. In this review, we therefore focused on a variety of neuromuscular diseases, among them amyotrophic lateral sclerosis (ALS), peripheral neuropathies, and spinal muscular atrophy (SMA), for which CSF diagnostics has emerged as a promising option for determining the disease itself and its progression. We focus on potentially valuable biomarkers among different disorders, such as neurofilaments, cytokines, other proteins, and lipids to determine their suitability, differentiating between different neurological disorders and their potential to determine early disease onset, disease progression, and treatment outcome. We further recommend novel approaches, e.g., the use of mass spectrometry as a promising alternative techniques to standard ELISA assays, potentially enhancing biomarker significance in clinical applications.
Collapse
|
23
|
Peng Y, Ma Y, Bao Y, Liu Z, Chen L, Dai F, Li Z. Electrospun PLGA/SF/artemisinin composite nanofibrous membranes for wound dressing. Int J Biol Macromol 2021; 183:68-78. [PMID: 33892031 DOI: 10.1016/j.ijbiomac.2021.04.021] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 03/31/2021] [Accepted: 04/03/2021] [Indexed: 12/23/2022]
Abstract
Combining biodegradable materials with natural plant components for wound dressing has been receiving significant attention. ART is a sesquiterpene lactone compound extracted from Artemisia annua L., possessing multiple pharmacological effects including antibacterial activity and anti-inflammatory property. Herein, the blended polylactic acid glycolic acid (PLGA)/silk fibroin (SF) membranes loaded with artemisinin (ART) are fabricated through electrospinning. With aid of SF, the fabricated membranes have a good sustained-release effect, and the accumulated ART release can reach 69% after three weeks. PLGA/SF/ART membranes exhibit favorable anti-inflammatory and cell compatibility in vitro evaluations. The in vivo experiment indicates that PLGA/SF/ART2 membranes can shorten the inflammation period and enhance skin regeneration in a full-thickness wound model through down-regulating the expressions of pro-inflammatory cytokines IL-1β and TNF-α. To sum up, the fabricated PLGA/SF/ART2 composite membranes with anti-inflammatory properties can be a proposal wound dressing for chronic wound healing.
Collapse
Affiliation(s)
- Yan Peng
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Yan Ma
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Yu Bao
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Zulan Liu
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Li Chen
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China
| | - Fangyin Dai
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; Key Laboratory for Sericulture Biology and Genetic Breeding, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China.
| | - Zhi Li
- State Key Laboratory of Silkworm Genome Biology, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, College of Sericulture, Textile and Biomass Science, Southwest University, Chongqing 400715, China.
| |
Collapse
|
24
|
Yazbeck R, Jaenisch SE, Abbott CA. Dipeptidyl peptidase 4 inhibitors: Applications in innate immunity? Biochem Pharmacol 2021; 188:114517. [PMID: 33722535 PMCID: PMC7954778 DOI: 10.1016/j.bcp.2021.114517] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 03/04/2021] [Accepted: 03/05/2021] [Indexed: 12/25/2022]
Abstract
Dipeptidyl peptidase (DPP)-4 inhibitors are a class of orally available, small molecule inhibitors that prolong the insulinotropic activity of the incretin hormone glucagon-like peptide-1 (GLP-1) and are highly effective for the treatment of Type-2 diabetes. DPP4 can also cleave several immunoregulatory peptides including chemokines. Emerging evidence continues to implicate DPP4 inhibitors as immunomodulators, with recent findings suggesting DPP4 inhibitors modify specific aspects of innate immunity. This review summarises recent insights into how DPP4 inhibitors could be implicated in endothelial, neutrophil and monocyte/macrophage mediated immunity. Additionally, this review highlights additional avenues of research with DPP4 inhibitors in the context of the COVID-19 pandemic.
Collapse
Affiliation(s)
- R Yazbeck
- College of Medicine and Public Health & Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia; College of Science and Engineering, Flinders University, Adelaide, Australia.
| | - S E Jaenisch
- College of Medicine and Public Health & Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia; College of Science and Engineering, Flinders University, Adelaide, Australia.
| | - C A Abbott
- College of Medicine and Public Health & Flinders Health and Medical Research Institute, Flinders University, Adelaide, Australia; College of Science and Engineering, Flinders University, Adelaide, Australia.
| |
Collapse
|
25
|
Ryan RYM, Seymour J, Loukas A, Lopez JA, Ikonomopoulou MP, Miles JJ. Immunological Responses to Envenomation. Front Immunol 2021; 12:661082. [PMID: 34040609 PMCID: PMC8141633 DOI: 10.3389/fimmu.2021.661082] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/01/2021] [Indexed: 01/05/2023] Open
Abstract
Venoms are complex mixtures of toxic compounds delivered by bite or sting. In humans, the consequences of envenomation range from self-limiting to lethal. Critical host defence against envenomation comprises innate and adaptive immune strategies targeted towards venom detection, neutralisation, detoxification, and symptom resolution. In some instances, venoms mediate immune dysregulation that contributes to symptom severity. This review details the involvement of immune cell subtypes and mediators, particularly of the dermis, in host resistance and venom-induced immunopathology. We further discuss established venom-associated immunopathology, including allergy and systemic inflammation, and investigate Irukandji syndrome as a potential systemic inflammatory response. Finally, this review characterises venom-derived compounds as a source of immune modulating drugs for treatment of disease.
Collapse
Affiliation(s)
- Rachael Y. M. Ryan
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
- School of Environment and Sciences, Griffith University, Nathan, QLD, Australia
| | - Jamie Seymour
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - Alex Loukas
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
| | - J. Alejandro Lopez
- School of Environment and Sciences, Griffith University, Nathan, QLD, Australia
- QIMR Berghofer Medical Research Institute, The University of Queensland, Herston, QLD, Australia
| | - Maria P. Ikonomopoulou
- Translational Venomics Group, Madrid Institute for Advanced Studies (IMDEA) in Food, CEI UAM+CSIC, Madrid, Spain
- Institute for Molecular Bioscience, University of Queensland, Brisbane, QLD, Australia
| | - John J. Miles
- Division of Tropical Health and Medicine, Australian Institute of Tropical Health and Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns, QLD, Australia
- Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
26
|
Gao A, Wang Y, Gao X, Tian W. LCZ696 ameliorates lipopolysaccharide-induced endothelial injury. Aging (Albany NY) 2021; 13:9582-9591. [PMID: 33839697 PMCID: PMC8064163 DOI: 10.18632/aging.202692] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 01/14/2021] [Indexed: 12/11/2022]
Abstract
Lipopolysaccharide (LPS)-induced endothelial dysfunction plays an important role in the pathogenesis of cardiovascular diseases. LCZ696, the dual-acting angiotensin receptor blocker, and neprilysin inhibitor has been used for the treatment of heart failure with reduced ejection fraction. Recent work suggests that LCZ696 therapy might have an anti-inflammatory effect in cardiovascular tissue. In the current study, we show that LCZ696 attenuates LPS-induced oxidative stress by reducing the production of intracellular reactive oxygen species (ROS) and the measurements of malonyl dialdehyde (MDA) level in human umbilical vascular endothelial cells (HUVECs). LCZ696 inhibits LPS-induced expressions and secretions of the pro-inflammatory cytokines, interleukin-6 (IL-6), interleukin-1α (IL-1α), and tumor necrosis factor β (TNF-β) as well as the chemokines, monocyte chemotactic protein 1 (MCP-1), and chemokine (C-X-C motif) ligand 1 protein (CXCL1). Additionally, we found that LCZ696 reduces LPS-induced expressions of vascular cell adhesion molecule 1 (VCAM-1) and P-selectin and the attachment of U937 monocytes to HUVECs. Mechanistically, LCZ696 prevents LPS-induced activation of the TLR4/Myd88 pathway and nuclear translocation of nuclear factor kappa-B (NF-κB) p65 factor. Based on these findings, we conclude that LCZ696 is capable of ameliorating LPS-induced endothelial dysfunction via anti-inflammatory properties.
