401
|
Mussbacher M, Salzmann M, Brostjan C, Hoesel B, Schoergenhofer C, Datler H, Hohensinner P, Basílio J, Petzelbauer P, Assinger A, Schmid JA. Cell Type-Specific Roles of NF-κB Linking Inflammation and Thrombosis. Front Immunol 2019; 10:85. [PMID: 30778349 PMCID: PMC6369217 DOI: 10.3389/fimmu.2019.00085] [Citation(s) in RCA: 428] [Impact Index Per Article: 71.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 01/11/2019] [Indexed: 12/22/2022] Open
Abstract
The transcription factor NF-κB is a central mediator of inflammation with multiple links to thrombotic processes. In this review, we focus on the role of NF-κB signaling in cell types within the vasculature and the circulation that are involved in thrombo-inflammatory processes. All these cells express NF-κB, which mediates important functions in cellular interactions, cell survival and differentiation, as well as expression of cytokines, chemokines, and coagulation factors. Even platelets, as anucleated cells, contain NF-κB family members and their corresponding signaling molecules, which are involved in platelet activation, as well as secondary feedback circuits. The response of endothelial cells to inflammation and NF-κB activation is characterized by the induction of adhesion molecules promoting binding and transmigration of leukocytes, while simultaneously increasing their thrombogenic potential. Paracrine signaling from endothelial cells activates NF-κB in vascular smooth muscle cells and causes a phenotypic switch to a “synthetic” state associated with a decrease in contractile proteins. Monocytes react to inflammatory situations with enforced expression of tissue factor and after differentiation to macrophages with altered polarization. Neutrophils respond with an extension of their life span—and upon full activation they can expel their DNA thereby forming so-called neutrophil extracellular traps (NETs), which exert antibacterial functions, but also induce a strong coagulatory response. This may cause formation of microthrombi that are important for the immobilization of pathogens, a process designated as immunothrombosis. However, deregulation of the complex cellular links between inflammation and thrombosis by unrestrained NET formation or the loss of the endothelial layer due to mechanical rupture or erosion can result in rapid activation and aggregation of platelets and the manifestation of thrombo-inflammatory diseases. Sepsis is an important example of such a disorder caused by a dysregulated host response to infection finally leading to severe coagulopathies. NF-κB is critically involved in these pathophysiological processes as it induces both inflammatory and thrombotic responses.
Collapse
Affiliation(s)
- Marion Mussbacher
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Manuel Salzmann
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Christine Brostjan
- Department of Surgery, General Hospital, Medical University of Vienna, Vienna, Austria
| | - Bastian Hoesel
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | | | - Hannes Datler
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Philipp Hohensinner
- Division of Cardiology, Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - José Basílio
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Peter Petzelbauer
- Skin and Endothelial Research Division, Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| | - Johannes A Schmid
- Institute of Vascular Biology and Thrombosis Research, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
402
|
Nandi S, Sproul EP, Nellenbach K, Erb M, Gaffney L, Freytes DO, Brown AC. Platelet-like particles dynamically stiffen fibrin matrices and improve wound healing outcomes. Biomater Sci 2019; 7:669-682. [PMID: 30608063 PMCID: PMC6385160 DOI: 10.1039/c8bm01201f] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Native platelets perform several critical functions within the context of wound healing, including participating in initial hemostasis and interacting with fibrin at the wound site to induce clot retraction. Platelet depletion or dysfunction due to trauma or disease can inhibit robust wound healing responses. There has been a focus recently on developing synthetic, non-immunogenic platelet mimetic technologies for the purpose of augmenting hemostatic responses in cases of deficient native platelet functionality. Here we describe the application of synthetic platelet-like particles (PLPs), capable of recapitulating the deformable platelet body and fibrin specificity found in native platelets, to enhance healing outcomes. We first demonstrate PLPs mimic activated platelet morphology and induce fibrin clot retraction. During clot retraction, native platelets generate forces within a fibrin network to stiffen the fibrin matrix; therefore, we hypothesized that our PLPs will likewise be able to stiffen provisional fibrin matrices. Due to previous studies indicating that increased matrix stiffness is linked to increased cellular migration, we further hypothesize that PLP-mediated fibrin stiffening will enhance cell migration and improve healing outcomes within in vitro and in vivo models of wound healing. PLPs were found to enhance fibroblast migration in in vitro models of early wound healing and enhance healing outcomes in an in vivo murine model of wound healing. These studies demonstrate the utility of PLPs for enhancing wound repair and also provide insight into the role of native platelet-mediated clot retraction in wound healing.
