251
|
Goncharov NV, Nadeev AD, Jenkins RO, Avdonin PV. Markers and Biomarkers of Endothelium: When Something Is Rotten in the State. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:9759735. [PMID: 29333215 PMCID: PMC5733214 DOI: 10.1155/2017/9759735] [Citation(s) in RCA: 132] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Accepted: 09/05/2017] [Indexed: 12/14/2022]
Abstract
Endothelium is a community of endothelial cells (ECs), which line the blood and lymphatic vessels, thus forming an interface between the tissues and the blood or lympha. This strategic position of endothelium infers its indispensable functional role in controlling vasoregulation, haemostasis, and inflammation. The state of endothelium is simultaneously the cause and effect of many diseases, and this is coupled with modifications of endothelial phenotype represented by markers and with biochemical profile of blood represented by biomarkers. In this paper, we briefly review data on the functional role of endothelium, give definitions of endothelial markers and biomarkers, touch on the methodological approaches for revealing biomarkers, present an implicit role of endothelium in some toxicological mechanistic studies, and survey the role of reactive oxygen species (ROS) in modulation of endothelial status.
Collapse
Affiliation(s)
- Nikolay V. Goncharov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology, Saint Petersburg, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint Petersburg, Russia
| | - Alexander D. Nadeev
- Sechenov Institute of Evolutionary Physiology and Biochemistry RAS, Saint Petersburg, Russia
- Institute of Cell Biophysics RAS, Pushchino, Russia
| | - Richard O. Jenkins
- School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | | |
Collapse
|
252
|
Subramanian N, Wu Z, Reister F, Sampaio KL, Frascaroli G, Cicin-Sain L, Mertens T. Naïve T cells are activated by autologous HCMV-infected endothelial cells through NKG2D and can control HCMV transmission in vitro. J Gen Virol 2017; 98:3068-3085. [PMID: 29165229 DOI: 10.1099/jgv.0.000976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Apart from classical antigen-presenting cells (APCs) like dendritic cells and macrophages, there are semiprofessional APCs such as endothelial cells (ECs) and Langerhans' cells. Human cytomegalovirus (HCMV) infects a wide range of cell types including the ECs which are involved in the trafficking and homing of T cells. By investigating the interaction of naïve T cells obtained from HCMV-seronegative umbilical cord blood with autologous HCMV-infected human umbilical vein ECs (HUVECs), we could show that the activation of naïve T cells occurred after 1 day of peripheral blood mononuclear cell (PBMC) exposure to HCMV-infected HUVECs. The percentage of activated T cells increased over time and the activation of naïve T cells was not induced by either autologous uninfected HUVECs or by autologous HCMV-infected fibroblasts. The activation of T cells occurred also when purified T cells were co-cultured with HCMV-infected HUVECs. In addition, in most of the donors only CD8+ T cells were activated, when the purified T cells were exposed to HCMV-infected HUVECs. The activation of naïve T cells was inhibited when the NKG2D receptor was blocked on the surface of T cells and among the different NKG2D ligands, we identified two ligands (ULBP4 and MICA) on HCMV-infected HUVECs which might be the interaction partners of the NKG2D receptor. Using a functional cell culture assay, we could show that these activated naïve T cells specifically inhibited HCMV transmission. Altogether, we identified a novel specific activation mechanism of naïve T cells from the umbilical cord by HCMV-infected autologous HUVECs through interaction with NKG2D.
Collapse
Affiliation(s)
| | - Zeguang Wu
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| | - Frank Reister
- Gynecology and Obstetrics Clinics, Ulm University Hospital, Ulm, Germany
| | | | - Giada Frascaroli
- Institute of Virology, Ulm University Medical Center, Ulm, Germany.,Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Luka Cicin-Sain
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Institute for Virology, Hannover Medical School, Hannover, Germany.,German Centre for Infection Research (DZIF), Location Hannover-Braunschweig, Germany
| | - Thomas Mertens
- Institute of Virology, Ulm University Medical Center, Ulm, Germany
| |
Collapse
|
253
|
Network pharmacology exploration reveals endothelial inflammation as a common mechanism for stroke and coronary artery disease treatment of Danhong injection. Sci Rep 2017; 7:15427. [PMID: 29133791 PMCID: PMC5684234 DOI: 10.1038/s41598-017-14692-3] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/09/2017] [Indexed: 12/17/2022] Open
Abstract
Although Danhong injection (DHI) is the most widely prescribed Chinese medicine for both stroke and coronary artery disease (CAD), its underlying common molecular mechanisms remain unclear. An integrated network pharmacology and experimental verification approach was used to decipher common pharmacological mechanisms of DHI on stroke and CAD treatment. A compound-target-disease & function-pathway network was constructed and analyzed, indicating that 37 ingredients derived from DH (Salvia miltiorrhiza Bge., Flos Carthami tinctorii and DHI) modulated 68 common targets shared by stroke and CAD. In-depth network analysis results of the top diseases, functions, pathways and upstream regulators implied that a common underlying mechanism linking DHI’s role in stroke and CAD treatment was inflammatory response in the process of atherosclerosis. Experimentally, DHI exerted comprehensive anti-inflammatory effects on LPS, ox-LDL or cholesterol crystal-induced NF-κB, c-jun and p38 activation, as well as IL-1β, TNF-α, and IL-10 secretion in vascular endothelial cells. Ten of 14 predicted ingredients were verified to have significant anti-inflammatory activities on LPS-induced endothelial inflammation. DHI exerts pharmacological efficacies on both stroke and CAD through multi-ingredient, multi-target, multi-function and multi-pathway mode. Anti-endothelial inflammation therapy serves as a common underlying mechanism. This study provides a new understanding of DHI in clinical application on cardiovascular and cerebrovascular diseases.
Collapse
|
254
|
Khaddaj Mallat R, Mathew John C, Kendrick DJ, Braun AP. The vascular endothelium: A regulator of arterial tone and interface for the immune system. Crit Rev Clin Lab Sci 2017; 54:458-470. [PMID: 29084470 DOI: 10.1080/10408363.2017.1394267] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
As the primary interface between the blood and various tissues of the body, the vascular endothelium exhibits a diverse range of roles and activities, all of which contribute to the overall health and function of the cardiovascular system. In this focused review, we discuss several key aspects of endothelial function, how this may be compromised and subsequent consequences. Specifically, we examine the dynamic regulation of arterial contractility and distribution of blood flow through the generation of chemical and electrical signaling events that impinge upon vascular smooth muscle. The endothelium can generate a diverse range of vasoactive compounds and signals, most of which act locally to adjust blood flow in a dynamic fashion to match tissue metabolism. Disruption of these vascular signaling processes (e.g. reduced nitric oxide bioavailability) is typically referred to as endothelial dysfunction, which is a recognized risk factor for cardiovascular disease in patients and occurs early in the development and progression of hypertension, atherosclerosis and tissue ischemia. Endothelial dysfunction is also associated with type-2 Diabetes and aging and increased mechanistic knowledge of the cellular changes contributing to these effects may provide important clues for interventional strategies. The endothelium also serves as the initial site of interaction for immune cells entering tissues in response to damage and acts to facilitate the actions of both the innate and acquired immune systems to interact with the vascular wall. In addition to representing the main cell type responsible for the formation of new blood vessels (i.e. angiogenesis) within the vasculature, the endothelium is also emerging as a source of extracellular vesicle or microparticles for the transport of signaling molecules and other cellular materials to nearby, or remote, sites in the body. The characteristics of released microparticles appear to change with the functional status of the endothelium; thus, these microparticles may represent novel biomarkers of endothelial health and more serious cardiovascular disease.
Collapse
Affiliation(s)
- Rayan Khaddaj Mallat
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Cini Mathew John
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Dylan J Kendrick
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| | - Andrew P Braun
- a Department of Physiology and Pharmacology, Cumming School of Medicine , University of Calgary, and Libin Cardiovascular Institute of Alberta , Calgary , Canada
| |
Collapse
|
255
|
Halper J. Basic Components of Vascular Connective Tissue and Extracellular Matrix. ADVANCES IN PHARMACOLOGY 2017; 81:95-127. [PMID: 29310805 DOI: 10.1016/bs.apha.2017.08.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Though the composition of the three layers constituting the blood vessel wall varies among the different types of blood vessels, and some layers may even be missing in capillaries, certain basic components, and properties are shared by all blood vessels, though each histologically distinct layer contains a unique complement of extracellular components, growth factors and cytokines, and cell types as well. The structure and composition of vessel layers informs and is informed by the function of the particular blood vessel. The adaptation of the composition and the resulting function of the extracellular matrix (ECM) to changes in circulation/blood flow and a variety of other extravascular stimuli can be characterized as remodeling spearheaded by vascular cells. There is a surprising amount of cell traffic among the three layers. It starts with endothelial cell mediated transmigration of inflammatory cells from the bloodstream into the subendothelium, and then into tissue adjoining the blood vessel. Smooth muscle cells and a variety of adventitial cells reside in tunica media and tunica externa, respectively. The latter cells are a mixture of progenitor/stem cells, fibroblasts, myofibroblasts, pericytes, macrophages, and dendritic cells and respond to endothelial injury by transdifferentiation as they travel into the two inner layers, intima and media for corrective mission in the ECM composition. This chapter addresses the role of various vascular cell types and ECM components synthesized by them in maintenance of normal structure and in their contribution to major pathological processes, such as atherosclerosis, organ fibrosis, and diabetic retinopathy.
Collapse
Affiliation(s)
- Jaroslava Halper
- College of Veterinary Medicine and AU/UGA Medical Partnership, The University of Georgia, Athens, GA, United States.
| |
Collapse
|
256
|
Hartlehnert M, Derksen A, Hagenacker T, Kindermann D, Schäfers M, Pawlak M, Kieseier BC, Meyer Zu Horste G. Schwann cells promote post-traumatic nerve inflammation and neuropathic pain through MHC class II. Sci Rep 2017; 7:12518. [PMID: 28970572 PMCID: PMC5624882 DOI: 10.1038/s41598-017-12744-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2016] [Accepted: 09/15/2017] [Indexed: 12/20/2022] Open
Abstract
The activation of T helper cells requires antigens to be exposed on the surface of antigen presenting cells (APCs) via MHC class II (MHC-II) molecules. Expression of MHC-II is generally limited to professional APCs, but other cell types can express MHC-II under inflammatory conditions. However, the importance of these conditional APCs is unknown. We and others have previously shown that Schwann cells are potentially conditional APCs, but the functional relevance of MHC-II expression by Schwann cells has not been studied in vivo. Here, we conditionally deleted the MHC-II β-chain from myelinating Schwann cells in mice and investigated how this influenced post-traumatic intraneural inflammation and neuropathic pain using the chronic constriction injury (CCI) model. We demonstrate that deletion of MHC-II in myelinating Schwann cells reduces thermal hyperalgesia and, to a lesser extent, also diminishes mechanical allodynia in CCI in female mice. This was accompanied by a reduction of intraneural CD4+ T cells and greater preservation of preferentially large-caliber axons. Activation of T helper cells by MHC-II on Schwann cells thus promotes post-traumatic axonal loss and neuropathic pain. Hence, we provide experimental evidence that Schwann cells gain antigen-presenting function in vivo and modulate local immune responses and diseases in the peripheral nerves.