Collapse
Affiliation(s)
- Aihong Gao
- Department of Cardiology, NO.215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, China
| | - Yu Wang
- Department of Cardiology, NO.215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, China
| | - Xiao Gao
- Department of Pathology, NO.215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, China
| | - Wei Tian
- Department of Pathology, NO.215 Hospital of Shaanxi Nuclear Industry, Xianyang 712000, China
| |
Collapse
|
27
|
Chang R, Mamun A, Dominic A, Le NT. SARS-CoV-2 Mediated Endothelial Dysfunction: The Potential Role of Chronic Oxidative Stress. Front Physiol 2021; 11:605908. [PMID: 33519510 PMCID: PMC7844210 DOI: 10.3389/fphys.2020.605908] [Citation(s) in RCA: 80] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2020] [Accepted: 12/09/2020] [Indexed: 01/08/2023] Open
Abstract
Endothelial cells have emerged as key players in SARS-CoV-2 infection and COVID-19 inflammatory pathologies. Dysfunctional endothelial cells can promote chronic inflammation and disease processes like thrombosis, atherosclerosis, and lung injury. In endothelial cells, mitochondria regulate these inflammatory pathways via redox signaling, which is primarily achieved through mitochondrial reactive oxygen species (mtROS). Excess mtROS causes oxidative stress that can initiate and exacerbate senescence, a state that promotes inflammation and chronic endothelial dysfunction. Oxidative stress can also activate feedback loops that perpetuate mitochondrial dysfunction, mtROS overproduction, and inflammation. In this review, we provide an overview of phenotypes mediated by mtROS in endothelial cells - such as mitochondrial dysfunction, inflammation, and senescence - as well as how these chronic states may be initiated by SARS-CoV-2 infection of endothelial cells. We also propose that SARS-CoV-2 activates mtROS-mediated feedback loops that cause long-term changes in host redox status and endothelial function, promoting cardiovascular disease and lung injury after recovery from COVID-19. Finally, we discuss the implications of these proposed pathways on long-term vascular health and potential treatments to address these chronic conditions.
Collapse
Affiliation(s)
- Ryan Chang
- College of Arts & Sciences, Washington University in St. Louis, St. Louis, MO, United States
| | - Abrar Mamun
- Wiess School of Natural Sciences, Rice University, Houston, TX, United States
| | - Abishai Dominic
- Department of Molecular and Cellular Medicine, College of Medicine, Texas A&M University, College Station, TX, United States
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Center for Cardiovascular Regeneration, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
28
|
Neutrophil-Derived Semaphorin 4D Induces Inflammatory Cytokine Production of Endothelial Cells via Different Plexin Receptors in Kawasaki Disease. BIOMED RESEARCH INTERNATIONAL 2021; 2020:6663291. [PMID: 33381571 PMCID: PMC7759398 DOI: 10.1155/2020/6663291] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 11/27/2020] [Accepted: 12/01/2020] [Indexed: 12/21/2022]
Abstract
Inflammation of endothelial cells (ECs) plays an important role in the pathogenesis of coronary artery lesions (CALs) in Kawasaki disease (KD). Semaphorin 4D (Sema4D) is the first semaphorin shown to have immunoregulatory functions by interacting with its receptors—plexin Bs. Recently, Sema4D has been reported to exert a proinflammatory effect on the endothelium and to be involved in cardiovascular disease. However, the role of Sema4D in KD remains unknown. This study was aimed at revealing the change of soluble Sema4D (sSema4D) in the serum of patients with KD and the effect of the sSema4D-plexin axis on the production of proinflammatory cytokines from human coronary endothelial cells (HCAECs) stimulated with sera from KD patients. Our results showed that serum sSema4D levels were specifically elevated in KD patients, especially in those with CALs, and correlated positively with disease severity and serum concentrations of interleukin- (IL-) 1β, IL-6, and IL-8. The disintegrin and metalloproteinase domain 17- (AMAM17-) mediated Sema4D shedding from neutrophils contributed to the elevation of sSema4D in the serum of KD patients. Furthermore, we found that Sema4D induced IL-1β production of HCAECs via plexin B2, whereas it promoted IL-6 and IL-8 production via plexin B1. Moreover, the expression of both plexin B1 and plexin B2 was upregulated in HCAECs treated with KD sera, and silencing of the two plexin receptors suppressed the overexpression of IL-1β, IL-6, and IL-8 in KD serum-treated HCAECs. Thus, our findings indicated that sSema4D released from neutrophils participates in the pathogenesis of KD-CALs by promoting inflammatory cytokine production of ECs via both plexin B1 and plexin B2, and Sema4D may be a novel predictor for KD-CALs and a candidate therapeutic target for anti-inflammatory strategies of KD.
Collapse
|
29
|
Whitmore HAB, Amarnani D, O'Hare M, Delgado-Tirado S, Gonzalez-Buendia L, An M, Pedron J, Bushweller JH, Arboleda-Velasquez JF, Kim LA. TNF-α signaling regulates RUNX1 function in endothelial cells. FASEB J 2020; 35:e21155. [PMID: 33135824 PMCID: PMC7821222 DOI: 10.1096/fj.202001668r] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/25/2020] [Accepted: 10/19/2020] [Indexed: 12/21/2022]
Abstract
Runt‐related transcription factor 1 (RUNX1) acts as a mediator of aberrant retinal angiogenesis and has been implicated in the progression of proliferative diabetic retinopathy (PDR). Patients with PDR, retinopathy of prematurity (ROP), and wet age‐related macular degeneration (wet AMD) have been found to have elevated levels of Tumor Necrosis Factor‐alpha (TNF‐α) in the eye. In fibrovascular membranes (FVMs) taken from patients with PDR RUNX1 expression was increased in the vasculature, while in human retinal microvascular endothelial cells (HRMECs), TNF‐α stimulation causes increased RUNX1 expression, which can be modulated by RUNX1 inhibitors. Using TNF‐α pathway inhibitors, we determined that in HRMECs, TNF‐α‐induced RUNX1 expression occurs via JNK activation, while NF‐κB and p38/MAPK inhibition did not affect RUNX1 expression. JNK inhibitors were also effective at stopping high d‐glucose‐stimulated RUNX1 expression. We further linked JNK to RUNX1 through Activator Protein 1 (AP‐1) and investigated the JNK‐AP‐1‐RUNX1 regulatory feedback loop, which can be modulated by VEGF. Additionally, stimulation with TNF‐α and d‐glucose had an additive effect on RUNX1 expression, which was downregulated by VEGF modulation. These data suggest that the downregulation of RUNX1 in conjunction with anti‐VEGF agents may be important in future treatments for the management of diseases of pathologic ocular angiogenesis.
Collapse
Affiliation(s)
- Hannah A B Whitmore
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Dhanesh Amarnani
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Michael O'Hare
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Santiago Delgado-Tirado
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Lucia Gonzalez-Buendia
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Miranda An
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Julien Pedron
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - John H Bushweller
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
| | - Joseph F Arboleda-Velasquez
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| | - Leo A Kim
- Schepens Eye Research Institute of Massachusetts Eye and Ear Infirmary, Boston, MA, USA.,Department of Ophthalmology, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
30
|
Targeted anti-inflammatory therapy is a new insight for reducing cardiovascular events: A review from physiology to the clinic. Life Sci 2020; 253:117720. [PMID: 32360620 DOI: 10.1016/j.lfs.2020.117720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/30/2022]
Abstract
Despite considerable progressions, cardiovascular disease (CVD) is still one of the major causes of mortality around the world, indicates an important and unmet clinical need. Recently, extensive studies have been performed on the role of inflammatory factors as either a major or surrogate factor in the pathophysiology of CVD. Epidemiological observations suggest the theory of the role of inflammatory mediators in the development of cardiovascular events. This may support the idea that targeted anti-inflammatory therapies, on the background of traditional validated medical therapies, can play a significant role in prevention and even reduction of cardiovascular disorders. Many randomized controlled trials have shown that drugs commonly useful for primary and secondary prevention of CVD have an anti-inflammatory mechanism. Further, many anti-inflammatory drugs are being examined because of their potential to reduce the risk of cardiovascular problems. In this study, we review the process of inflammation in the development of cardiovascular events, both in vivo and clinical evidence in immunotherapy for CVD.