Collapse
Affiliation(s)
- Seema Nandi
- Joint Department of Biomedical Engineering, North Carolina State University and The University of North Carolina at Chapel Hill, Raleigh, NC 27695, USA.
| | | | | | | | | | | | | |
Collapse
|
403
|
Schaaf MB, Houbaert D, Meçe O, Agostinis P. Autophagy in endothelial cells and tumor angiogenesis. Cell Death Differ 2019; 26:665-679. [PMID: 30692642 PMCID: PMC6460396 DOI: 10.1038/s41418-019-0287-8] [Citation(s) in RCA: 153] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/07/2019] [Accepted: 01/10/2019] [Indexed: 12/14/2022] Open
Abstract
In mammalian cells, autophagy is the major pathway for the degradation and recycling of obsolete and potentially noxious cytoplasmic materials, including proteins, lipids, and whole organelles, through the lysosomes. Autophagy maintains cellular and tissue homeostasis and provides a mechanism to adapt to extracellular cues and metabolic stressors. Emerging evidence unravels a critical function of autophagy in endothelial cells (ECs), the major components of the blood vasculature, which delivers nutrients and oxygen to the parenchymal tissue. EC-intrinsic autophagy modulates the response of ECs to various metabolic stressors and has a fundamental role in redox homeostasis and EC plasticity. In recent years moreover, genetic evidence suggests that autophagy regulates pathological angiogenesis, a hallmark of solid tumors. In the hypoxic, nutrient-deprived, and pro-angiogenic tumor microenvironment, heightened autophagy in the blood vessels is emerging as a critical mechanism enabling ECs to dynamically accommodate their higher bioenergetics demands to the extracellular environment and connect with other components of the tumor stroma through paracrine signaling. In this review, we provide an overview of the major cellular mechanisms regulated by autophagy in ECs and discuss their potential role in tumor angiogenesis, tumor growth, and response to anticancer therapy. Vascular homeostasis relies on the proper behavior of endothelial cells (ECs). Emerging evidence indicate a critical role of autophagy, a vesicular process for lysosomal degradation of cytoplasmic content, in EC biology. While EC-intrinsic autophagy promotes EC function and quiescent state through redox homeostasis and possibly metabolic control, a role for EC-associated autophagy in cancer seems more complex. ![]()
Collapse
Affiliation(s)
- Marco B Schaaf
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Diede Houbaert
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Odeta Meçe
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium
| | - Patrizia Agostinis
- Cell Death Research & Therapy (CDRT) Laboratory, Department for Cellular and Molecular Medicine, KU Leuven University of Leuven, Leuven, Belgium.