Collapse
Affiliation(s)
- Maike Hartlehnert
- Department of Neurology, University Hospital Münster, Münster, Germany
| | - Angelika Derksen
- Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany
| | - Tim Hagenacker
- Department of Neurology, University of Duisburg-Essen, Essen, Germany
| | - David Kindermann
- Department of Neurology, University of Duisburg-Essen, Essen, Germany
| | - Maria Schäfers
- Department of Neurology, University of Duisburg-Essen, Essen, Germany
| | - Mathias Pawlak
- Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Bernd C Kieseier
- Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany
| | - Gerd Meyer Zu Horste
- Department of Neurology, University Hospital Münster, Münster, Germany. .,Department of Neurology, Heinrich-Heine-University, Medical Faculty, Düsseldorf, Germany.
| |
Collapse
|
257
|
Giovannini S, Tinelli G, Biscetti F, Straface G, Angelini F, Pitocco D, Mucci L, Landolfi R, Flex A. Serum high mobility group box-1 and osteoprotegerin levels are associated with peripheral arterial disease and critical limb ischemia in type 2 diabetic subjects. Cardiovasc Diabetol 2017; 16:99. [PMID: 28789654 PMCID: PMC5549317 DOI: 10.1186/s12933-017-0581-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 07/28/2017] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND High mobility group box-1 (HMGB-1) is a nuclear protein also acting as inflammatory mediator, whilst osteoprotegerin (OPG), member of tumor necrosis factor receptor superfamily, is indicated as marker of vascular calcification. Peripheral artery disease (PAD) and type 2 diabetes (T2D) are clinical conditions characterized by elevated serum inflammatory markers and vascular calcification enhancement. The aim of this study was to investigate the potential role of HMGB-1, OPG and several inflammatory mediators such as C-reactive protein (HsCRP), tumor necrosis factor-alpha and interleukin-6 (IL-6) on the presence and severity of peripheral artery disease in patients with T2D. METHODS In this retrospective observational study, we have analyzed HMGB-1, OPG and inflammatory cytokines serum levels in 1393 type 2 diabetic patients with PAD and without PAD (WPAD). RESULTS HMGB-1 (7.89 ± 15.23 ng/mL), OPG (6.54 ± 7.76 pmol/L), HsCRP (15.6 ± 14.4 mg/L) and IL-6 (56.1 ± 28.6 pg/mL) serum levels were significantly higher in patients with PAD than in those WPAD (3.02 ± 8.12 ng/mL, P ˂ 0.001; 2.98 ± 2.01 pmol/L, P < 0.001; 7.05 ± 4.4 mg/L, P < 0.001; 37.5 ± 20.2 pg/mL, P < 0.001 respectively). Moreover HMGB-1 (P < 0.001), OPG (P < 0.001), HsCRP (P < 0.001) and IL-6 (P < 0.001) serum levels were positively correlated with clinical severity of PAD. HMGB-1 (adjusted OR 12.32; 95% CI 3.56-23.54, P = 0.023) and OPG (adjusted OR 3.53; 95% CI 1.54-6.15, P = 0.019) resulted independent determinants of PAD in patients with T2D after adjusting for the conventional cardiovascular risk factor and established inflammatory mediators. CONCLUSIONS In T2D patients HMGB-1 and OPG serum levels are higher in patients affected by PAD and independently associated with its occurrence and clinical severity.
Collapse
Affiliation(s)
- Silvia Giovannini
- Department of Gerontology and Geriatrics, A. Gemelli Foundation, Catholic University of the Sacred Heart, School of Medicine, Rome, Italy
| | - Giovanni Tinelli
- Department of Vascular Surgery, A. Gemelli Foundation, Catholic University of the Sacred Heart, School of Medicine, Rome, Italy
| | - Federico Biscetti
- Rheumatology and Affine Sciences Institute, A. Gemelli Foundation, Catholic University of the Sacred Heart, School of Medicine, Rome, Italy.,Laboratory of Vascular Biology and Genetics, Catholic University School of Medicine, Rome, Italy
| | - Giuseppe Straface
- Vascular Medicine and Atherothrombosis Laboratory, Department of Experimental Medicine, Sapienza University of Rome, Polo Pontino, Italy
| | - Flavia Angelini
- Laboratory of Vascular Biology and Genetics, Catholic University School of Medicine, Rome, Italy
| | - Dario Pitocco
- Department of Medicine, A. Gemelli Foundation, Catholic University School of Medicine, Rome, Italy
| | - Luciana Mucci
- Laboratory of Vascular Biology and Genetics, Catholic University School of Medicine, Rome, Italy.,Department of Medicine, A. Gemelli Foundation, Catholic University School of Medicine, Rome, Italy
| | - Raffaele Landolfi
- Department of Medicine, A. Gemelli Foundation, Catholic University School of Medicine, Rome, Italy
| | - Andrea Flex
- Laboratory of Vascular Biology and Genetics, Catholic University School of Medicine, Rome, Italy. .,Department of Medicine, A. Gemelli Foundation, Catholic University School of Medicine, Rome, Italy.
| |
Collapse
|
258
|
Dai J, Fang P, Saredy J, Xi H, Ramon C, Yang W, Choi ET, Ji Y, Mao W, Yang X, Wang H. Metabolism-associated danger signal-induced immune response and reverse immune checkpoint-activated CD40 + monocyte differentiation. J Hematol Oncol 2017; 10:141. [PMID: 28738836 PMCID: PMC5525309 DOI: 10.1186/s13045-017-0504-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Accepted: 06/26/2017] [Indexed: 01/16/2023] Open
Abstract
Adaptive immunity is critical for disease progression and modulates T cell (TC) and antigen-presenting cell (APC) functions. Three signals were initially proposed for adaptive immune activation: signal 1 antigen recognition, signal 2 co-stimulation or co-inhibition, and signal 3 cytokine stimulation. In this article, we propose to term signal 2 as an immune checkpoint, which describes interactions of paired molecules leading to stimulation (stimulatory immune checkpoint) or inhibition (inhibitory immune checkpoint) of an immune response. We classify immune checkpoint into two categories: one-way immune checkpoint for forward signaling towards TC only, and two-way immune checkpoint for both forward and reverse signaling towards TC and APC, respectively. Recently, we and others provided evidence suggesting that metabolic risk factors (RF) activate innate and adaptive immunity, involving the induction of immune checkpoint molecules. We summarize these findings and suggest a novel theory, metabolism-associated danger signal (MADS) recognition, by which metabolic RF activate innate and adaptive immunity. We emphasize that MADS activates the reverse immune checkpoint which leads to APC inflammation in innate and adaptive immunity. Our recent evidence is shown that metabolic RF, such as uremic toxin or hyperhomocysteinemia, induced immune checkpoint molecule CD40 expression in monocytes (MC) and elevated serum soluble CD40 ligand (sCD40L) resulting in CD40+ MC differentiation. We propose that CD40+ MC is a novel pro-inflammatory MC subset and a reliable biomarker for chronic kidney disease severity. We summarize that CD40:CD40L immune checkpoint can induce TC and APC activation via forward stimulatory, reverse stimulatory, and TC contact-independent immune checkpoints. Finally, we modeled metabolic RF-induced two-way stimulatory immune checkpoint amplification and discussed potential signaling pathways including AP-1, NF-κB, NFAT, STAT, and DNA methylation and their contribution to systemic and tissue inflammation.
Collapse
Affiliation(s)
- Jin Dai
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian road, Hangzhou, 310006, Zhejiang, China.,Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Pu Fang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Jason Saredy
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hang Xi
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Cueto Ramon
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - William Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Eric T Choi
- Department of Surgery, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, 210029, China
| | - Wei Mao
- Department of Cardiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, 54 Youdian road, Hangzhou, 310006, Zhejiang, China.
| | - Xiaofeng Yang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.,Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA
| | - Hong Wang
- Center for Metabolic Disease Research, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA. .,Department of Pharmacology, Temple University School of Medicine, 3500 N. Broad Street, Philadelphia, PA, 19140, USA.
| |
Collapse
|
259
|
Dunst J, Kamena F, Matuschewski K. Cytokines and Chemokines in Cerebral Malaria Pathogenesis. Front Cell Infect Microbiol 2017; 7:324. [PMID: 28775960 PMCID: PMC5517394 DOI: 10.3389/fcimb.2017.00324] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 07/03/2017] [Indexed: 12/18/2022] Open
Abstract
Cerebral malaria is among the major causes of malaria-associated mortality and effective adjunctive therapeutic strategies are currently lacking. Central pathophysiological processes involved in the development of cerebral malaria include an imbalance of pro- and anti-inflammatory responses to Plasmodium infection, endothelial cell activation, and loss of blood-brain barrier integrity. However, the sequence of events, which initiates these pathophysiological processes as well as the contribution of their complex interplay to the development of cerebral malaria remain incompletely understood. Several cytokines and chemokines have repeatedly been associated with cerebral malaria severity. Increased levels of these inflammatory mediators could account for the sequestration of leukocytes in the cerebral microvasculature present during cerebral malaria, thereby contributing to an amplification of local inflammation and promoting cerebral malaria pathogenesis. Herein, we highlight the current knowledge on the contribution of cytokines and chemokines to the pathogenesis of cerebral malaria with particular emphasis on their roles in endothelial activation and leukocyte recruitment, as well as their implication in the progression to blood-brain barrier permeability and neuroinflammation, in both human cerebral malaria and in the murine experimental cerebral malaria model. A better molecular understanding of these processes could provide the basis for evidence-based development of adjunct therapies and the definition of diagnostic markers of disease progression.
Collapse
Affiliation(s)
- Josefine Dunst
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany.,Institute of Chemistry and Biochemistry, Free UniversityBerlin, Germany.,Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Faustin Kamena
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany.,Institute of Chemistry and Biochemistry, Free UniversityBerlin, Germany.,Molecular Parasitology, Institute of Biology, Humboldt UniversityBerlin, Germany
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection BiologyBerlin, Germany.,Institute of Chemistry and Biochemistry, Free UniversityBerlin, Germany
| |
Collapse
|
260
|
Abstract
Cryptococcus species are encapsulated fungi found in the environment that predominantly cause disease in immunocompromised hosts after inhalation into the lungs. Even with contemporary antifungal regimens, patients with cryptococcosis continue to have high morbidity and mortality rates. The development of more effective therapies may depend on our understanding of the cellular and molecular mechanisms by which the host promotes sterilizing immunity against the fungus. This review will highlight our current knowledge of how Cryptococcus, primarily the species C. neoformans, is sensed by the mammalian host and how subsequent signaling pathways direct the anti-cryptococcal response by effector cells of the innate immune system.
Collapse
Affiliation(s)
- Lena J Heung
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
261
|
Boule LA, Kovacs EJ. Alcohol, aging, and innate immunity. J Leukoc Biol 2017; 102:41-55. [PMID: 28522597 DOI: 10.1189/jlb.4ru1016-450r] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/24/2017] [Accepted: 04/13/2017] [Indexed: 12/15/2022] Open
Abstract
The global population is aging: in 2010, 8% of the population was older than 65 y, and that is expected to double to 16% by 2050. With advanced age comes a heightened prevalence of chronic diseases. Moreover, elderly humans fair worse after acute diseases, namely infection, leading to higher rates of infection-mediated mortality. Advanced age alters many aspects of both the innate and adaptive immune systems, leading to impaired responses to primary infection and poor development of immunologic memory. An often overlooked, yet increasingly common, behavior in older individuals is alcohol consumption. In fact, it has been estimated that >40% of older adults consume alcohol, and evidence reveals that >10% of this group is drinking more than the recommended limit by the National Institute on Alcohol Abuse and Alcoholism. Alcohol consumption, at any level, alters host immune responses, including changes in the number, phenotype, and function of innate and adaptive immune cells. Thus, understanding the effect of alcohol ingestion on the immune system of older individuals, who are already less capable of combating infection, merits further study. However, there is currently almost nothing known about how drinking alters innate immunity in older subjects, despite innate immune cells being critical for host defense, resolution of inflammation, and maintenance of immune homeostasis. Here, we review the effects of aging and alcohol consumption on innate immune cells independently and highlight the few studies that have examined the effects of alcohol ingestion in aged individuals.