Collapse
|
31
|
Haghighatdoost F, Gholami A, Hariri M. Effect of grape polyphenols on selected inflammatory mediators: A systematic review and meta-analysis randomized clinical trials. EXCLI JOURNAL 2020; 19:251-267. [PMID: 32327953 PMCID: PMC7174578 DOI: 10.17179/excli2020-1011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/25/2020] [Indexed: 12/17/2022]
Abstract
Grapes contain different polyphenols and might prevent inflammation by reducing Nitric Oxide (NO) inactivation through antioxidative enzymes. The aim of this article was to demonstrate the effects of grape polyphenols on the selected inflammatory mediators, such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-a), and high-sensitivity C-reactive protein (hs-CRP). To find papers assessing the effects of grape polyphenols on inflammatory mediators, electronic data bases, including ISI web of science, PubMed/Medline, SCOPUS, and Google scholar, were searched up to March 2019. Delphi checklist was used for evaluating the qualities of the included articles. The protocol was registered in PROSPERO (No. CRD42019116695). The mean changes in the intervention and control groups were calculated by subtracting the end values from the baselines. Then, the difference between the two changes was measured and utilized as the effect size in meta-analysis. 9 and 8 articles were included in the systematic review and meta-analysis, respectively. Our results indicated that grape polyphenols did not reduce hs-CRP levels, but omission of one article could lead to a significant reduction in hs-CRP (Weight Mean Difference (WMD): −0.54 mg/L, 95 % CI: −1.02, -0.06; P=0.026, I2=0.0 %). Regarding IL-6 and TNF-α, no significant changes were observed in the intervention compared to the control group (WMD: 0.04 pg/mL, 95 % CI: −0.02, 0.28; P=0.744, I2=0.0 %, WMD: -0.10 pg/mL, 95 % CI: −0.25, 0.05; P=0.183, I2=0.0 %, respectively). We found no beneficial effects of grape polyphenols on the selected inflammatory mediators. Still, more studies with higher doses of polyphenols, longer treatment durations, different sources of grape polyphenols, and larger numbers of participants are required.
Collapse
Affiliation(s)
- Fahimeh Haghighatdoost
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ali Gholami
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.,Department of Epidemiology & Biostatistics, School of Public Health, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Mitra Hariri
- Noncommunicable Diseases Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| |
Collapse
|
32
|
Rossi M, Buonuomo PS, Battafarano G, Conforti A, Mariani E, Algeri M, Pelle S, D'Agostini M, Macchiaiolo M, De Vito R, Gonfiantini MV, Jenkner A, Rana I, Bartuli A, Del Fattore A. Dissecting the mechanisms of bone loss in Gorham-Stout disease. Bone 2020; 130:115068. [PMID: 31525474 DOI: 10.1016/j.bone.2019.115068] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 09/11/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022]
Abstract
Gorham-Stout disease (GSD) is a rare disorder characterized by progressive osteolysis and angiomatous proliferation. Since the mechanisms leading to bone loss in GSD are not completely understood, we performed histological, serum, cellular and molecular analyses of 7 patients. Increased vessels, osteoclast number and osteocyte lacunar area were revealed in patients' bone biopsies. Biochemical analysis of sera showed high levels of ICTP, Sclerostin, VEGF-A and IL-6. In vitro experiments revealed increased osteoclast differentiation and activity, and impaired mineralization ability of osteoblasts. To evaluate the involvement of systemic factors in GSD, control cells were treated with patients' sera and displayed an increase of osteoclastogenesis, bone resorption activity and a reduction of osteoblast function. Interestingly, GSD sera stimulated the vessel formation by endothelial cells EA.hy926. These results suggest that bone cell autonomous alterations with the cooperation of systemic factors are involved in massive bone loss and angiomatous proliferation observed in GSD patients.
Collapse
Affiliation(s)
- Michela Rossi
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Giulia Battafarano
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy
| | - Antonella Conforti
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Rome, Italy
| | - Eda Mariani
- Research Laboratories, Bambino Gesù Children's Hospital, Rome, Italy
| | - Mattia Algeri
- Department of Pediatric Hematology and Oncology, Bambino Gesù Children's Hospital, Rome, Italy
| | | | | | - Marina Macchiaiolo
- Rare Diseases and Medical Genetic Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Rita De Vito
- Histopathology, Bambino Gesù Children's Hospital, Rome, Italy
| | | | - Alessandro Jenkner
- Division of Immunology and Infectious Diseases Department of Pediatrics, Bambino Gesù Children Hospital, Rome, Italy
| | - Ippolita Rana
- Rare Diseases and Medical Genetic Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Andrea Bartuli
- Rare Diseases and Medical Genetic Unit, Bambino Gesù Children's Hospital, Rome, Italy
| | - Andrea Del Fattore
- Bone Physiopathology Group, Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, Rome, Italy.
| |
Collapse
|
33
|
Zhang Z, Wu Y, Wu B, Qi Q, Li H, Lu H, Fan C, Feng C, Zuo J, Niu L, Tang W. DZ2002 ameliorates fibrosis, inflammation, and vasculopathy in experimental systemic sclerosis models. Arthritis Res Ther 2019; 21:290. [PMID: 31842999 PMCID: PMC6916442 DOI: 10.1186/s13075-019-2074-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 11/28/2019] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Systemic sclerosis is a multisystem inflammatory and vascular lesion leading to extensive tissue fibrosis. A reversible S-adenosyl-l-homocysteine hydrolase (SAHH) inhibitor, DZ2002, modulates the pathologic processes of various inflammatory diseases and autoimmune diseases. This study is designed to investigate the therapeutic potentiality of DZ2002 for experimental systemic sclerosis models. METHODS The anti-inflammatory and anti-fibrotic features of DZ2002 and its mechanisms were investigated in a bleomycin (BLM)-induced dermal fibrosis mice model. The effects of DZ2002 on expression of extracellular matrix components and TGF-β signaling in human dermal fibroblasts were analyzed. Simultaneously, the effects of DZ2002 on macrophage activation and endothelial cell adhesion molecule expression were also evaluated. RESULTS DZ2002 significantly attenuated dermal fibrosis in BLM-induced mice. Consistently, DZ2002 inhibited the expression of various molecules associated with dermal fibrosis, including transforming growth factor β1, connective tissue growth factor, tumor necrosis factor-α, interferon-γ, IL-1β, IL-4, IL-6, IL-10, IL-12p40, IL-17A, and monocyte chemotactic protein 1 in the lesional skin of BLM-induced mice. Furthermore, DZ2002 decreased the proportion of macrophages, neutrophils, and T cells (especially T helper cells) in the skin tissue of BLM-induced mice. In addition, DZ2002 attenuated both M1 macrophage and M2 macrophage differentiation in vivo and in vitro. Importantly, DZ2002 directly reversed the profibrotic phenotype of transforming growth factor-β1-treated dermal fibroblasts and suppressed ICAM-1, VCAM-1, VEGF, bFGF, and ET-1 expression in endothelial cells. Finally, our investigations showed that DZ2002 relieved systemic sclerosis by regulating fibrosis TGF-β/Smad signaling pathway. CONCLUSIONS DZ2002 prevents the development of experimental dermal fibrosis by reversing the profibrotic phenotype of various cell types and would be a potential drug for the treatment of systemic sclerosis.