| |
Collapse
|
404
|
Hemostasis based on a novel 'two-path unifying theory' and classification of hemostatic disorders. Blood Coagul Fibrinolysis 2019; 29:573-584. [PMID: 30063477 DOI: 10.1097/mbc.0000000000000765] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
: Hemostasis is the most important protective mechanism for human survival following harmful vascular damage caused by internal disease or external injury. Physiological mechanism of hemostasis is partially understood. Hemostasis can be initiated by either intravascular injury or external bodily injury involving two different levels of damage [i.e., limited to the endothelium or combined with extravascular tissue (EVT)]. In intravascular injury, traumatic damage limited to local endothelium typically is of no consequence, but disease-induced endothelial damage associated with systemic endothelial injury seen in sepsis and other critical illnesses could cause generalized 'endotheliopathy'. It triggers no bleeding but promotes serious endothelial molecular response. If intravascular local trauma extends beyond the endothelium and into EVT, it causes intravascular 'bleeding' and initiate 'clotting' via normal hemostasis. In external bodily injury, local traumatic damage always extends to the endothelium and EVT, and triggers 'bleeding' and 'clotting'. Systemic endotheliopathy activates only unusually large von Willebrand factor multimers (ULVWF) path and mediates 'microthrombogenesis', producing 'microthrombi' strings. This partial activation of hemostasis with ULVWF path leads to vascular microthrombotic disease. But localized traumatic injury extending to the endothelium and EVT activates both ULVWF and tissue factor paths. Combined activation of ULVWF and tissue factor paths provides normal hemostasis in external bodily injury, but causes 'macrothrombus' formation in intravascular injury. This 'two-path unifying theory' concept succinctly elucidates simplified nature of hemostasis in intravascular and external bodily injuries. It also clarifies different pathogenesis of every hemorrhagic disease and thrombotic disorder related to internal vascular disease and external vascular injury.
Collapse
|
405
|
Thrombogenesis and thrombotic disorders based on 'two-path unifying theory of hemostasis': philosophical, physiological, and phenotypical interpretation. Blood Coagul Fibrinolysis 2019; 29:585-595. [PMID: 30234545 DOI: 10.1097/mbc.0000000000000769] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
: Hemostasis, endowed to human to protect lives, is a process of logical blood clotting system to prevent blood loss in vascular injury. However, the notion that deadly thrombosis occurs as a result of normal hemostasis in intravascular injury could encounter with conceptual skepticism because the term 'thrombosis' automatically conjures up as serious disease. According to 'two-path unifying theory', normal hemostasis is initiated only by vascular injury through activated unusually large von Willebrand factor (ULVWF) path and/or activated tissue factor (TF) path. When these two equally important paths are unified in normal hemostasis, clotting at external bodily injury site is initiated for wound healing, but in intravascular injury 'blood clots' is formed to produce a disease called 'thrombosis'. As microthrombi from ULVWF path and fibrin clots from TF path become unified, macrothrombus would be formed via thrombogenesis. However, if ULVWF path and TF path cannot be unified due to lone ULVWF path activation, partial hemostasis produces only microthrombi seen in endotheliopathy-associated vascular microthrombotic disease. In real life, in-vivo fibrin clot cannot be formed alone via normal hemostasis because bleeding vascular injury always activates both ULVWF and TF paths. Without vascular injury, microthrombi due to activated ULVWF path occur in ADAMTS13 deficiency in thrombotic thrombocytopenic purpura, and fibrin clots due to activated TF path occur in acute promyelocytic leukemia. These two conditions can be called pathologic hemostasis. Three thrombogenic pathways produce three thrombotic disorders, which include macrothrombosis, microthrombosis and true DIC through macrothrombogenesis, microthrombogenesis and fibrinogenesis in both physiologic and pathological hemostasis.