Collapse
Affiliation(s)
- Lisbeth A Boule
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery (GITES), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA; .,The Mucosal Inflammation Program (MIP), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA.,The Investigations in Metabolism, Aging, Gender and Exercise (IMAGE) Research Group, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA; and
| | - Elizabeth J Kovacs
- Department of Surgery, Division of GI, Trauma, and Endocrine Surgery (GITES), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA; .,The Mucosal Inflammation Program (MIP), University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA.,The Investigations in Metabolism, Aging, Gender and Exercise (IMAGE) Research Group, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA; and.,The Immunology Graduate Program, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
262
|
Pirro M, Bianconi V, Paciullo F, Mannarino MR, Bagaglia F, Sahebkar A. Lipoprotein(a) and inflammation: A dangerous duet leading to endothelial loss of integrity. Pharmacol Res 2017; 119:178-187. [DOI: 10.1016/j.phrs.2017.02.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Revised: 01/08/2017] [Accepted: 02/02/2017] [Indexed: 12/15/2022]
|
263
|
Mechanisms of endothelial activation in sepsis and cell culture models to study the heterogeneous host response. Int J Artif Organs 2017; 40:9-14. [PMID: 28218355 DOI: 10.5301/ijao.5000560] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/27/2017] [Indexed: 01/26/2023]
Abstract
Sepsis is currently viewed as a fundamental disintegration of control functions from intracellular signalling to immunoregulatory and neuroendocrine mechanisms. The immediate threat in sepsis is invasive infection, and the need to activate immune defense mechanisms to clear the pathogen before irreparable damage occurs. In the process of pathogen elimination, however, the systemic host response to infection may cause collateral damage to the endothelium and may lead to the destruction of host tissues.A number of experimental models have been developed to monitor endothelial activation and to study endothelial dysfunction under septic conditions. Here, we review the application of these models to assess the highly variable host response in sepsis and to investigate the efficacy of adsorbent-based extracorporeal therapies. We also highlight the need for efficient diagnostic tools, which are indispensable to select patients who are likely to benefit from distinct adjunctive therapies.
Collapse
|
264
|
Nombela-Arrieta C, Isringhausen S. The Role of the Bone Marrow Stromal Compartment in the Hematopoietic Response to Microbial Infections. Front Immunol 2017; 7:689. [PMID: 28163704 PMCID: PMC5247475 DOI: 10.3389/fimmu.2016.00689] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/23/2016] [Indexed: 12/18/2022] Open
Abstract
Continuous production of blood cells unfolds within a complex three-dimensional tissue scaffold established by highly organized stromal cell networks of mesenchymal, neural, and vascular origin inside bone marrow (BM) cavities. Collectively, stromal cells have been shown to serve two principal roles; first as primary participants of bone remodeling and metabolism and second as master regulators of different stages of blood cell development and production. Indeed, ample evidence demonstrates that stromal cells can sense and integrate systemic signals to shape hematopoietic responses and that these regulatory mechanisms are subverted in multiple pathologic conditions. Microbial infections are stressors that elicit potent inflammatory reactions and induce substantial alterations of hematopoietic output. Whether the cellular components of the BM stromal microenvironment are targeted by infections and participate in infection-induced hematopoiesis has not been investigated in sufficient detail to date. In this manuscript, we provide a succinct updated overview of the different cell populations that are currently known to form BM stroma. We discuss experimental evidence demonstrating that different stromal components are actively damaged or functionally altered by pathogens and/or ensuing inflammatory signals and review how these effects are known to contribute to the hematologic manifestations observed during infections.
Collapse
|
265
|
Initiation of acute graft-versus-host disease by angiogenesis. Blood 2017; 129:2021-2032. [PMID: 28096092 DOI: 10.1182/blood-2016-08-736314] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Accepted: 01/08/2017] [Indexed: 01/06/2023] Open
Abstract
The inhibition of inflammation-associated angiogenesis ameliorates inflammatory diseases by reducing the recruitment of tissue-infiltrating leukocytes. However, it is not known if angiogenesis has an active role during the initiation of inflammation or if it is merely a secondary effect occurring in response to stimuli by tissue-infiltrating leukocytes. Here, we show that angiogenesis precedes leukocyte infiltration in experimental models of inflammatory bowel disease and acute graft-versus-host disease (GVHD). We found that angiogenesis occurred as early as day+2 after allogeneic transplantation mainly in GVHD typical target organs skin, liver, and intestines, whereas no angiogenic changes appeared due to conditioning or syngeneic transplantation. The initiation phase of angiogenesis was not associated with classical endothelial cell (EC) activation signs, such as Vegfa/VEGFR1+2 upregulation or increased adhesion molecule expression. During early GVHD at day+2, we found significant metabolic and cytoskeleton changes in target organ ECs in gene array and proteomic analyses. These modifications have significant functional consequences as indicated by profoundly higher deformation in real-time deformability cytometry. Our results demonstrate that metabolic changes trigger alterations in cell mechanics, leading to enhanced migratory and proliferative potential of ECs during the initiation of inflammation. Our study adds evidence to the hypothesis that angiogenesis is involved in the initiation of tissue inflammation during GVHD.
Collapse
|
266
|
Abstract
Chemokines are a family of small cytokines that share a typical key structure that is stabilized by disulfide bonds between the cysteine residues at the NH2-terminal of the protein, and they are secreted by a great variety of cells in several different conditions. Their function is directly dependent on their interactions with their receptors. Chemokines are involved in cell maturation and differentiation, infection, autoimmunity, cancer, and, in general, in any situation where immune components are involved. However, their role in postfracture inflammation and fracture healing is not yet well established. In this article, we will discuss the response of chemokines to bone fracture and their potential roles in postfracture inflammation and healing based on data from our studies and from other previously published studies.
Collapse
Affiliation(s)
- Bouchra Edderkaoui
- Musculoskeletal Disease Center, Loma Linda VA Health Care Systems, Loma Linda, CA, USA
- Department of Medicine, Loma Linda University, Loma Linda, CA, USA
- *Correspondence: Bouchra Edderkaoui,
| |
Collapse
|
267
|
van Niekerk G, Loos B, Nell T, Engelbrecht AM. Autophagy--A free meal in sickness-associated anorexia. Autophagy 2016; 12:727-34. [PMID: 27050464 DOI: 10.1080/15548627.2016.1147672] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022] Open
Abstract
Activation of the immune system is metabolically costly, yet a hallmark of an infection is a reduction in appetite with a subsequent reduction in metabolite provision. What is the functional value of decreasing nutrient intake when an infection imposes large demands on metabolic parameters? Here, we propose that sickness-associated anorexia (SAA) upregulates the ancient process of autophagy systemically, thereby profoundly controlling not only immune- but also nonimmune-competent cells. This allows an advanced impact on the resolution of an infection through direct pathogen killing, enhancement of epitope presentation and the contribution toward the clearance of noxious factors. By rendering a 'free meal,' autophagy is thus most fundamentally harnessed during an anorexic response in order to promote both host tolerance and resistance. These findings strongly suggest a reassessment of numerous SAA-related clinical applications and a re-evaluation of current efforts in patient care.
Collapse
Affiliation(s)
- Gustav van Niekerk
- a Department of Physiological Sciences , Stellenbosch University , Stellenbosch , South Africa
| | - Ben Loos
- b Department of Physiological Sciences , Faculty of Natural Sciences, Stellenbosch University , Stellenbosch , South Africa
| | - Theo Nell
- b Department of Physiological Sciences , Faculty of Natural Sciences, Stellenbosch University , Stellenbosch , South Africa
| | - Anna-Mart Engelbrecht
- b Department of Physiological Sciences , Faculty of Natural Sciences, Stellenbosch University , Stellenbosch , South Africa
| |
Collapse
|
268
|
Lubkin A, Torres VJ. Bacteria and endothelial cells: a toxic relationship. Curr Opin Microbiol 2016; 35:58-63. [PMID: 28013162 DOI: 10.1016/j.mib.2016.11.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 11/26/2016] [Accepted: 11/30/2016] [Indexed: 12/24/2022]
Abstract
Pathogenic bacteria use the bloodstream as a highway for getting around the body, and thus have to find ways to enter and exit through the endothelium. Many bacteria approach this problem by producing toxins that can breach the endothelial barrier through diverse creative mechanisms, including directly killing endothelial cells (ECs), weakening the cytoskeleton within ECs, and breaking the junctions between ECs. Toxins can also modulate the immune response by influencing endothelial biology, and can modulate endothelial function by influencing the response of leukocytes. Understanding these interactions, in both the in vitro and in vivo contexts, is of critical importance for designing new therapies for sepsis and other severe bacterial diseases.
Collapse
Affiliation(s)
- Ashira Lubkin
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, United States
| | - Victor J Torres
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, United States.
| |
Collapse
|
269
|
Machado-Pereira M, Santos T, Bernardino L, Ferreira R. Vascular inter-regulation of inflammation: molecular and cellular targets for CNS therapy. J Neurochem 2016; 140:692-702. [PMID: 27925201 DOI: 10.1111/jnc.13914] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2016] [Revised: 11/07/2016] [Accepted: 11/21/2016] [Indexed: 12/17/2022]
Abstract
Angiogenesis and inflammation are clearly interconnected and interdependent processes that are dysregulated in a series of systemic and brain pathologies. Herein, key aspects regarding endothelial cell function and tissue remodelling that are particularly affected or aggravated by inflammation are presented. Most importantly, the cellular and molecular mechanisms involved in the vascular regulation of the inflammatory processes occurring in several brain disorders and how they impact on disease/injury progression are detailed, highlighting potential targets for therapy. Finally, nanomedicine-based approaches designed to overcome limitations pertaining to low systemic bioavailability, light, pH and temperature sensitivity and/or rapid degradation of these targets, and to optimize their mode of action are discussed. Ultimately, we expect this review to provide new insight and to suggest novel approaches for the treatment of blood-brain barrier dysfunction per se or as a means to treat the injured or diseased central nervous system.
Collapse
Affiliation(s)
- Marta Machado-Pereira
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| | - Tiago Santos
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| | - Liliana Bernardino
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| | - Raquel Ferreira
- Health Sciences Research Centre (CICS-UBI), University of Beira Interior, Rua Marquês d'Ávila e Bolama, Covilhã, Portugal
| |
Collapse
|
270
|
Extensive Characterization and Comparison of Endothelial Cells Derived from Dermis and Adipose Tissue: Potential Use in Tissue Engineering. PLoS One 2016; 11:e0167056. [PMID: 27902740 PMCID: PMC5130240 DOI: 10.1371/journal.pone.0167056] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Accepted: 11/08/2016] [Indexed: 02/04/2023] Open
Abstract
Tissue-engineered constructs need to become quickly vascularized in order to ensure graft take. One way of achieving this is to incorporate endothelial cells (EC) into the construct. The adipose tissue stromal vascular fraction (adipose-SVF) might provide an alternative source for endothelial cells as adipose tissue can easily be obtained by liposuction. Since adipose-EC are now gaining more interest in tissue engineering, we aimed to extensively characterize endothelial cells from adipose tissue (adipose-EC) and compare them with endothelial cells from dermis (dermal-EC). The amount of endothelial cells before purification varied between 4–16% of the total stromal population. After MACS selection for CD31 positive cells, a >99% pure population of endothelial cells was obtained within two weeks of culture. Adipose- and dermal-EC expressed the typical endothelial markers PECAM-1, ICAM-1, Endoglin, VE-cadherin and VEGFR2 to a similar extent, with 80–99% of the cell population staining positive. With the exception of CXCR4, which was expressed on 29% of endothelial cells, all other chemokine receptors (CXCR1, 2, 3, and CCR2) were expressed on less than 5% of the endothelial cell populations. Adipose-EC proliferated similar to dermal-EC, but responded less to the mitogens bFGF and VEGF. A similar migration rate was found for both adipose-EC and dermal-EC in response to bFGF. Sprouting of adipose-EC and dermal-EC was induced by bFGF and VEGF in a 3D fibrin matrix. After stimulation of adipose-EC and dermal-EC with TNF-α an increased secretion was seen for PDGF-BB, but not uPA, PAI-1 or Angiopoietin-2. Furthermore, secretion of cytokines and chemokines (IL-6, CCL2, CCL5, CCL20, CXCL1, CXCL8 and CXCL10) was also upregulated by both adipose- and dermal-EC. The similar characteristics of adipose-EC compared to their dermal-derived counterpart make them particularly interesting for skin tissue engineering. In conclusion, we show here that adipose tissue provides for an excellent source of endothelial cells for tissue engineering purposes, since they are readily available, and easily isolated and amplified.