Collapse
Affiliation(s)
- Zongwang Zhang
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Baoshan District, Shanghai, 200444, China
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yanwei Wu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Bing Wu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qing Qi
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Heng Li
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huimin Lu
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chen Fan
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chunlan Feng
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Jianping Zuo
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Lili Niu
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Baoshan District, Shanghai, 200444, China.
| | - Wei Tang
- Laboratory of Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
34
|
Basurto L, Gregory MA, Hernández SB, Sánchez-Huerta L, Martínez AD, Manuel-Apolinar L, Avelar FJ, Alonso LAM, Sánchez-Arenas R. Monocyte chemoattractant protein-1 (MCP-1) and fibroblast growth factor-21 (FGF-21) as biomarkers of subclinical atherosclerosis in women. Exp Gerontol 2019; 124:110624. [DOI: 10.1016/j.exger.2019.05.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 05/28/2019] [Accepted: 05/28/2019] [Indexed: 11/30/2022]
|
35
|
Sun Y, Wang S, Zhao L, Zhang B, Chen H. IFN-γ and TNF-α aggravate endothelial damage caused by CD123-targeted CAR T cell. Onco Targets Ther 2019; 12:4907-4925. [PMID: 31417286 PMCID: PMC6600319 DOI: 10.2147/ott.s205678] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 05/13/2019] [Indexed: 01/08/2023] Open
Abstract
Background: CD123-targeted chimeric antigen receptor (CAR) T cell (CART123) for the treatment of acute myeloid leukemia (AML) and blastic plasmacytoid dendritic cell neoplasm has exhibited potential in clinical trials. However, capillary leakage syndrome, which is associated with endothelial cells damage, is under intensive focus in CART123 therapy. Purpose: The present study aimed to explore the change in CD123 in endothelial cells and the injury to endothelial cells caused by CART123. Methods: The expression of CD123 and cytotoxicity were assessed by flow cytometry. Cytokine release was assessed by ELISA. An in vitro co-culture model was designed to mimic the status, wherein CART123 was stimulated and cytokines were released. Results: In the current study, CART123 exhibited cytotoxicity and the effects of cytokine production on endothelium, and the upregulation of CD123 enhanced the cytotoxicity. The addition of interferon (IFN)-γ and tumor necrosis factor (TNF)-α neutralizing antibodies can effectively reverse the upregulation of CD123 on the endothelial cells caused by CART123, while the cytotoxicity of CART123 in AML cell lines was not affected in vitro. Second, we proved that CD123 expresses in CART123 and would be upregulated after activation, putatively causing an overactivated and fratricide effect. Conclusion: In summary, this study identified that the expression of CD123 on endothelial cells could be upregulated when co-cultured with CART123. Furthermore, IFN-γ and TNF-α could aggravate endothelial damage caused by CART123 in vitro.
Collapse
Affiliation(s)
- Yao Sun
- Academy of Military Medical Sciences , Beijing 100071, People's Republic of China.,Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Shenyu Wang
- Academy of Military Medical Sciences , Beijing 100071, People's Republic of China.,Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Long Zhao
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Bin Zhang
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| | - Hu Chen
- Department of Hematopoietic Stem Cell Transplantation, The Fifth Medical Center of Chinese PLA General Hospital (Former 307th Hospital of PLA), the Research Institute of Hematopoietic Stem Cell of the People's Liberation Army, Beijing 100071, People's Republic of China.,Beijing Key Laboratory of Hematopoietic Stem Cell Therapy and Transformation Research , Beijing 100071, People's Republic of China
| |
Collapse
|
36
|
Ginkgolide B Mediated Alleviation of Inflammatory Cascades and Altered Lipid Metabolism in HUVECs via Targeting PCSK-9 Expression and Functionality. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7284767. [PMID: 31281844 PMCID: PMC6590504 DOI: 10.1155/2019/7284767] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/13/2019] [Accepted: 05/26/2019] [Indexed: 01/06/2023]
Abstract
The potential of oxidized-LDL (Ox-LDL) to elicit inflammatory responses in macrophages leading to the atherosclerosis (AS) progression is well known. Since proprotein convertase subtilisin/Kexin-9 (PCSK-9), the posttranslational regulator of LDL-receptor, is associated with elevated LDL in the circulation, the present report was aimed to uncover the ameliorative effects of Ginkgolide B, a terpenic lactone from Ginkgo biloba, against Ox-LDL-induced alterations in cholesterol metabolism in HUVECs. Consequently, our results demonstrated that incubation with Ox-LDL significantly upregulated the PCSK-9 expression in HUVECs, which was significantly downregulated, both at mRNA and protein level, after Ginkgolide B treatment via subsequent suppression of sterol element binding protein (SREBP-2) expression. Moreover, Ginkgolide B-mediated inhibition of PCSK-9 activity was also validated by in silico methods which revealed that it interferes the PSCK-9 interaction with LDL-receptor (LDL-R). Interestingly, Ox-LDL-induced LDL-R expression was further enhanced by Ginkgolide B treatment in HUVECs. Moreover, Ginkgolide B treatment lead to downregulation of lectin-like Ox-LDL receptor (LOX-1) and NADPH oxidase (NOX-4) expression which was upregulated in Ox-LDL-treated HUVECs, along with the attenuation of mitochondrial ROS generation. Furthermore, Ginkgolide B significantly inhibited the augmented expression of intercellular adhesion molecule-1 (ICAM-1) and vascular adhesion molecule-1 (VCAM-1) in Ox-LDL-activated HUVECs. Ginkgolide B also significantly ameliorated the inflammatory response in Ox-LDL-activated HUVECs by suppressing the expression of IL-1α, IL-1β, IL-6, CXCL-1, CXCL-2, and monocyte chemotactic protein (MCP-1), at mRNA and protein level. Our in vitro and in silico study established that Ginkgolide B alleviated the Ox-LDL-induced inflammatory cascades and altered lipid metabolism in HUVECs by suppressing the PCSK-9 and, thus, could be established as a treasured alternative therapeutic candidate in the atherosclerosis management.
Collapse
|
37
|
Dimou P, Wright RD, Budge KL, Midgley A, Satchell SC, Peak M, Beresford MW. The human glomerular endothelial cells are potent pro-inflammatory contributors in an in vitro model of lupus nephritis. Sci Rep 2019; 9:8348. [PMID: 31171837 PMCID: PMC6554346 DOI: 10.1038/s41598-019-44868-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 05/24/2019] [Indexed: 02/02/2023] Open
Abstract
Juvenile-onset lupus nephritis (LN) affects up to 80% of juvenile-onset systemic lupus erythematosus patients (JSLE). As the exact role of human renal glomerular endothelial cells (GEnCs) in LN has not been fully elucidated, the aim of this study was to investigate their involvement in LN. Conditionally immortalised human GEnCs (ciGEnCs) were treated with pro-inflammatory cytokines known to be involved in LN pathogenesis and also with LPS. Secretion and surface expression of pro-inflammatory proteins was quantified via ELISA and flow cytometry. NF-κΒ and STAT-1 activation was investigated via immunofluorescence. Serum samples from JSLE patients and from healthy controls were used to treat ciGEnCs to determine via qRT-PCR potential changes in the mRNA levels of pro-inflammatory genes. Our results identified TNF-α, IL-1β, IL-13, IFN-γ and LPS as robust in vitro stimuli of ciGEnCs. Each of them led to significantly increased production of different pro-inflammatory proteins, including; IL-6, IL-10, MCP-1, sVCAM-1, MIP-1α, IP-10, GM-CSF, M-CSF, TNF-α, IFN-γ, VCAM-1, ICAM-1, PD-L1 and ICOS-L. TNF-α and IL-1β were shown to activate NF-κB, whilst IFN-γ activated STAT-1. JSLE patient serum promoted IL-6 and IL-1β mRNA expression. In conclusion, our in vitro model provides evidence that human GEnCs play a pivotal role in LN-associated inflammatory process.
Collapse
Affiliation(s)
- Paraskevi Dimou
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Rachael D Wright
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Kelly L Budge
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Angela Midgley
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | | | - Matthew Peak
- NIHR Alder Hey Clinical Research Facility, Alder Hey Children's NHS Foundation Trust, Liverpool, UK
| | - Michael W Beresford
- Department of Women's and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK. .,NIHR Alder Hey Clinical Research Facility, Alder Hey Children's NHS Foundation Trust, Liverpool, UK. .,Department of Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK.
| |
Collapse
|
38
|
Schröder S, Juerß D, Kriesen S, Manda K, Hildebrandt G. Immunomodulatory properties of low-dose ionizing radiation on human endothelial cells. Int J Radiat Biol 2018; 95:23-32. [DOI: 10.1080/09553002.2018.1486515] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Affiliation(s)
- Sabine Schröder
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Rostock, Germany
| | - Dajana Juerß
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Rostock, Germany
| | - Stephan Kriesen
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Rostock, Germany
| | - Katrin Manda
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Rostock, Germany
| | - Guido Hildebrandt
- Department of Radiotherapy and Radiation Oncology, University of Rostock, Rostock, Germany
| |
Collapse
|
39
|
Pich C, Meylan P, Mastelic-Gavillet B, Nguyen TN, Loyon R, Trang BK, Moser H, Moret C, Goepfert C, Hafner J, Levesque MP, Romero P, Jandus C, Michalik L. Induction of Paracrine Signaling in Metastatic Melanoma Cells by PPARγ Agonist Rosiglitazone Activates Stromal Cells and Enhances Tumor Growth. Cancer Res 2018; 78:6447-6461. [PMID: 30185551 DOI: 10.1158/0008-5472.can-18-0912] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2018] [Revised: 07/27/2018] [Accepted: 08/28/2018] [Indexed: 01/10/2023]
Abstract
In addition to improving insulin sensitivity in type 2 diabetes, the thiazolidinedione family of compounds and the pharmacologic activation of their best-characterized target PPARγ have been proposed as a therapeutic option for cancer treatment. In this study, we reveal a new mode of action for the thiazolidinedione rosiglitazone that can contribute to tumorigenesis. Rosiglitazone activated a tumorigenic paracrine communication program in a subset of human melanoma cells that involves the secretion of cytokines, chemokines, and angiogenic factors. This complex blend of paracrine signals activated nonmalignant fibroblasts, endothelial cells, and macrophages in a tumor-friendly way. In agreement with these data, rosiglitazone promoted human melanoma development in xenografts, and tumors exposed to rosiglitazone exhibited enhanced angiogenesis and inflammation. Together, these findings establish an important tumorigenic action of rosiglitazone in a subset of melanoma cells. Although studies conducted on cohorts of diabetic patients report overall benefits of thiazolidinediones in cancer prevention, our data suggest that exposure of established tumors to rosiglitazone may be deleterious.Significance: These findings uncover a novel mechanism by which the thiazolidinedione compound rosiglitazone contributes to tumorigenesis, thus highlighting a potential risk associated with its use in patients with established tumors. Cancer Res; 78(22); 6447-61. ©2018 AACR.
Collapse
Affiliation(s)
- Christine Pich
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Patrick Meylan
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Beatris Mastelic-Gavillet
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Thanh Nhan Nguyen
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Romain Loyon
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Bao Khanh Trang
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Hélène Moser
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Catherine Moret
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Christine Goepfert
- COMPATH, Institute of Animal Pathology, University of Bern, Bern, Switzerland
| | - Jürg Hafner
- Department of Dermatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Mitchell P Levesque
- Department of Dermatology, University Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Pedro Romero
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Camilla Jandus
- Department of Oncology, University of Lausanne, Ludwig Cancer Research Center, Lausanne, Switzerland
| | - Liliane Michalik
- Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland.
| |
Collapse
|
40
|
Interaction with an endothelial lumen increases neutrophil lifetime and motility in response to P aeruginosa. Blood 2018; 132:1818-1828. [PMID: 30143504 DOI: 10.1182/blood-2018-05-848465] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/14/2018] [Indexed: 12/12/2022] Open
Abstract
Neutrophil infiltration into tissues is essential for host defense and pathogen clearance. Although many of the signaling pathways involved in the transendothelial migration of neutrophils are known, the role of the endothelium in regulating neutrophil behavior in response to infection within interstitial tissues remains unclear. Here we developed a microscale 3-dimensional (3D) model that incorporates an endothelial lumen, a 3D extracellular matrix, and an intact bacterial source to model the host microenvironment. Using this system, we show that an endothelial lumen significantly increased neutrophil migration toward a source of Pseudomonas aeruginosa Surprisingly, we found neutrophils, which were thought to be short-lived cells in vitro, migrate for up to 24 hours in 3D in the presence of an endothelial lumen and bacteria. In addition, we found that endothelial cells secrete inflammatory mediators induced by the presence of P aeruginosa, including granulocyte-macrophage colony-stimulating factor (GM-CSF), a known promoter of neutrophil survival, and interleukin (IL)-6, a proinflammatory cytokine. We found that pretreatment of neutrophils with a blocking antibody against the IL-6 receptor significantly reduced neutrophil migration to P aeruginosa but did not alter neutrophil lifetime, indicating that secreted IL-6 is an important signal between endothelial cells and neutrophils that mediates migration. Taken together, these findings demonstrate an important role for endothelial paracrine signaling in neutrophil migration and survival.
Collapse
|
41
|
Ma CY, Xu ZY, Wang SP, Peng HY, Liu F, Liu JH, Ren FX. Change of Inflammatory Factors in Patients with Acute Coronary Syndrome. Chin Med J (Engl) 2018; 131:1444-1449. [PMID: 29893361 PMCID: PMC6006811 DOI: 10.4103/0366-6999.233953] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Acute coronary syndrome (ACS) is closely related to unstable plaques and secondary thrombosis. The inflammatory cells in plaques and their inflammatory products may be the cause for plaque instability and ruptures. The study aimed to disclose the changes of inflammatory factors including serum intracellular adhesion molecule-1 (ICAM-1), chitinase-3-like protein 1 (YKL-40), and lipoprotein-associated phospholipase A2 (Lp-PLA2) in patients with ACS and its clinical significance. METHODS A total of 120 patients with coronary heart disease (CHD) were categorized into 2 groups: 69 with ACS and 51 with stable angina pectoris (SAP); 20 patients with chest pain and normal angiography served as a control group. The 120 patients with CHD were categorized into single-vessel disease group, double-vessel disease group, and three-vessel disease group based on the number of coronary artery stenosis. The severity of coronary artery stenosis was quantified based on coronary angiography using Gensini score. They were further divided into mild CHD group with its Gensini score <26 (n = 36), moderate CHD group with its Gensini score being 26-54 (n = 48) and severe CHD group with its Gensini score >54 (n = 36). Serum levels of ICAM-1, YKL-40, and Lp-PLA2 of different groups were determined by enzyme-linked immunosorbent assay. Correlation between ICAM-1, YKL-40, Lp-PLA2, and Gensini score was analyzed. RESULTS The levels of serum inflammatory factors ICAM-1, YKL-40, and Lp-PLA2 were significantly higher in the ACS group than those in control group and SAP group (all P < 0.05); and compared with control group, no significant difference was observed in terms of the serum ICAM-1, YKL-40, and Lp-PLA2 levels in the SAP group (P > 0.05).The levels of serum ICAM-1, YKL-40, and Lp-PLA2 were not significantly different among control group, single-vessel disease group, double-vessel disease group, and three-vessel disease group (all P > 0.05). The levels of serum ICAM-1, YKL-40, and Lp-PLA2 were not significantly different among control group, mild CHD group (Gensini score <26), moderate CHD group (Gensini score 26-54), and severe CHD group (Gensini score >54) (all P > 0.05). Nonparametric Spearman correlation analysis showed that the levels of serum ICAM-1, YKL-40, and Lp-PLA2 were not correlated with the Gensini score in CHD patients (r = 0.093, r = -0.149, and r = -0.085, all P > 0.05; respectively). CONCLUSIONS The serum levels of ICAM-1, YKL-40, and Lp-PLA2 were correlated with different clinical types of CHD, but not well correlated the severity and extent of artery stenosis, suggesting that ICAM-1, YKL-40, and Lp-PLA2 might be involved in occurrence of instability of atherosclerotic plaque, and might reflect the severity of CHD mostly through reflecting the plaque stability.