Collapse
|
271
|
Lobo-Silva D, Carriche GM, Castro AG, Roque S, Saraiva M. Balancing the immune response in the brain: IL-10 and its regulation. J Neuroinflammation 2016; 13:297. [PMID: 27881137 PMCID: PMC5121946 DOI: 10.1186/s12974-016-0763-8] [Citation(s) in RCA: 290] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 11/11/2016] [Indexed: 12/15/2022] Open
Abstract
Background The inflammatory response is critical to fight insults, such as pathogen invasion or tissue damage, but if not resolved often becomes detrimental to the host. A growing body of evidence places non-resolved inflammation at the core of various pathologies, from cancer to neurodegenerative diseases. It is therefore not surprising that the immune system has evolved several regulatory mechanisms to achieve maximum protection in the absence of pathology. Main body The production of the anti-inflammatory cytokine interleukin (IL)-10 is one of the most important mechanisms evolved by many immune cells to counteract damage driven by excessive inflammation. Innate immune cells of the central nervous system, notably microglia, are no exception and produce IL-10 downstream of pattern recognition receptors activation. However, whereas the molecular mechanisms regulating IL-10 expression by innate and acquired immune cells of the periphery have been extensively addressed, our knowledge on the modulation of IL-10 expression by central nervous cells is much scattered. This review addresses the current understanding on the molecular mechanisms regulating IL-10 expression by innate immune cells of the brain and the implications of IL-10 modulation in neurodegenerative disorders. Conclusion The regulation of IL-10 production by central nervous cells remains a challenging field. Answering the many remaining outstanding questions will contribute to the design of targeted approaches aiming at controlling deleterious inflammation in the brain.
Collapse
Affiliation(s)
- Diogo Lobo-Silva
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, Braga, Portugal
| | - Guilhermina M Carriche
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
| | - A Gil Castro
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, Braga, Portugal
| | - Susana Roque
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal.,ICVS/3B's PT Government Associate Laboratory, Braga, Portugal
| | - Margarida Saraiva
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal. .,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
272
|
Li X, Fang P, Yang WY, Chan K, Lavallee M, Xu K, Gao T, Wang H, Yang X. Mitochondrial ROS, uncoupled from ATP synthesis, determine endothelial activation for both physiological recruitment of patrolling cells and pathological recruitment of inflammatory cells. Can J Physiol Pharmacol 2016; 95:247-252. [PMID: 27925481 DOI: 10.1139/cjpp-2016-0515] [Citation(s) in RCA: 59] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Mitochondrial reactive oxygen species (mtROS) are signaling molecules, which drive inflammatory cytokine production and T cell activation. In addition, cardiovascular diseases, cancers, and autoimmune diseases all share a common feature of increased mtROS level. Both mtROS and ATP are produced as a result of electron transport chain activity, but it remains enigmatic whether mtROS could be generated independently from ATP synthesis. A recent study shed light on this important question and found that, during endothelial cell (EC) activation, mtROS could be upregulated in a proton leak-coupled, but ATP synthesis-uncoupled manner. As a result, EC could upregulate mtROS production for physiological EC activation without compromising mitochondrial membrane potential and ATP generation, and consequently without causing mitochondrial damage and EC death. Thus, a novel pathophysiological role of proton leak in driving mtROS production was uncovered for low grade EC activation, patrolling immunosurveillance cell trans-endothelial migration and other signaling events without compromising cellular survival. This new working model explains how mtROS could be increasingly generated independently from ATP synthesis and endothelial damage or death. Mapping the connections among mitochondrial metabolism, physiological EC activation, patrolling cell migration, and pathological inflammation is significant towards the development of novel therapies for inflammatory diseases and cancers.
Collapse
Affiliation(s)
- Xinyuan Li
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Pu Fang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - William Y Yang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Kylie Chan
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Muriel Lavallee
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Keman Xu
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Tracy Gao
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Hong Wang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| | - Xiaofeng Yang
- Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA.,Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research, Department of Pharmacology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140, USA
| |
Collapse
|
273
|
Navarro R, Compte M, Álvarez-Vallina L, Sanz L. Immune Regulation by Pericytes: Modulating Innate and Adaptive Immunity. Front Immunol 2016; 7:480. [PMID: 27867386 PMCID: PMC5095456 DOI: 10.3389/fimmu.2016.00480] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Accepted: 10/20/2016] [Indexed: 01/22/2023] Open
Abstract
Pericytes (PC) are mural cells that surround endothelial cells in small blood vessels. PC have traditionally been credited with structural functions, being essential for vessel maturation and stabilization. However, an accumulating body of evidence suggests that PC also display immune properties. They can respond to a series of pro-inflammatory stimuli and are able to sense different types of danger due to their expression of functional pattern-recognition receptors, contributing to the onset of innate immune responses. In this context, PC not only secrete a variety of chemokines but also overexpress adhesion molecules such as ICAM-1 and VCAM-1 involved in the control of immune cell trafficking across vessel walls. In addition to their role in innate immunity, PC are involved in adaptive immunity. It has been reported that interaction with PC anergizes T cells, which is attributed, at least in part, to the expression of PD-L1. As components of the tumor microenvironment, PC can also modulate the antitumor immune response. However, their role is complex, and further studies will be required to better understand the crosstalk of PC with immune cells in order to consider them as potential therapeutic targets. In any case, PC will be looked at with new eyes by immunologists from now on.
Collapse
Affiliation(s)
- Rocío Navarro
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda , Madrid , Spain
| | - Marta Compte
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda , Madrid , Spain
| | - Luis Álvarez-Vallina
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain; Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus, Denmark
| | - Laura Sanz
- Molecular Immunology Unit, Hospital Universitario Puerta de Hierro Majadahonda , Madrid , Spain
| |
Collapse
|
274
|
Hara T, Nakashima Y, Sakai Y, Nishio H, Motomura Y, Yamasaki S. Kawasaki disease: a matter of innate immunity. Clin Exp Immunol 2016; 186:134-143. [PMID: 27342882 PMCID: PMC5054572 DOI: 10.1111/cei.12832] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/22/2016] [Indexed: 12/26/2022] Open
Abstract
Kawasaki disease (KD) is an acute systemic vasculitis of childhood that does not have a known cause or aetiology. The epidemiological features (existence of epidemics, community outbreaks and seasonality), unique age distribution and clinical symptoms and signs of KD suggest that the disease is caused by one or more infectious environmental triggers. However, KD is not transmitted person-to-person and does not occur in clusters within households, schools or nurseries. KD is a self-limited illness that is not associated with the production of autoantibodies or the deposition of immune complexes, and it rarely recurs. Regarding the underlying pathophysiology of KD, innate immune activity (the inflammasome) is believed to play a role in the development of KD vasculitis, based on the results of studies with animal models and the clinical and laboratory findings of KD patients. Animal studies have demonstrated that innate immune pathogen-associated molecular patterns (PAMPs) can cause vasculitis independently of acquired immunity and have provided valuable insights regarding the underlying mechanisms of this phenomenon. To validate this concept, we recently searched for KD-specific PAMPs and identified such molecules with high specificity and sensitivity. These molecules have structures similar to those of microbe-associated molecular patterns (MAMPs), as shown by liquid chromatography-tandem mass spectrometry. We propose herein that KD is an innate immune disorder resulting from the exposure of a genetically predisposed individual to microbe-derived innate immune stimulants and that it is not a typical infectious disease.
Collapse
Affiliation(s)
- T Hara
- Fukuoka Children's Hospital.
- Department of Pediatrics, Graduate School of Medical Sciences.
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan.
| | - Y Nakashima
- Department of Pediatrics, Graduate School of Medical Sciences
| | - Y Sakai
- Department of Pediatrics, Graduate School of Medical Sciences
| | - H Nishio
- Department of Pediatrics, Graduate School of Medical Sciences
| | - Y Motomura
- Department of Pediatrics, Graduate School of Medical Sciences
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - S Yamasaki
- Division of Molecular Immunology, Research Center for Infectious Diseases, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
275
|
SIRT1 inhibits inflammatory response partly through regulation of NLRP3 inflammasome in vascular endothelial cells. Mol Immunol 2016; 77:148-56. [PMID: 27505710 DOI: 10.1016/j.molimm.2016.07.018] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Revised: 06/25/2016] [Accepted: 07/29/2016] [Indexed: 11/23/2022]
Abstract
Emerging evidence has indicated that vascular endothelial cells (ECs) not only form the barrier between blood and the vessel wall but also serve as conditional innate immune cells. Our previous study found that SIRT1, a class III histone deacetylase, inhibits the inflammatory response in ECs. Recent studies revealed that SIRT1 also participates in the modulation of immune responses. Although the NLRP3 inflammasome is known to be a crucial component of the innate immune system, there is no direct evidence demonstrating the anti-inflammatory effect of SIRT1 on ECs through the NLRP3 inflammasome. In this study, we observed that lipopolysaccharide (LPS) and adenosine triphosphate (ATP) triggered the activation of NLRP3 inflammasome in human umbilical vein ECs (HUVECs). Moreover, SIRT1 expression was reduced in HUVECs stimulated with LPS and ATP. SIRT1 activator inhibited the expression of monocyte chemotactic protein-1 (MCP-1) and C-reactive protein (CRP), whereas SIRT1 knockdown resulted in significant increases in MCP-1 and CRP levels in HUVECs stimulated with LPS and ATP. Importantly, the lack of SIRT1 enhanced NLRP3 inflammasome activation and subsequent caspase-1 cleavage. On the other hand, NLRP3 siRNA blocked the activation of the NLRP3 inflammasome in HUVECs stimulated with LPS plus ATP. Further study revealed that NLRP3 inflammasome blockade significantly reduced MCP-1 and CRP production in HUVECs. In vivo studies indicated that implantation of the periarterial carotid collar inhibited arterial SIRT1 expression in rabbits. Meanwhile, treatment with a SIRT1 activator decreased the expression levels of MCP-1 and CRP in collared arteries and the interleukin (IL)-1β level in serum. Taken together, these findings indicate that NLRP3 inflammasome activation promoted endothelial inflammation and that SIRT1 inhibits the inflammatory response partly through regulation of the NLRP3 inflammasome in ECs.
Collapse
|
276
|
Pantsulaia I, Ciszewski WM, Niewiarowska J. Senescent endothelial cells: Potential modulators of immunosenescence and ageing. Ageing Res Rev 2016; 29:13-25. [PMID: 27235855 DOI: 10.1016/j.arr.2016.05.011] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2016] [Revised: 05/24/2016] [Accepted: 05/24/2016] [Indexed: 02/08/2023]
Abstract
Recent studies have demonstrated that the accumulation of senescent endothelial cells may be the primary cause of cardiovascular diseases. Because of their multifunctional properties, endothelial cells actively take part in stimulating the immune system and inflammation. In addition, ageing is characterized by the progressive deterioration of immune cells and a decline in the activation of the immune response. This results in a loss of the primary function of the immune system, which is eliminating damaged/senescent cells and neutralizing potential sources of harmful inflammatory reactions. In this review, we discuss cellular senescence and the senescence-associated secretory phenotype (SASP) of endothelial cells and summarize the link between endothelial cells and immunosenescence. We describe the possibility that age-related changes in Toll-like receptors (TLRs) and microRNAs can affect the phenotypes of senescent endothelial cells and immune cells via a negative feedback loop aimed at restraining the excessive pro-inflammatory response. This review also addresses the following questions: how do senescent endothelial cells influence ageing or age-related changes in the inflammatory burden; what is the connection between ECs and immunosenescence, and what are the crucial hypothetical pathways linking endothelial cells and the immune system during ageing.
Collapse
|
277
|
Ambrozova G, Fidlerova T, Verescakova H, Koudelka A, Rudolph TK, Woodcock SR, Freeman BA, Kubala L, Pekarova M. Nitro-oleic acid inhibits vascular endothelial inflammatory responses and the endothelial-mesenchymal transition. Biochim Biophys Acta Gen Subj 2016; 1860:2428-2437. [PMID: 27431604 DOI: 10.1016/j.bbagen.2016.07.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Revised: 06/24/2016] [Accepted: 07/14/2016] [Indexed: 11/15/2022]
Abstract
BACKGROUND Inflammatory-mediated pathological processes in the endothelium arise as a consequence of the dysregulation of vascular homeostasis. Of particular importance are mediators produced by stimulated monocytes/macrophages inducing activation of endothelial cells (ECs). This is manifested by excessive soluble pro-inflammatory mediator production and cell surface adhesion molecule expression. Nitro-fatty acids are endogenous products of metabolic and inflammatory reactions that display immuno-regulatory potential and may represent a novel therapeutic strategy to treat inflammatory diseases. The purpose of our study was to characterize the effects of nitro-oleic acid (OA-NO2) on inflammatory responses and the endothelial-mesenchymal transition (EndMT) in ECs that is a consequence of the altered healing phase of the immune response. METHODS The effect of OA-NO2 on inflammatory responses and EndMT was determined in murine macrophages and murine and human ECs using Western blotting, ELISA, immunostaining, and functional assays. RESULTS OA-NO2 limited the activation of macrophages and ECs by reducing pro-inflammatory cytokine production and adhesion molecule expression through its modulation of STAT, MAPK and NF-κB-regulated signaling. OA-NO2 also decreased transforming growth factor-β-stimulated EndMT and pro-fibrotic phenotype of ECs. These effects are related to the downregulation of Smad2/3. CONCLUSIONS The study shows the pleiotropic effect of OA-NO2 on regulating EC-macrophage interactions during the immune response and suggests a role for OA-NO2 in the regulation of vascular endothelial immune and fibrotic responses arising during chronic inflammation. GENERAL SIGNIFICANCE These findings propose the OA-NO2 may be useful as a novel therapeutic agent for treatment of cardiovascular disorders associated with dysregulation of the endothelial immune response.
Collapse
Affiliation(s)
- Gabriela Ambrozova
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Tana Fidlerova
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65, Brno, Czech Republic
| | - Hana Verescakova
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65, Brno, Czech Republic; Faculty of Science, Institute of Experimental Biology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Adolf Koudelka
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65, Brno, Czech Republic; Faculty of Science, Institute of Experimental Biology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic
| | - Tanja K Rudolph
- Faculty of Science, Institute of Experimental Biology, Masaryk University, Kamenice 5, 625 00 Brno, Czech Republic; Heart Centre, University Hospital of Cologne, Kerpener Str. 62, 50937 Cologne, Germany
| | - Steven R Woodcock
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, USA
| | - Bruce A Freeman
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, 4200 Fifth Ave, Pittsburgh, PA 15260, USA
| | - Lukas Kubala
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65, Brno, Czech Republic; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic
| | - Michaela Pekarova
- Department of Free Radical Pathophysiology, Institute of Biophysics, Academy of Sciences of the Czech Republic, Kralovopolska 135, 612 65, Brno, Czech Republic; International Clinical Research Center - Center of Biomolecular and Cellular Engineering, St. Anne's University Hospital, Pekarska 53, 656 91, Brno, Czech Republic.
| |
Collapse
|
278
|
van Niekerk G, Isaacs AW, Nell T, Engelbrecht AM. Sickness-Associated Anorexia: Mother Nature's Idea of Immunonutrition? Mediators Inflamm 2016; 2016:8071539. [PMID: 27445441 PMCID: PMC4942670 DOI: 10.1155/2016/8071539] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 02/06/2023] Open
Abstract
During an infection, expansion of immune cells, assembly of antibodies, and the induction of a febrile response collectively place continual metabolic strain on the host. These considerations also provide a rationale for nutritional support in critically ill patients. Yet, results from clinical and preclinical studies indicate that aggressive nutritional support does not always benefit patients and may occasionally be detrimental. Moreover, both vertebrates and invertebrates exhibit a decrease in appetite during an infection, indicating that such sickness-associated anorexia (SAA) is evolutionarily conserved. It also suggests that SAA performs a vital function during an infection. We review evidence signifying that SAA may present a mechanism by which autophagic flux is upregulated systemically. A decrease in serum amino acids during an infection promotes autophagy not only in immune cells, but also in nonimmune cells. Similarly, bile acids reabsorbed postprandially inhibit hepatic autophagy by binding to farnesoid X receptors, indicating that SAA may be an attempt to conserve autophagy. In addition, augmented autophagic responses may play a critical role in clearing pathogens (xenophagy), in the presentation of epitopes in nonprovisional antigen presenting cells and the removal of damaged proteins and organelles. Collectively, these observations suggest that some patients might benefit from permissive underfeeding.
Collapse
Affiliation(s)
- Gustav van Niekerk
- Department of Physiological Sciences, Private Bag X1, Matieland, Stellenbosch 7600, South Africa
| | - Ashwin W. Isaacs
- Department of Physiological Sciences, Private Bag X1, Matieland, Stellenbosch 7600, South Africa
| | - Theo Nell
- Department of Physiological Sciences, Private Bag X1, Matieland, Stellenbosch 7600, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Private Bag X1, Matieland, Stellenbosch 7600, South Africa
| |
Collapse
|
279
|
Kim JH, Jeong HS, Kim KM, Lee YJ, Jung MH, Park JJ, Kim JP, Woo SH. Extra-Esophageal Pepsin from Stomach Refluxate Promoted Tonsil Hypertrophy. PLoS One 2016; 11:e0152336. [PMID: 27058240 PMCID: PMC4825923 DOI: 10.1371/journal.pone.0152336] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 03/11/2016] [Indexed: 11/24/2022] Open
Abstract
Background Gastroesophageal reflux is associated with numerous pathologic conditions of the upper aerodigestive tract. Gastric pepsin within reflux contributes to immunologic reactions in the tonsil. In this study, we aimed to find the relationships between pepsin and tonsillar hypertrophy. Methods and finding We explored the notion whether tonsillar hypertrophy was due to pepsin-mediated gastric reflux in tonsil hypertrophy. Fifty-four children with tonsil hypertrophy and 30 adults with tonsillitis were recruited before surgical treatment. Blood and tonsil tissues from each patient were harvested for analysis of changes in lymphocyte and macrophage numbers coupled with histological and biochemical analysis. Pepsin was expressed at different levels in tonsil tissues from each tonsillar hypertrophy. Pepsin-positive cells were found in the crypt epithelium, surrounding the lymphoid follicle with developing fibrosis, and also surrounding the lymphoid follicle that faced the crypt. And also, pepsin staining was well correlated with damaged tonsillar squamous epithelium and TGF-β1 and iNOS expression in the tonsil section. In addition, pepsin and TGF-β1-positive cells were co-localized with CD68-positive cells in the crypt and surrounding germinal centers. In comparison of macrophage responsiveness to pepsin, peripheral blood mononuclear cells (PBMNCs) were noticeably larger in the presence of activated pepsin in the child group. Furthermore, CD11c and CD163-positive cells were significantly increased by activated pepsin. However, this was not seen for the culture of PBMNCs from the adult group. Conclusions The lymphocytes and monocytes are in a highly proliferative state in the tonsillar hypertrophy and associated with increased expression of pro-inflammatory factors as a result of exposure to stomach reflux pepsin.
Collapse
Affiliation(s)
- Jin Hyun Kim
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
| | - Han-Sin Jeong
- Department of Otorhinolaryngology-Head and Neck Surgery, Sungkyunkwan University School of Medicine, Samsung Medical Center, Seoul, Korea
| | - Kyung Mi Kim
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
| | - Ye Jin Lee
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
| | - Myeong Hee Jung
- Biomedical Research Institute, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
| | - Jung Je Park
- Department of Otolaryngology, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
| | - Jin Pyeong Kim
- Department of Otolaryngology, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
| | - Seung Hoon Woo
- Department of Otolaryngology, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
- Institute of Health Sciences, Gyeongsang National University Hospital, Jinju, Gyeongsangnamdo, Korea
- * E-mail:
| |
Collapse
|
280
|
D'Angelo W, Acharya D, Wang R, Wang J, Gurung C, Chen B, Bai F, Guo YL. Development of Antiviral Innate Immunity During In Vitro Differentiation of Mouse Embryonic Stem Cells. Stem Cells Dev 2016; 25:648-59. [PMID: 26906411 DOI: 10.1089/scd.2015.0377] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The innate immunity of embryonic stem cells (ESCs) has recently emerged as an important issue in ESC biology and in ESC-based regenerative medicine. We have recently reported that mouse ESCs (mESCs) do not have a functional type I interferon (IFN)-based antiviral innate immunity. They are deficient in expressing IFN in response to viral infection and have limited ability to respond to IFN. Using fibroblasts (FBs) as a cell model, the current study investigated the development of antiviral mechanisms during in vitro differentiation of mESCs. We demonstrate that mESC-differentiated FBs (mESC-FBs) share extensive similarities with naturally differentiated FBs in morphology, marker expression, and growth pattern, but their development of antiviral mechanisms lags behind. Nonetheless, the antiviral mechanisms are inducible during mESC differentiation as demonstrated by the transition of nuclear factor kappa B (NFκB), a key transcription factor for IFN expression, from its inactive state in mESCs to its active state in mESC-FBs and by increased responses of mESC-FBs to viral stimuli and IFN during their continued in vitro propagation. Together with our previously published study, the current data provide important insights into molecular basis for the deficiency of IFN expression in mESCs and the development of antiviral innate immunity during mESC differentiation.
Collapse
Affiliation(s)
- William D'Angelo
- 1 Department of Biological Sciences, the University of Southern Mississippi , Hattiesburg, Mississippi
| | - Dhiraj Acharya
- 1 Department of Biological Sciences, the University of Southern Mississippi , Hattiesburg, Mississippi
| | - Ruoxing Wang
- 2 Department of Cancer Biology, University of Pennsylvania School of Medicine , Philadelphia, Pennsylvania
| | - Jundi Wang
- 1 Department of Biological Sciences, the University of Southern Mississippi , Hattiesburg, Mississippi
| | - Chandan Gurung
- 1 Department of Biological Sciences, the University of Southern Mississippi , Hattiesburg, Mississippi
| | - Bohan Chen
- 1 Department of Biological Sciences, the University of Southern Mississippi , Hattiesburg, Mississippi
| | - Fengwei Bai
- 1 Department of Biological Sciences, the University of Southern Mississippi , Hattiesburg, Mississippi
| | - Yan-Lin Guo
- 1 Department of Biological Sciences, the University of Southern Mississippi , Hattiesburg, Mississippi
| |
Collapse
|
281
|
Lerner TR, de Souza Carvalho-Wodarz C, Repnik U, Russell MR, Borel S, Diedrich CR, Rohde M, Wainwright H, Collinson LM, Wilkinson RJ, Griffiths G, Gutierrez MG. Lymphatic endothelial cells are a replicative niche for Mycobacterium tuberculosis. J Clin Invest 2016; 126:1093-108. [PMID: 26901813 PMCID: PMC4767353 DOI: 10.1172/jci83379] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Accepted: 01/04/2016] [Indexed: 12/13/2022] Open
Abstract
In extrapulmonary tuberculosis, the most common site of infection is within the lymphatic system, and there is growing recognition that lymphatic endothelial cells (LECs) are involved in immune function. Here, we identified LECs, which line the lymphatic vessels, as a niche for Mycobacterium tuberculosis in the lymph nodes of patients with tuberculosis. In cultured primary human LECs (hLECs), we determined that M. tuberculosis replicates both in the cytosol and within autophagosomes, but the bacteria failed to replicate when the virulence locus RD1 was deleted. Activation by IFN-γ induced a cell-autonomous response in hLECs via autophagy and NO production that restricted M. tuberculosis growth. Thus, depending on the activation status of LECs, autophagy can both promote and restrict replication. Together, these findings reveal a previously unrecognized role for hLECs and autophagy in tuberculosis pathogenesis and suggest that hLECs are a potential niche for M. tuberculosis that allows establishment of persistent infection in lymph nodes.