Collapse
Affiliation(s)
- Cai-Yun Ma
- Department of Cardiology, Aviation General Hospital, Beijing 100016, China
| | - Zhen-Ye Xu
- Department of Cardiology, Beijing Daxing Hospital, Capital Medical University, Beijing 102600, China
| | - Shao-Ping Wang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Hong-Yu Peng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Fang Liu
- Department of Cardiology, Aviation General Hospital, Beijing 100016, China
| | - Jing-Hua Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing 100029, China
| | - Feng-Xue Ren
- Department of Cardiology, Aviation General Hospital, Beijing 100016, China
| |
Collapse
|
42
|
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
|
43
|
Modulation of Inflammatory Reactions by Low-Dose Ionizing Radiation: Cytokine Release of Murine Endothelial Cells Is Dependent on Culture Conditions. J Immunol Res 2018; 2018:2856518. [PMID: 29967799 PMCID: PMC6008836 DOI: 10.1155/2018/2856518] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/14/2018] [Accepted: 04/05/2018] [Indexed: 01/04/2023] Open
Abstract
Background In many European countries, patients with a variety of chronical inflammatory diseases are treated with low-dose radiotherapy (LD-RT). In contrast to high-dose irradiation given to tumor patients, little is known about radiobiological mechanisms underlying this clinical successful LD-RT application. The objective of this study was to gain a better insight into the modulation of inflammatory reactions after LD-RT on the basis of endothelial cells (EC) as major participants and regulators of inflammation. Methods Three murine EC lines were cultivated under 2D and 3D culture conditions and irradiated with doses from 0.01 Gy to 2 Gy. To simulate an inflammatory situation, cells were activated with TNF-α. After LD-RT, a screening of numerous inflammatory markers was determined by multiplex assay, followed by detailed analyses of four cytokines (KC, MCP-1, RANTES, and G-CSF). Additionally, the monocyte binding to EC was analyzed. Results Cytokine concentrations were dependent on culture condition, IR dose, time point after IR, and EC origin. IR caused nonlinear dose-dependent effects on secretion of the proinflammatory cytokines KC, MCP-1, and RANTES. The monocyte adhesion was significantly enhanced after IR as well as activation. Conclusions The study shows that LD-RT, also using very low radiation doses, has a clear immunomodulatory effect on EC as major participants and regulators of inflammation.
Collapse
|
44
|
Thakkar V, Patterson KA, Stevens W, Wilson M, Roddy J, Sahhar J, Proudman S, Hissaria P, Nikpour M. Increased serum levels of adhesion molecules ICAM-1 and VCAM-1 in systemic sclerosis are not specific for pulmonary manifestations. Clin Rheumatol 2018; 37:1563-1571. [PMID: 29687288 DOI: 10.1007/s10067-018-4081-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/15/2018] [Accepted: 03/23/2018] [Indexed: 01/15/2023]
Abstract
Studies suggest elevated serum intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) levels may be markers of pulmonary arterial hypertension in systemic sclerosis (SSc-PAH). We sought to evaluate whether ICAM-1 and VCAM-1 levels are useful screening biomarkers for incident SSc-PAH. In this cross-sectional study, four groups were selected from the Australian Scleroderma Cohort Study: group 1 (n = 15) had definite PAH; group 2 (n = 19) had interstitial lung disease (ILD); group 3 (n = 30) were SSc-controls; and group 4 (n = 34) were healthy controls. Serum ICAM-1 and VCAM-1 levels were measured using the Millipore Milliplex MAP Human 2-Plex Panel. There were no differences in ICAM-1 levels in the PAH versus ILD group (263.0 ± 85.4 vs 380.4 ± 168.3 ng/mL, p = 0.136), SSc-controls (263.0 ± 85.4 vs 253.1 ± 98.0 ng/mL, p = 1.00), or healthy controls (263.0 ± 85.4 vs 201.8 ± 57.2 ng/mL, p = 0.093). Similarly, there were no differences in VCAM-1 level in PAH versus ILD groups (1476.2 ± 434.9 vs 1424.8 ± 527.6 ng/mL, p = 1.00) and SSc-controls (1476.2 ± 434.9 vs 1409.5 ± 341.1 ng/mL, p = 1.00). SSc subjects had significantly higher levels of ICAM-1 (297.4 ± 134.0 vs 201.8 ± 57.2 ng/mL, p < 0.0001) and VCAM-1 compared to healthy controls (1432.7 ± 427.4 vs 1125.6 ± 273.4 ng/mL, p < 0.0001). Neither ICAM-1 nor VCAM-1 is a specific screening biomarker of SSc-PAH. Instead, increased levels of these adhesion molecules in SSc, irrespective of pulmonary complications, suggest that they may play a role in SSc pathogenesis.
Collapse
Affiliation(s)
- Vivek Thakkar
- Department of Rheumatology, Liverpool Hospital, Liverpool BC, NSW, 2170, Australia.,School of Medicine, Western Sydney University, Campbelltown, NSW, Australia.,South Western Clinical School, University of New South Wales, Liverpool, Australia.,Faculty of Medicine and Health Sciences, Macquarie University, Sydney, Australia.,Department of Rheumatology, St Vincent's Hospital (Melbourne), 41 Parade, Fitzroy, VIC, 3065, Australia.,Department of Medicine, The University of Melbourne at St Vincent's Hospital, 41 Parade, Fitzroy, VIC, 3065, Australia
| | - Karen A Patterson
- Flinders University, Bedford Park, South Australia, Australia.,Commonwealth Scientific and Industrial Research Organisation CSIRO, Adelaide, South Australia, Australia
| | - Wendy Stevens
- Department of Rheumatology, St Vincent's Hospital (Melbourne), 41 Parade, Fitzroy, VIC, 3065, Australia
| | - Michelle Wilson
- Department of Rheumatology, St Vincent's Hospital (Melbourne), 41 Parade, Fitzroy, VIC, 3065, Australia
| | - Janet Roddy
- Department of Rheumatology, Royal Perth Hospital, GPO Box X2213, Perth, WA, 6001, Australia
| | - Joanne Sahhar
- Department of Rheumatology, Monash Health & Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia.,Department of Medicine, Monash Health & Monash University, 246 Clayton Road, Clayton, VIC, 3168, Australia
| | - Susanna Proudman
- Rheumatology Unit, Royal Adelaide Hospital, Port Road, Adelaide, SA, 5000, Australia.,Discipline of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Pravin Hissaria
- Discipline of Medicine, University of Adelaide, Adelaide, SA, 5000, Australia.,Department of Clinical Immunology, Royal Adelaide Hospital, North Terrace, Adelaide, SA, 5000, Australia
| | - Mandana Nikpour
- Department of Rheumatology, St Vincent's Hospital (Melbourne), 41 Parade, Fitzroy, VIC, 3065, Australia. .,Department of Medicine, The University of Melbourne at St Vincent's Hospital, 41 Parade, Fitzroy, VIC, 3065, Australia.
| |
Collapse
|
45
|
Kolliker Frers RA, Cosentino V, Tau J, Kerzberg EM, Urdapilleta A, Chiocconi M, Kogan N, Otero-Losada M, Capani F. Immune-Mediated Inflammation Promotes Subclinical Atherosclerosis in Recent-Onset Psoriatic Arthritis Patients without Conventional Cardiovascular Risk Factors. Front Immunol 2018. [PMID: 29535705 PMCID: PMC5834432 DOI: 10.3389/fimmu.2018.00139] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Studies on the inflammatory burden in recent-onset psoriatic arthritis (PsA) patients without conventional cardiovascular risk factors (CVRFs) are not available. This preliminary study focuses on cardiovascular risk in cutaneous psoriasis (CPs) and recent-onset PsA patients. Blood biochemistry (glucose, cholesterol, uric acid, lipid profile and apolipoprotein B) was analyzed using standard kits. Proatherogenic inflammation markers, C-reactive protein (CRP) and interleukin-6 (IL-6), and endothelial activators monocyte chemoattractant protein-1 (MCP-1) and soluble intercellular adhesion molecule-1 (sICAM-1), were determined by enzyme-linked immunosorbent assay. Ultrasound images allowed measuring carotid intima-media thickness (cIMT). Our study first shows an increase in cIMT, and in serum levels of sICAM-1 and CRP in recent-onset PsA patients not presenting conventional CVRFs over the non-medicated time-period, from disease diagnosis to the beginning of pharmacological treatment, compared with healthy subjects. The outcome highlights the importance of monitoring serum level of sICAM1, CRP, and cIMT, and the value of primary prevention in psoriatic patients even with no history of cardiovascular events.