Collapse
Affiliation(s)
- Thomas R. Lerner
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
| | - Cristiane de Souza Carvalho-Wodarz
- Department of Drug Delivery, Helmholtz Institute for Pharmaceutical Research Saarland, Helmholtz Centre for Infection Research, Saarland University, Saarbrücken, Germany
| | - Urska Repnik
- Department of Biosciences, University of Oslo, Blindernveien, Oslo, Norway
| | - Matthew R.G. Russell
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Sophie Borel
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
| | - Collin R. Diedrich
- Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
| | - Manfred Rohde
- Central Facility for Microscopy, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Helen Wainwright
- Department of Anatomical Pathology, University of Cape Town Faculty of Health Sciences and National Health Laboratory Service, Groote Schuur Hospital, Cape Town, South Africa
| | - Lucy M. Collinson
- Electron Microscopy Science Technology Platform, The Francis Crick Institute, Lincoln’s Inn Fields Laboratory, London, United Kingdom
| | - Robert J. Wilkinson
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
- Clinical Infectious Diseases Research Initiative, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, South Africa
- Department of Medicine, Imperial College London, London, United Kingdom
| | - Gareth Griffiths
- Department of Biosciences, University of Oslo, Blindernveien, Oslo, Norway
| | - Maximiliano G. Gutierrez
- Host-pathogen interactions in tuberculosis laboratory, The Francis Crick Institute, Mill Hill Laboratory, London, United Kingdom
| |
Collapse
|
282
|
Nymo S, Gustavsen A, Nilsson PH, Lau C, Espevik T, Mollnes TE. Human Endothelial Cell Activation by Escherichia coli and Staphylococcus aureus Is Mediated by TNF and IL-1β Secondarily to Activation of C5 and CD14 in Whole Blood. THE JOURNAL OF IMMUNOLOGY 2016; 196:2293-9. [PMID: 26800874 DOI: 10.4049/jimmunol.1502220] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/22/2015] [Indexed: 11/19/2022]
Abstract
Endothelial cells (EC) play a central role in inflammation. E-selectin and ICAM-1 expression are essential for leukocyte recruitment and are good markers of EC activation. Most studies of EC activation are done in vitro using isolated mediators. The aim of the present study was to examine the relative importance of pattern recognition systems and downstream mediators in bacteria-induced EC activation in a physiological relevant human model, using EC incubated with whole blood. HUVEC were incubated with human whole blood. Escherichia coli- and Staphylococcus aureus-induced EC activation was measured by E-selectin and ICAM-1 expression using flow cytometry. The mAb 18D11 was used to neutralize CD14, and the lipid A analog eritoran was used to block TLR4/MD2. C5 cleavage was inhibited using eculizumab, and C5aR1 was blocked by an antagonist. Infliximab and canakinumab were used to neutralize TNF and IL-1β. The EC were minimally activated when bacteria were incubated in serum, whereas a substantial EC activation was seen when the bacteria were incubated in whole blood. E. coli-induced activation was largely CD14-dependent, whereas S. aureus mainly caused a C5aR1-mediated response. Combined CD14 and C5 inhibition reduced E-selectin and ICAM-1 expression by 96 and 98% for E. coli and by 70 and 75% for S. aureus. Finally, the EC activation by both bacteria was completely abolished by combined inhibition of TNF and IL-1β. E. coli and S. aureus activated EC in a CD14- and C5-dependent manner with subsequent leukocyte secretion of TNF and IL-1β mediating the effect.
Collapse
Affiliation(s)
- Stig Nymo
- Research Laboratory, Nordland Hospital, 8092 Bodø, Norway; Faculty of Health Sciences, K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway; Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, 0424 Oslo, Norway; K. G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
| | - Alice Gustavsen
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, 0424 Oslo, Norway; K. G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
| | - Per H Nilsson
- Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, 0424 Oslo, Norway; K. G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway
| | - Corinna Lau
- Research Laboratory, Nordland Hospital, 8092 Bodø, Norway; Faculty of Health Sciences, K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway
| | - Terje Espevik
- Center of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway; and Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 7491 Trondheim, Norway
| | - Tom Eirik Mollnes
- Research Laboratory, Nordland Hospital, 8092 Bodø, Norway; Faculty of Health Sciences, K. G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, 9037 Tromsø, Norway; Department of Immunology, Oslo University Hospital, Rikshospitalet, University of Oslo, 0424 Oslo, Norway; K. G. Jebsen Inflammation Research Center, University of Oslo, 0424 Oslo, Norway; Center of Molecular Inflammation Research, Norwegian University of Science and Technology, 7491 Trondheim, Norway; and
| |
Collapse
|
283
|
Mangold CA, Masser DR, Stanford DR, Bixler GV, Pisupati A, Giles CB, Wren JD, Ford MM, Sonntag WE, Freeman WM. CNS-wide Sexually Dimorphic Induction of the Major Histocompatibility Complex 1 Pathway With Aging. J Gerontol A Biol Sci Med Sci 2016; 72:16-29. [PMID: 26786204 DOI: 10.1093/gerona/glv232] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 12/09/2015] [Indexed: 01/01/2023] Open
Abstract
The major histocompatibility complex I (MHCI) pathway, which canonically functions in innate immune viral antigen presentation and detection, is functionally pleiotropic in the central nervous system (CNS). Alternative roles include developmental synapse pruning, regulation of synaptic plasticity, and inhibition of neuronal insulin signaling; all processes altered during brain aging. Upregulation of MHCI components with aging has been reported; however, no systematic examination of MHCI cellular localization, expression, and regulation across CNS regions, life span, and sexes has been reported. In the mouse, MHCI is expressed by neurons and microglia, and MHCI components and receptors (H2-K1, H2-D1, β2M, Lilrb3, Klra2, CD247) display markedly different expression profiles across the hippocampus, cortex, cerebellum, brainstem, and retina. MHCI components, receptors, associated inflammatory transcripts (IL1α, IL1β, IL6, TNFα), and TAP (transporter associated with antigen processing) components are induced with aging and to a greater degree in female than male mice across CNS regions. H2-K1 and H2-D1 expression is associated with differential CG and non-CG promoter methylation across CNS regions, ages, and between sexes, and concomitant increased expression of proinflammatory genes. Meta-analysis of human brain aging data also demonstrates age-related increases in MHCI. Induction of MHCI signaling could contribute to altered synapse regulation and impaired synaptic plasticity with aging.
Collapse
Affiliation(s)
- Colleen A Mangold
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey
| | - Dustin R Masser
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey.,Department of Physiology, University of Oklahoma Health Sciences Center.,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - David R Stanford
- Department of Physiology, University of Oklahoma Health Sciences Center.,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Georgina V Bixler
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey
| | - Aditya Pisupati
- MD/PhD Program, College of Medicine, Pennsylvania State University, Hershey
| | - Cory B Giles
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation
| | - Jonathan D Wren
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation
| | - Matthew M Ford
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton
| | - William E Sonntag
- Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| | - Willard M Freeman
- Department of Pharmacology, College of Medicine, Pennsylvania State University, Hershey. .,Department of Physiology, University of Oklahoma Health Sciences Center.,Reynolds Oklahoma Center on Aging, University of Oklahoma Health Sciences Center
| |
Collapse
|
284
|
Mai J, Nanayakkara G, Lopez-Pastrana J, Li X, Li YF, Wang X, Song A, Virtue A, Shao Y, Shan H, Liu F, Autieri MV, Kunapuli SP, Iwakura Y, Jiang X, Wang H, Yang XF. Interleukin-17A Promotes Aortic Endothelial Cell Activation via Transcriptionally and Post-translationally Activating p38 Mitogen-activated Protein Kinase (MAPK) Pathway. J Biol Chem 2016; 291:4939-54. [PMID: 26733204 DOI: 10.1074/jbc.m115.690081] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Indexed: 01/18/2023] Open
Abstract
Interleukin-17 (IL-17)-secreting T helper 17 cells were recently identified as a CD4(+) T helper subset and implicated in various inflammatory and autoimmune diseases. The issues of whether and by what mechanism hyperlipidemic stress induces IL-17A to activate aortic endothelial cells (ECs) and enhance monocyte adhesion remained largely unknown. Using biochemical, immunological, microarray, experimental data mining analysis, and pathological approaches focused on primary human and mouse aortic ECs (HAECs and MAECs) and our newly generated apolipoprotein E (ApoE)(-/-)/IL-17A(-/-) mice, we report the following new findings. 1) The hyperlipidemia stimulus oxidized low density lipoprotein up-regulated IL-17 receptor(s) in HAECs and MAECs. 2) IL-17A activated HAECs and increased human monocyte adhesion in vitro. 3) A deficiency of IL-17A reduced leukocyte adhesion to endothelium in vivo. 3) IL-17A activated HAECs and MAECs via up-regulation of proinflammatory cytokines IL-6, granulocyte-macrophage colony-stimulating factor (GM-CSF), chemokine CXC motif ligand 1 (CXCL1), and CXCL2. 4) IL-17A activated ECs specifically via the p38 mitogen-activated protein kinases (MAPK) pathway; the inhibition of p38 MAPK in ECs attenuated IL-17A-mediated activation by ameliorating the expression of the aforementioned proinflammatory cytokines, chemokines, and EC adhesion molecules including intercellular adhesion molecule 1. Taken together, our results demonstrate for the first time that IL-17A activates aortic ECs specifically via p38 MAPK pathway.
Collapse
Affiliation(s)
- Jietang Mai
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and Departments of Pharmacology and
| | - Gayani Nanayakkara
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Jahaira Lopez-Pastrana
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Xinyuan Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and Departments of Pharmacology and
| | - Ya-Feng Li
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Xin Wang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Ai Song
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Anthony Virtue
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and Departments of Pharmacology and
| | - Ying Shao
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Huimin Shan
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Fang Liu
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Michael V Autieri
- Physiology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Satya P Kunapuli
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and Physiology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania 19140 and
| | - Yoichiro Iwakura
- Center for Experimental Medicine, Institute of Medical Science, University of Tokyo, Tokyo 108-8639, Japan
| | - Xiaohua Jiang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and
| | - Hong Wang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and Departments of Pharmacology and
| | - Xiao-Feng Yang
- From the Centers for Metabolic Disease Research, Cardiovascular Research, and Thrombosis Research and Departments of Pharmacology and
| |
Collapse
|
285
|
Shoda T, Futamura K, Orihara K, Emi-Sugie M, Saito H, Matsumoto K, Matsuda A. Recent advances in understanding the roles of vascular endothelial cells in allergic inflammation. Allergol Int 2016; 65:21-9. [PMID: 26666487 DOI: 10.1016/j.alit.2015.08.001] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 07/30/2015] [Accepted: 08/10/2015] [Indexed: 12/14/2022] Open
Abstract
Allergic disorders commonly involve both chronic tissue inflammation and remodeling caused by immunological reactions to various antigens on tissue surfaces. Due to their anatomical location, vascular endothelial cells are the final responders to interact with various exogenous factors that come into contact with the epithelial surface, such as pathogen-associated molecular patterns (PAMPs) and antigens. Recent studies have shed light on the important roles of endothelial cells in the development and exacerbation of allergic disorders. For instance, endothelial cells have the greatest potential to produce several key molecules that are deeply involved in allergic inflammation, such as periostin and thymus and activation-regulated chemokine (TARC/CCL17). Additionally, endothelial cells were recently shown to be important functional targets for IL-33--an essential regulator of allergic inflammation. Notably, almost all endothelial cell responses and functions involved in allergic inflammation are not suppressed by corticosteroids. These corticosteroid-refractory endothelial cell responses and functions include TNF-α-associated angiogenesis, leukocyte adhesion, IL-33-mediated responses and periostin and TARC production. Therefore, these unique responses and functions of endothelial cells may be critically involved in the pathogenesis of various allergic disorders, especially their refractory processes. Here, we review recent studies, including ours, which have elucidated previously unknown pathophysiological roles of vascular endothelial cells in allergic inflammation and discuss the possibility of endothelium-targeted therapy for allergic disorders.
Collapse
Affiliation(s)
- Tetsuo Shoda
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan.
| | - Kyoko Futamura
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kanami Orihara
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Maiko Emi-Sugie
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hirohisa Saito
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Kenji Matsumoto
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Akio Matsuda
- Department of Allergy and Immunology, National Research Institute for Child Health and Development, Tokyo, Japan
| |
Collapse
|
286
|
Konukoglu D, Uzun H. Endothelial Dysfunction and Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 956:511-540. [DOI: 10.1007/5584_2016_90] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
287
|
Carman CV, Martinelli R. T Lymphocyte-Endothelial Interactions: Emerging Understanding of Trafficking and Antigen-Specific Immunity. Front Immunol 2015; 6:603. [PMID: 26635815 PMCID: PMC4657048 DOI: 10.3389/fimmu.2015.00603] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 11/10/2015] [Indexed: 12/26/2022] Open
Abstract
Antigen-specific immunity requires regulated trafficking of T cells in and out of diverse tissues in order to orchestrate lymphocyte development, immune surveillance, responses, and memory. The endothelium serves as a unique barrier, as well as a sentinel, between the blood and the tissues, and as such it plays an essential locally tuned role in regulating T cell migration and information exchange. While it is well established that chemoattractants and adhesion molecules are major determinants of T cell trafficking, emerging studies have now enumerated a large number of molecular players as well as a range of discrete cellular remodeling activities (e.g., transmigratory cups and invadosome-like protrusions) that participate in directed migration and pathfinding by T cells. In addition to providing trafficking cues, intimate cell-cell interaction between lymphocytes and endothelial cells provide instruction to T cells that influence their activation and differentiation states. Perhaps the most intriguing and underappreciated of these "sentinel" roles is the ability of the endothelium to act as a non-hematopoietic "semiprofessional" antigen-presenting cell. Close contacts between circulating T cells and antigen-presenting endothelium may play unique non-redundant roles in shaping adaptive immune responses within the periphery. A better understanding of the mechanisms directing T cell trafficking and the antigen-presenting role of the endothelium may not only increase our knowledge of the adaptive immune response but also empower the utility of emerging immunomodulatory therapeutics.
Collapse
Affiliation(s)
- Christopher V Carman
- Center for Vascular Biology Research, Department of Medicine and Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| | - Roberta Martinelli
- Center for Vascular Biology Research, Department of Medicine and Emergency Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School , Boston, MA , USA
| |
Collapse
|
288
|
Abstract
We have recently demonstrated that brain endothelial cells cross-present parasite antigen during mouse experimental cerebral malaria (ECM). Here we describe a 2-d protocol to detect cross-presentation by isolating the brain microvessels and incubating them with a reporter cell line that expresses lacZ upon detection of the relevant peptide-major histocompatibility complex. After X-gal staining, a typical positive result consists of hundreds of blue spots, compared with fewer than 20 spots from a naive brain. The assay is generalizable to other disease contexts by using reporter cells that express appropriate specific T cell receptors. Also described is the protocol for culturing endothelial cells from brain microvessels isolated from naive mice. After 7-10 d, an in vitro cross-presentation assay can be performed by adding interferon-γ, antigen (e.g., Plasmodium berghei-infected red blood cells) and reporter cells in sequence over 3 d. This is useful for comparing different antigen forms or for probing the effects of various interventions.
Collapse
|
289
|
Coelho-Santos V, Leitão RA, Cardoso FL, Palmela I, Rito M, Barbosa M, Brito MA, Fontes-Ribeiro CA, Silva AP. The TNF-α/NF-κB signaling pathway has a key role in methamphetamine-induced blood-brain barrier dysfunction. J Cereb Blood Flow Metab 2015; 35:1260-71. [PMID: 25899299 PMCID: PMC4528012 DOI: 10.1038/jcbfm.2015.59] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 02/17/2015] [Accepted: 03/13/2015] [Indexed: 01/09/2023]
Abstract
Methamphetamine (METH) is a psychostimulant that causes neurologic and psychiatric abnormalities. Recent studies have suggested that its neurotoxicity may also result from its ability to compromise the blood-brain barrier (BBB). Herein, we show that METH rapidly increased the vesicular transport across endothelial cells (ECs), followed by an increase of paracellular transport. Moreover, METH triggered the release of tumor necrosis factor-alpha (TNF-α), and the blockade of this cytokine or the inhibition of nuclear factor-kappa B (NF-κB) pathway prevented endothelial dysfunction. Since astrocytes have a crucial role in modulating BBB function, we further showed that conditioned medium obtained from astrocytes previously exposed to METH had a negative impact on barrier properties also via TNF-α/NF-κB pathway. Animal studies corroborated the in vitro results. Overall, we show that METH directly interferes with EC properties or indirectly via astrocytes through the release of TNF-α and subsequent activation of NF-κB pathway culminating in barrier dysfunction.
Collapse
Affiliation(s)
- Vanessa Coelho-Santos
- 1] Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal [2] Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ricardo A Leitão
- 1] Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal [2] Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Filipa L Cardoso
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Inês Palmela
- Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Manuel Rito
- Neurosurgery Service, Coimbra Hospital and University Centre, Coimbra, Portugal
| | - Marcos Barbosa
- 1] Neurosurgery Service, Coimbra Hospital and University Centre, Coimbra, Portugal [2] Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria A Brito
- 1] Research Institute for Medicines (iMed.ULisboa), Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal [2] Department of Biochemistry and Human Biology, Faculdade de Farmácia, Universidade de Lisboa, Lisboa, Portugal
| | - Carlos A Fontes-Ribeiro
- 1] Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal [2] Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana P Silva
- 1] Laboratory of Pharmacology and Experimental Therapeutics, Faculty of Medicine, University of Coimbra, Coimbra, Portugal [2] Institute for Biomedical Imaging and Life Sciences (IBILI), Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
290
|
Guo YL, Carmichael GG, Wang R, Hong X, Acharya D, Huang F, Bai F. Attenuated Innate Immunity in Embryonic Stem Cells and Its Implications in Developmental Biology and Regenerative Medicine. Stem Cells 2015; 33:3165-73. [PMID: 26086534 DOI: 10.1002/stem.2079] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Accepted: 05/16/2015] [Indexed: 12/14/2022]
Abstract
Embryonic stem cells (ESCs) represent a promising cell source for regenerative medicine. Intensive research over the past 2 decades has led to the feasibility of using ESC-differentiated cells (ESC-DCs) in regenerative medicine. However, increasing evidence indicates that ESC-DCs generated by current differentiation methods may not have equivalent cellular functions to their in vivo counterparts. Recent studies have revealed that both human and mouse ESCs as well as some types of ESC-DCs lack or have attenuated innate immune responses to a wide range of infectious agents. These findings raise important concerns for their therapeutic applications since ESC-DCs, when implanted to a wound site of a patient, where they would likely be exposed to pathogens and inflammatory cytokines. Understanding whether an attenuated immune response is beneficial or harmful to the interaction between host and grafted cells becomes an important issue for ESC-based therapy. A substantial amount of recent evidence has demonstrated that the lack of innate antiviral responses is a common feature to ESCs and other types of pluripotent cells. This has led to the hypothesis that mammals may have adapted different antiviral mechanisms at different stages of organismal development. The underdeveloped innate immunity represents a unique and uncharacterized property of ESCs that may have important implications in developmental biology, immunology, and in regenerative medicine.
Collapse
Affiliation(s)
- Yan-Lin Guo
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Gordon G Carmichael
- The Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Ruoxing Wang
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Xiaoxiao Hong
- The Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Dhiraj Acharya
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Faqing Huang
- The Department of Chemistry and Biochemistry, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| | - Fengwei Bai
- The Department of Biological Sciences, University of Southern Mississippi, Hattiesburg, Mississippi, USA
| |
Collapse
|
291
|
Activated Brain Endothelial Cells Cross-Present Malaria Antigen. PLoS Pathog 2015; 11:e1004963. [PMID: 26046849 PMCID: PMC4457820 DOI: 10.1371/journal.ppat.1004963] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2014] [Accepted: 05/19/2015] [Indexed: 12/31/2022] Open
Abstract
In the murine model of cerebral malaria caused by P. berghei ANKA (PbA), parasite-specific CD8+ T cells directly induce pathology and have long been hypothesized to kill brain endothelial cells that have internalized PbA antigen. We previously reported that brain microvessel fragments from infected mice cross-present PbA epitopes, using reporter cells transduced with epitope-specific T cell receptors. Here, we confirm that endothelial cells are the population responsible for cross-presentation in vivo, not pericytes or microglia. PbA antigen cross-presentation by primary brain endothelial cells in vitro confers susceptibility to killing by CD8+ T cells from infected mice. IFNγ stimulation is required for brain endothelial cross-presentation in vivo and in vitro, which occurs by a proteasome- and TAP-dependent mechanism. Parasite strains that do not induce cerebral malaria were phagocytosed and cross-presented less efficiently than PbA in vitro. The main source of antigen appears to be free merozoites, which were avidly phagocytosed. A human brain endothelial cell line also phagocytosed P. falciparum merozoites. Besides being the first demonstration of cross-presentation by brain endothelial cells, our results suggest that interfering with merozoite phagocytosis or antigen processing may be effective strategies for cerebral malaria intervention.
Collapse
|
292
|
Mouratidis PX, George AJ. Regulation of indoleamine 2,3-dioxygenase in primary human saphenous vein endothelial cells. J Inflamm Res 2015; 8:97-106. [PMID: 26056484 PMCID: PMC4446016 DOI: 10.2147/jir.s82202] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Indoleamine 2,3-dioxygenase (IDO) is an enzyme associated with the regulation of immune responses. Cytokines such as IFNγ induce its expression in endothelial cells originating from immune-privileged sites. In this study, we investigate regulators of IDO in primary endothelial cells from a non-immune-privileged site and determine whether IDO expression affects immune cell behavior. METHODS IDO expression was determined using real-time quantitative polymerase chain reaction and immunoblotting. IDO activity was estimated using an IDO enzyme assay. Primary cells were transfected using microporation, and T-cell migration was determined using a cell transmigration assay. RESULTS IDO is expressed in human saphenous vein endothelial cells after stimulation with IFNγ but not after treatment with TNFα, IL-1β, IL-2, IL-4, IL-6, or IL-10. VEGFβ and heparin negatively regulate IFNγ-driven increases in IDO. Overexpression of IDO in endothelial cells does not affect transmigration of T-cells. CONCLUSION IDO is expressed in human saphenous vein endothelial cells after stimulation with IFNγ. Heparin and angiogenesis stimulators such as VEGFβ negatively regulate its expression.
Collapse
|
293
|
Hu Y, Liu K, Yan M, Zhang Y, Wang Y, Ren L. Effects and mechanisms of icariin on atherosclerosis. Int J Clin Exp Med 2015; 8:3585-3589. [PMID: 26064253 PMCID: PMC4443087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2014] [Accepted: 02/27/2015] [Indexed: 06/04/2023]
Abstract
PURPOSE Icariin, a flavonoid isolated from the traditional Chinese herbal medicine Epimedium brevicornum Maxim, has been shown to process anti-inflammatory, antioxidative actions and anti-atherosclerosis activity in vivo and in vitro. The purpose of this study was to investigate the effects and mechanisms of icariin on atherosclerosis by human umbilical vein endothelial cells (HUVECs). METHODS The effects of icariin on the activity of HUVECs induced by oxidized low-density lipoprotein (ox-LDL) were detected by MTT assay. Then we studied the effects of icariin on the adhesion of monocyte with HUVECs induced by ox-LDL. The secretion of E-selectin, intercellular adhesion molecule (ICAM-1) and vascular cell adhesion molecule (VCAM-1) by HUVECs were measured by enzyme-linked immunosorbent assay (ELISA) method. Finally the mRNA levels of ICAM-1, VCAM-1, E-selectin of HUVECs were analyzed by real time RT-PCR. RESULTS MTT result indicated that icariin (10, 20, 40 μmol/L) could inhibit HUVECs injury induced by ox-LDL in a concentration-dependent manner (P < 0.05). The adhesion of monocyte with HUVECs induced by ox-LDL was inhibited by icariin in a concentration-dependent manner (P < 0.05). The levels of ICAM-1, VCAM-1, E-selectin of icariin groups were significantly decreased in a concentration-dependent manner compared with ox-LDL-simulated group (P < 0.05). The mRNA expressions of ICAM-1, VCAM-1, E-selectin of icariin groups were also downregulated significantly compared with ox-LDL-simulated group (P < 0.05). CONCLUSIONS Icariin can prevent atherosclerotic lesion. Its mechanism may be that it can defend against the oxidation damage to HUVECs, inhibit the adhesion of monocyte to HUVECs, and reduce the secretion and expression of adhesion molecules including ICAM-1, VCAM-1, E-selectin.