Collapse
Affiliation(s)
- Rodolfo A Kolliker Frers
- Laboratorio de Citoarquitectura y Plasticidad Neuronal (LCPN), Instituto de Investigaciones Cardiológicas, ININCA-UBA-CONICET, Buenos Aires, Argentina.,Unidad de Reumatología, Hospital JM Ramos Mejía, Buenos Aires, Argentina
| | - Vanesa Cosentino
- Unidad de Reumatología, Hospital JM Ramos Mejía, Buenos Aires, Argentina
| | - Julia Tau
- Laboratorio de Investigación Ocular, Departamento de Patología, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Eduardo M Kerzberg
- Unidad de Reumatología, Hospital JM Ramos Mejía, Buenos Aires, Argentina
| | - Adriana Urdapilleta
- Departamento de Radiología, Hospital JM Ramos Mejía, Buenos Aires, Argentina
| | - Monica Chiocconi
- Laboratorio de determinaciones hormonales, Sección de metabolismo de lípidos e hidratos de carbono, División Endocrinología, Departamento de Diagnóstico y Tratamiento, Hospital JM Ramos Mejía, Buenos Aires, Argentina
| | - Nora Kogan
- Sección Psoriasis, División Dermatología, Departamento de Clínica, Hospital JM Ramos Mejía, Buenos Aires, Argentina
| | - Matilde Otero-Losada
- Laboratorio de HPLC, Instituto de Investigaciones Cardiológicas, University of Buenos Aires, National Research Council, ININCA-UBA-CONICET, Buenos Aires, Argentina
| | - Francisco Capani
- Laboratorio de Citoarquitectura y Plasticidad Neuronal (LCPN), Instituto de Investigaciones Cardiológicas, ININCA-UBA-CONICET, Buenos Aires, Argentina.,Departamento de Biología, Universidad John F Kennedy, Buenos Aires, Argentina.,Universidad Autónoma de Chile, Santiago, Chile
| |
Collapse
|
46
|
Trommer S, Leimert A, Bucher M, Schumann J. Impact of Unsaturated Fatty Acids on Cytokine-Driven Endothelial Cell Dysfunction. Int J Mol Sci 2017; 18:ijms18122739. [PMID: 29258201 PMCID: PMC5751340 DOI: 10.3390/ijms18122739] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2017] [Revised: 12/08/2017] [Accepted: 12/15/2017] [Indexed: 12/21/2022] Open
Abstract
Polyunsaturated fatty acids (PUFA) are reported to exert prophylactic and acute therapeutic effects in diseases linked to endothelial dysfunction. In the present study, the consequences of a PUFA enrichment of endothelial cells (cell line TIME) on cell viability, expression of the cytokines interleukin-6 (IL-6), interleukin-8 (IL-8), granulocyte-macrophage colony-stimulating factor (GM-CSF), and monocyte chemoattractant protein 1 (MCP-1), synthesis of the adhesion molecules intercellular adhesion molecule 1 (ICAM-1) and vascular adhesion molecule 1 (VCAM-1), and production of the coagulation factors plasminogen activator inhibitor-1 (PAI-1), von Willebrand factor (vWF), and tissue factor (TF) was analyzed in parallel. PUFA of both the n3 and the n6 family were investigated in a physiologically relevant concentration of 15 µM, and experiments were performed in both the presence and the absence of the pro-inflammatory cytokines interleukin-1β (IL-1β), tumor necrosis factor-α (TNF-α), and interferon-γ (IFN-γ). Supplementation of the culture medium with particular fatty acids was found to have a promoting effect on cellular production of the cytokines IL-6, IL-8, GM-CSF, and MCP-1. Further on, PUFA treatment in the absence of a stimulant diminished the percentage of endothelial cells positive for ICAM-1, and adversely affected the stimulation-induced upregulation of VCAM-1. Cell viability and production of coagulation factors were not or only marginally affected by supplemented fatty acids. Altogether, the data indicate that PUFA of either family are only partially able to counterbalance the destructive consequences of an endothelial dysfunction.
Collapse
Affiliation(s)
- Simon Trommer
- Clinic for Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), 06120 Halle (Saale), Germany.
| | - Anja Leimert
- Clinic for Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), 06120 Halle (Saale), Germany.
| | - Michael Bucher
- Clinic for Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), 06120 Halle (Saale), Germany.
| | - Julia Schumann
- Clinic for Anesthesiology and Surgical Intensive Care, University Hospital Halle (Saale), 06120 Halle (Saale), Germany.
| |
Collapse
|
47
|
Chang MC, Lee JJ, Chen YJ, Lin SI, Lin LD, Jein-Wen Liou E, Huang WL, Chan CP, Huang CC, Jeng JH. Lysophosphatidylcholine induces cytotoxicity/apoptosis and IL-8 production of human endothelial cells: Related mechanisms. Oncotarget 2017; 8:106177-106189. [PMID: 29290940 PMCID: PMC5739725 DOI: 10.18632/oncotarget.22425] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 10/29/2017] [Indexed: 12/31/2022] Open
Abstract
Increased levels of oxidized low-density lipoprotein oxLDL) are shown to elevate the risk of cardiovascular diseases such as atherosclerosis, thrombosis, stroke, and myocardial infarction. This is possibly due to the toxic effects of oxLDLs on vascular cells. Various oxLDLs including lysophosphatidylcholine (LPC) and 7-ketocholesterol injure vascular endothelial cells and stimulate inflammatory reaction. However the toxicity of LPC on endothelial cells is not clear. In this study, human endothelial cells were exposed to LPC. Cytotoxicity was measured by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide assay. Propidium iodide (PI) staining or PI/Annexin V dual staining flow cytometry were used to determine cell cycle progression and apoptosis. Reactive oxygen species (ROS) level was analyzed by DCFH-DA labeling flow cytometry. RNA and protein expression of endothelial cells was studied by reverse transcriptase-polymerase chain reaction and western blotting. IL-8 secretion was measured by enzyme-linked immunosorbant assay. LPC showed cytotoxicity to endothelial cells (>50 µg/ml). LPC induced cell cycle arrest and apoptosis with concomitant inhibition of cdc2 and cyclin B1 expression. LPC stimulated intracellular ROS production and ATM/Chk2, ATR/Chk1 and Akt activation. IL-8 expression and secretion in endothelial cells were induced by LPC. LPC-induced apoptosis, and IL-8 expression/secretion was attenuated by LY294002, a PI3K/Akt inhibitor. These results reveal that LPC is involved in the pathogenesis of atherosclerosis and vascular diseases by stimulation of inflammation and injury to endothelial cells. These events are related to ROS, ATM/Chk2, ATR/Chk2 and PI3K/Akt signaling. Understanding the toxic mechanisms of LPC is useful for future prevention and treatment atherosclerosis.
Collapse
Affiliation(s)
- Mei-Chi Chang
- Biomedical Science Team, Chang Gung University of Science and Technology, Kwei-Shan, Taoyuan City, Taiwan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Jang-Jaer Lee
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Yi-Jane Chen
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | - Szu-I Lin
- Department of Dentistry, Municipal Taoyuan Hospital, Taoyuan City, Taiwan
| | - Li-Deh Lin
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| | | | - Wei-Ling Huang
- Department of Dentistry, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | - Chiu-Po Chan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Chi-Chia Huang
- Department of Dentistry, Cardinal Tien Hospital, New Taipei City, Taiwan
| | - Jiiang-Huei Jeng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
48
|
Hirano Y, Yang WL, Aziz M, Zhang F, Sherry B, Wang P. MFG-E8-derived peptide attenuates adhesion and migration of immune cells to endothelial cells. J Leukoc Biol 2017; 101:1201-1209. [PMID: 28096298 DOI: 10.1189/jlb.3a0416-184rr] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Revised: 12/20/2016] [Accepted: 12/28/2016] [Indexed: 02/05/2023] Open
Abstract
Milk fat globule-epidermal growth factor-factor 8 (MFG-E8) plays an immunomodulatory role in inflammatory diseases. MFG-E8-derived short peptide (MSP68) greatly reduces neutrophil infiltration and injury in the lung during sepsis. In this study, we examined the effect of MSP68 on chemotaxis of various immune cells and its regulatory mechanism. Bone marrow-derived neutrophils (BMDNs) from C57BL/6 mice, human monocyte THP-1 cell line, and human T lymphocyte Jurkat cell line were used for adhesion and migration assays using a Transwell method in the presence of MSP68. Treatment with MSP68 significantly inhibited the BMDN and THP-1 cell but not Jurkat cell adhesion on the TNF-α-stimulated pulmonary artery endothelial cell (PAEC) monolayer dose-dependently. MSP68 also significantly reduced BMDN adhesion on VCAM-1-coated wells dose dependently. Surface plasmon resonance (SPR) analysis revealed that MSP68 efficiently recognized integrin α4β1 (receptor for VCAM-1) at the dissociation constant (KD) of 1.53 × 10-7 M. These findings implicate that MSP68 prevents neutrophil adhesion to the activated endothelial cells by interfering with the binding between integrin α4β1 on neutrophils and VCAM-1 on endothelial cells. Moreover, MSP68 significantly attenuated the migration of BMDN and THP-1 cells but not Jurkat cells to their chemoattractants. Pretreatment with MSP68 inhibited the transmigration of BMDNs across the PAECs toward chemoattractants, fMLP, MIP-2, and complement fragment 5a (C5a) dose-dependently. Finally, we identified that the activation of p38 MAPK in BMDNs by fMLP was inhibited by MSP68. Thus, MSP68 attenuates extravasation of immune cells through the endothelial cell lining into inflamed tissue, implicating MSP68 to be a novel, therapeutic agent for inflammatory diseases caused by excessive immune cell infiltration.
Collapse
Affiliation(s)
- Yohei Hirano
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Emergency and Critical Care Medicine, Juntendo University and Urayasu Hospital, Chiba, Japan; and
| | - Weng-Lang Yang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA.,Department of Surgery, Hofstra Northwell School of Medicine, Manhasset, New York, USA
| | - Monowar Aziz
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Fangming Zhang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Barbara Sherry
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA
| | - Ping Wang
- Center for Immunology and Inflammation, The Feinstein Institute for Medical Research, Manhasset, New York, USA; .,Department of Surgery, Hofstra Northwell School of Medicine, Manhasset, New York, USA
| |
Collapse
|
49
|
Chang MC, Chen YJ, Liou EJW, Tseng WY, Chan CP, Lin HJ, Liao WC, Chang YC, Jeng PY, Jeng JH. 7-Ketocholesterol induces ATM/ATR, Chk1/Chk2, PI3K/Akt signalings, cytotoxicity and IL-8 production in endothelial cells. Oncotarget 2016; 7:74473-74483. [PMID: 27740938 PMCID: PMC5342680 DOI: 10.18632/oncotarget.12578] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 10/06/2016] [Indexed: 01/01/2023] Open
Abstract
Cardiovascular diseases (atherosclerosis, stroke, myocardiac infarction etc.) are the major systemic diseases of elder peoples in the world. This is possibly due to increased levels of oxidized low-density lipoproteins (oxLDLs) such as 7-ketocholesterol (7-KC) and lysophosphatidylcholine (LPC) that damage vascular endothelial cells, induce inflammatory responses, to elevate the risk of cardiovascular diseases, Alzheimer's disease, and age-related macular degeneration. However the toxic effects of 7-KC on endothelial cells are not known. In this study, 7-KC showed cytotoxicity to endothelial cells at concentrations higher than 10 µg/ml. 7-KC stimulated ATM/Chk2, ATR-Chk1 and p53 signaling pathways in endothelial cells. 7-KC also induced G0/G1 cell cycle arrest and apoptosis with an inhibition of Cyclin dependent kinase 1 (Cdk1) and cyclin B1 expression. Secretion and expression of IL-8 in endothelial cells were stimulated by 7-KC. 7-KC further induced intracellular ROS production as shown by increase in DCF fluorescence and Akt phosphorylation. LY294002 attenuated the 7-KC-induced apoptosis and IL-8 mRNA expression of endothelial cells. These results indicate that oxLDLs such as 7-KC may contribute to the pathogenesis of atherosclerosis, thrombosis and cardiovascular diseases by induction of endothelial damage, apoptosis and inflammatory responses. These events are associated with ROS production, activation of ATM/Chk2, ATR/Chk1, p53 and PI3K/Akt signaling pathways.
Collapse
Affiliation(s)
- Mei-Chi Chang
- Biomedical Science Team, Chang Gung University of Science and Technology, Kwei-Shan, Taoyuan, Taiwan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Yi-Jane Chen
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital
| | | | - Wan-Yu Tseng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital
| | - Chiu-Po Chan
- Department of Dentistry, Chang Gung Memorial Hospital, Taipei, Taiwan
| | - Hseuh-Jen Lin
- Department of Dentistry, Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Wan-Chuen Liao
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital
| | - Ya-Ching Chang
- Department of Dentistry, Mackey Memorial Hospital, Taipei, Taiwan
| | - Po-Yuan Jeng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital
| | - Jiiang-Huei Jeng
- School of Dentistry and Department of Dentistry, National Taiwan University Medical College and National Taiwan University Hospital
| |
Collapse
|
50
|
Yang X, Li M, Haghiac M, Catalano PM, O'Tierney-Ginn P, Hauguel-de Mouzon S. Causal relationship between obesity-related traits and TLR4-driven responses at the maternal-fetal interface. Diabetologia 2016; 59:2459-2466. [PMID: 27535280 PMCID: PMC5583648 DOI: 10.1007/s00125-016-4073-6] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/18/2016] [Indexed: 12/21/2022]
Abstract
AIMS/HYPOTHESIS Obesity triggers complex inflammatory networks within the innate immune system. During pregnancy, the placenta amplifies the low-grade inflammation through activation of Toll-like receptor 4 (TLR4) signalling pathways. The purpose of this study was to investigate the impact of obesity on placental TLR4 expression and inflammatory signals. The secondary aim was to analyse the placental cell type responsible for TLR4 activation. METHODS Thirty-nine women recruited at term-scheduled Caesarean section were grouped according to their pre-gravid BMI (<25 kg/m(2) and >30 kg/m(2)). Placenta, venous maternal and cord blood were obtained at delivery for analysis. Data were analysed with linear regression and Spearman's rank correlation coefficient analysis. RESULTS TLR4, IL6 and IL8 expression was increased three- to ninefold (p < 0.001) in the placenta of obese vs lean women. There was a positive correlation between placental TLR4 and maternal systemic and placental IL6 and IL8 concentrations. Placental TLR4 expression was correlated with maternal pre-gravid BMI, insulin resistance index, plasma insulin and C-reactive protein (r = 0.57, 0.31, 0.35, 0.53, respectively; p < 0.001) but not with plasma glucose, maternal age, gestational age and gestational weight gain (r < 0.2; p > 0.1). TLR4 was located in both trophoblast and macrovascular endothelial cells lining fetal vasculature. Lipopolysaccharide-induced TLR4 activation was more robust in trophoblasts than in endothelial vascular cells (100-fold vs tenfold; p < 0.001). CONCLUSIONS/INTERPRETATION Trophoblastic TLR4 is strongly implicated in the propagation of placental inflammation. Placental inflammation is related to maternal metabolic conditions such as pre-gravid BMI, whilst gestational weight gain or gestational age are not. These results implicate the pre-gravid condition as a significant contributor to metabolic inflammation in late pregnancy.
Collapse
Affiliation(s)
- Xiaohua Yang
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Ming Li
- Epidemiology and Biostatistics Department, School of Medicine, Case Western Reserve University, Cleveland, USA
| | - Maricela Haghiac
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Patrick M Catalano
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Perrie O'Tierney-Ginn
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA
| | - Sylvie Hauguel-de Mouzon
- Center for Reproductive Health, MetroHealth Medical Center, Case Western Reserve University School of Medicine, 2500 MetroHealth Dr., Cleveland, OH, 44109-1998, USA.
| |
Collapse
|