Collapse
Affiliation(s)
- Yanwu Hu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University Changchun 130021, Jilin, China
| | - Kai Liu
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University Changchun 130021, Jilin, China
| | - Mengtong Yan
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University Changchun 130021, Jilin, China
| | - Yang Zhang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University Changchun 130021, Jilin, China
| | - Yadi Wang
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University Changchun 130021, Jilin, China
| | - Liqun Ren
- Department of Experimental Pharmacology and Toxicology, School of Pharmaceutical Science, Jilin University Changchun 130021, Jilin, China
| |
Collapse
|
294
|
Howland SW, Claser C, Poh CM, Gun SY, Rénia L. Pathogenic CD8+ T cells in experimental cerebral malaria. Semin Immunopathol 2015; 37:221-31. [PMID: 25772948 DOI: 10.1007/s00281-015-0476-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/01/2015] [Indexed: 11/26/2022]
Abstract
Cerebral malaria (CM) is one the major complications occurring during malaria infection. The mechanisms leading to this syndrome are still not completely understood. Although it is clear that parasite sequestration is the key initiation factor, the downstream pathological processes are still highly debated. The experimental cerebral malaria (ECM) model, in which susceptible mice are infected with Plasmodium berghei ANKA, has led to the identification of CD8(+) T cells as the major mediator of ECM death. In this review, we discuss the recent advances and future developments in the understanding of the role of CD8(+) T cells in CM.
Collapse
Affiliation(s)
- Shanshan Wu Howland
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | | | | | | |
Collapse
|
295
|
Chen Z, Wen L, Martin M, Hsu CY, Fang L, Lin FM, Lin TY, Geary MJ, Geary GG, Zhao Y, Johnson DA, Chen JW, Lin SJ, Chien S, Huang HD, Miller YI, Huang PH, Shyy JYJ. Oxidative stress activates endothelial innate immunity via sterol regulatory element binding protein 2 (SREBP2) transactivation of microRNA-92a. Circulation 2014; 131:805-14. [PMID: 25550450 DOI: 10.1161/circulationaha.114.013675] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Oxidative stress activates endothelial innate immunity and disrupts endothelial functions, including endothelial nitric oxide synthase-derived nitric oxide bioavailability. Here, we postulated that oxidative stress induces sterol regulatory element-binding protein 2 (SREBP2) and microRNA-92a (miR-92a), which in turn activate endothelial innate immune response, leading to dysfunctional endothelium. METHODS AND RESULTS Using cultured endothelial cells challenged by diverse oxidative stresses, hypercholesterolemic zebrafish, and angiotensin II-infused or aged mice, we demonstrated that SREBP2 transactivation of microRNA-92a (miR-92a) is oxidative stress inducible. The SREBP2-induced miR-92a targets key molecules in endothelial homeostasis, including sirtuin 1, Krüppel-like factor 2, and Krüppel-like factor 4, leading to NOD-like receptor family pyrin domain-containing 3 inflammasome activation and endothelial nitric oxide synthase inhibition. In endothelial cell-specific SREBP2 transgenic mice, locked nucleic acid-modified antisense miR-92a attenuates inflammasome, improves vasodilation, and ameliorates angiotensin II-induced and aging-related atherogenesis. In patients with coronary artery disease, the level of circulating miR-92a is inversely correlated with endothelial cell-dependent, flow-mediated vasodilation and is positively correlated with serum level of interleukin-1β. CONCLUSIONS Our findings suggest that SREBP2-miR-92a-inflammasome exacerbates endothelial dysfunction during oxidative stress. Identification of this mechanism may help in the diagnosis or treatment of disorders associated with oxidative stress, innate immune activation, and endothelial dysfunction.
Collapse
Affiliation(s)
- Zhen Chen
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.).
| | - Liang Wen
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Marcy Martin
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Chien-Yi Hsu
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Longhou Fang
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Feng-Mao Lin
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Ting-Yang Lin
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - McKenna J Geary
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Greg G Geary
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Yongli Zhao
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - David A Johnson
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Jaw-Wen Chen
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Shing-Jong Lin
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Shu Chien
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Hsien-Da Huang
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Yury I Miller
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - Po-Hsun Huang
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.)
| | - John Y-J Shyy
- From Department of Medicine, School of Medicine (Z.C., L.W., M.M., L.F., T.-Y.L., M.J.C., Y.I.M., J.Y.-J.S.) and Department of Bioengineering (S.C.), University of California, San Diego; Department of Cardiovascular Sciences, Houston Methodist Medical Institute, Houston (L.F.); Biochemistry and Molecular Biology Graduate Program (M.M.) and Division of Biomedical Sciences, School of Medicine (D.A.J.), University of California, Riverside; Division of Cardiology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Cardiovascular Research Center, National Yang-Ming University, Taipei, Taiwan (C.-Y.H., J.-W.C., S.-J.L., P.-H.H.); Institute of Bioinformatics and Systems Biology and Department of Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan (F.-M.L., H.-D.H.); Department of Kinesiology and Health Sciences, California State University, San Bernardino (G.G.); and Cardiovascular Research Center, Medical School, Xi'an Jiaotong University, Xi'an, China (Y.Z., J.Y.-J.S.).
| |
Collapse
|
296
|
Shao Y, Cheng Z, Li X, Chernaya V, Wang H, Yang XF. Immunosuppressive/anti-inflammatory cytokines directly and indirectly inhibit endothelial dysfunction--a novel mechanism for maintaining vascular function. J Hematol Oncol 2014; 7:80. [PMID: 25387998 PMCID: PMC4236671 DOI: 10.1186/s13045-014-0080-6] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 10/13/2014] [Indexed: 12/14/2022] Open
Abstract
Endothelial dysfunction is a pathological status of the vascular system, which can be broadly defined as an imbalance between endothelium-dependent vasoconstriction and vasodilation. Endothelial dysfunction is a key event in the progression of many pathological processes including atherosclerosis, type II diabetes and hypertension. Previous reports have demonstrated that pro-inflammatory/immunoeffector cytokines significantly promote endothelial dysfunction while numerous novel anti-inflammatory/immunosuppressive cytokines have recently been identified such as interleukin (IL)-35. However, the effects of anti-inflammatory cytokines on endothelial dysfunction have received much less attention. In this analytical review, we focus on the recent progress attained in characterizing the direct and indirect effects of anti-inflammatory/immunosuppressive cytokines in the inhibition of endothelial dysfunction. Our analyses are not only limited to the importance of endothelial dysfunction in cardiovascular disease progression, but also expand into the molecular mechanisms and pathways underlying the inhibition of endothelial dysfunction by anti-inflammatory/immunosuppressive cytokines. Our review suggests that anti-inflammatory/immunosuppressive cytokines serve as novel therapeutic targets for inhibiting endothelial dysfunction, vascular inflammation and cardio- and cerebro-vascular diseases.
Collapse
Affiliation(s)
- Ying Shao
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Zhongjian Cheng
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Xinyuan Li
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Valeria Chernaya
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Hong Wang
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA.
| | - Xiao-feng Yang
- Department of Pharmacology, Center for Metabolic Disease Research and Cardiovascular Research Center, Temple University School of Medicine, MERB 1059, 3500 North Broad Street, Philadelphia, PA, 19140, USA. .,Department of Microbiology and Immunology, Temple University School of Medicine, Philadelphia, PA, 19140, USA.
| |
Collapse
|
297
|
Silva LMR, Vila-Viçosa MJM, Cortes HCE, Taubert A, Hermosilla C. Suitable in vitro Eimeria arloingi macromeront formation in host endothelial cells and modulation of adhesion molecule, cytokine and chemokine gene transcription. Parasitol Res 2014; 114:113-24. [DOI: 10.1007/s00436-014-4166-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 09/23/2014] [Indexed: 10/24/2022]
|
298
|
Wang S, Lei T, Zhang K, Zhao W, Fang L, Lai B, Han J, Xiao L, Wang N. Xenobiotic pregnane X receptor (PXR) regulates innate immunity via activation of NLRP3 inflammasome in vascular endothelial cells. J Biol Chem 2014; 289:30075-81. [PMID: 25202020 DOI: 10.1074/jbc.m114.578781] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Pregnane X receptor (PXR) is a member of nuclear receptor superfamily and responsible for the detoxification of xenobiotics. Our previously study demonstrated that PXR is expressed in endothelial cells (ECs) and acts as a master regulator of detoxification genes to protect ECs against xenobiotics. Vascular endothelial cells are key sentinel cells to sense the pathogens and xenobiotics. In this study, we examined the potential function of PXR in the regulation of innate immunity in vasculatures. Treatments with PXR agonists or overexpression of a constitutively active PXR in cultured ECs increased gene expression of the key pattern recognition receptors, including Toll-like receptors (TLR-2, -4, -9) and NOD-like receptors (NOD-1 and -2 and NLRP3). In particular, PXR agonism triggered the activation of NLRP3 inflammasome and the ensuing cleavage and maturation of caspase-1 and interleukin-1β (IL-1β). Conversely, selective antagonism or gene silencing of PXR abrogated NLRP3 inflammasome activation. In addition, we identified NLRP3 as a transcriptional target of PXR by using the promoter-reporter and ChIP assays. In summary, our findings revealed a novel regulatory mechanism of innate immune by PXR, which may act as a master transcription factor controlling the convergence between the detoxification of xenobiotics and the innate immunity against them.
Collapse
Affiliation(s)
- Shaolan Wang
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and
| | - Ting Lei
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and
| | - Kang Zhang
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and
| | - Wenxiang Zhao
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and
| | - Li Fang
- Institute of Cardiovascular Science, Peking University, Beijing 100191, China
| | - Baochang Lai
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and
| | - Jie Han
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and
| | - Lei Xiao
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and
| | - Nanping Wang
- From the Cardiovascular Research Center, School of Medicine, Xi'an Jiaotong University, Xi'an 710061 and Institute of Cardiovascular Science, Peking University, Beijing 100191, China
| |
Collapse
|
299
|
Firasat S, Hecker M, Binder L, Asif AR. Advances in endothelial shear stress proteomics. Expert Rev Proteomics 2014; 11:611-9. [DOI: 10.1586/14789450.2014.933673] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
300
|
Nymo S, Niyonzima N, Espevik T, Mollnes TE. Cholesterol crystal-induced endothelial cell activation is complement-dependent and mediated by TNF. Immunobiology 2014; 219:786-92. [PMID: 25053140 DOI: 10.1016/j.imbio.2014.06.006] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Accepted: 06/26/2014] [Indexed: 11/16/2022]
Abstract
Cholesterol crystals are known to be a hallmark of atherosclerosis with recent studies demonstrating deposition of these crystals in early fatty streak formation as well as penetrating the intima following plaque rupture. Inflammation has also become a central focus in atheroma development and endothelial cell activation is recognized as necessary for the recruitment of inflammatory cells to the plaque. However, the extent to which cholesterol crystals can induce inflammation and activate endothelial cells is not known. To investigate this, we developed a novel model activating human umbilical vein endothelial cells using lepirudin anticoagulated human whole blood. We found that cholesterol crystals caused a marked and dose-dependent increase in the adhesion molecules E-selectin and ICAM-1 on the surface of the endothelial cells after incubation with whole blood. There was no activation of the cells when the crystals were incubated in medium alone, or in human serum, despite substantial crystal-induced complement activation in serum. Complement inhibitors at the C3 and C5 levels reduced the whole blood induced endothelial cell activation by up to 89% (p<0.05) and abolished TNF release (p<0.01). Finally, the TNF inhibitor infliximab reduced endothelial activation to background levels (p<0.05). In conclusion, these data demonstrate that endothelial activation by cholesterol crystals is mediated by complement-dependent TNF release, and suggests that complement-inhibition might have a role in alleviating atherosclerosis-induced inflammation.
Collapse
Affiliation(s)
- Stig Nymo
- Department of Laboratory Medicine, Nordland Hospital, Bodø, Norway.
| | - Nathalie Niyonzima
- Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway; Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Terje Espevik
- Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway; Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Tom Eirik Mollnes
- Department of Laboratory Medicine, Nordland Hospital, Bodø, Norway; Institute of Clinical Medicine, University of Tromsø, Tromsø, Norway; Centre of Molecular Inflammation Research, Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway; Department of Immunology, Oslo University Hospital Rikshospitalet, Oslo, Norway; K.G. Jebsen IRC, University of Oslo, Oslo, Norway
| |
Collapse
|