Collapse
|
406
|
Lion A, Esnault E, Kut E, Guillory V, Trapp-Fragnet L, Soubies SM, Chanteloup N, Niepceron A, Guabiraba R, Marc D, Eterradossi N, Trapp S, Quéré P. Chicken endothelial cells are highly responsive to viral innate immune stimuli and are susceptible to infections with various avian pathogens. Avian Pathol 2019; 48:121-134. [PMID: 30556415 DOI: 10.1080/03079457.2018.1556386] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
It is well established that the endothelium plays a prominent role in the pathogenesis of various infectious diseases in mammals. However, little is known about the role of endothelial cells (EC) as targets for avian pathogens and their contribution to the pathogenesis of infectious diseases in galliform birds. First, we explored the innate immune response of primary chicken aortic endothelial cells (pchAEC), obtained from 18-day-old embryos, to stimulation with pathogen-associated molecular patterns or recombinant chicken interferons (type I, II and III IFNs). In spite of the abundant expression of a number of innate immune receptors, marked cytokine responses to stimulation with pathogen-associated molecular patterns were only seen in pchAEC treated with the TLR3 agonist polyI:C (pI:C) and the MDA5 agonist liposome-complexed polyI:C (L-pI:C), as was assessed by quantitative PCR and luciferase-based IFN-I/NFκB reporter assays. Treatments of pchAEC with IFN-α, IFN-γ and IFN-λ resulted in STAT1-phosphorylation/activation, as was revealed by immunoblotting. Next, we demonstrated that pchAEC are susceptible to infection with a variety of poultry pathogens, including Marek's disease virus (MDV), infectious bursal disease virus (IBDV), avian pathogenic Escherichia coli (APEC) and Eimeria tenella. Our data highlight that chicken EC are potential targets for viral, bacterial and protozoan pathogens in gallinaceous poultry and may partake in the inflammatory and antimicrobial response. The pchAEC infection model used herein will allow further studies interrogating avian pathogen interactions with vascular EC. RESEARCH HIGHLIGHTS Use of a well-defined primary chicken aortic endothelial cell (pchAEC) culture model for studying avian host-pathogen interactions. pchAEC are responsive to innate immune stimulation with viral pathogen-associated molecular patterns and chicken type I, II and III interferons. pchAEC are susceptible to infections with economically important poultry pathogens, including MDV, IBDV, APEC and Eimeria tenella.
Collapse
Affiliation(s)
- Adrien Lion
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Evelyne Esnault
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Emmanuel Kut
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Vanaïque Guillory
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Laetitia Trapp-Fragnet
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Sébastien M Soubies
- b ANSES, Unité Virologie, Immunologie, Parasitologie Aviaire et Cunicole , Ploufragan , France
| | - Nathalie Chanteloup
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Alisson Niepceron
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Rodrigo Guabiraba
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Daniel Marc
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Nicolas Eterradossi
- b ANSES, Unité Virologie, Immunologie, Parasitologie Aviaire et Cunicole , Ploufragan , France
| | - Sascha Trapp
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| | - Pascale Quéré
- a INRA, Université François Rabelais, UMR1282 Infectiologie et Santé Publique , Nouzilly , France
| |
Collapse
|
407
|
Ma T, Li Z, Wang S. Production of Bioactive Recombinant Reteplase by Virus-Based Transient Expression System in Nicotiana benthamiana. FRONTIERS IN PLANT SCIENCE 2019; 10:1225. [PMID: 31649696 PMCID: PMC6791962 DOI: 10.3389/fpls.2019.01225] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Accepted: 09/04/2019] [Indexed: 05/08/2023]
Abstract
To explore a cost-effective alternative method to produce the recombinant thrombolytic drug Reteplase (rPA), a plant viral amplicon-based gene expression system was employed to transiently express bioactive Strep II-tagged recombinant rPA in Nicotiana benthamiana leaves via agro-infiltration. Several gene expression cassettes were designed, synthesized in vitro, and then cloned into Tobacco mosaic virus RNA-based overexpression vector. Codon optimization, subcellular targeting, and the effect of attached Strep-tag II were assessed to identify conditions that maximized expression levels of the recombinant rPA in tobacco leaves. We found that codon-optimized rPA with N-terminal Strep-tag II that was aimed to the endoplasmic reticulum as target provided the highest amount of biologically active protein, i.e., up to ∼50 mg from per kilogram fresh weight leaf biomass in less than 1 week. Furthermore, the recombinant rPA was conveniently purified from inoculated leaf extracts by a one-step purification procedure via the Strep-tag II. The plant-made rPA was glycosylated with molecular mass of ∼45.0 kDa, and its in vitro fibrinolysis activity was equivalent to the commercial available rPA. These results indicate that the plant viral amplicon-based system offers a simple and highly effective approach for cost-effective large-scale production of recombinant rPA.
Collapse
Affiliation(s)
- Ting Ma
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in the Western China, School of Life Science, Ningxia Universisty, Yinchuan, China
| | - Zhiying Li
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in the Western China, School of Life Science, Ningxia Universisty, Yinchuan, China
- Key Laboratory of Modern Molecular Breeding for Dominant and Special Crops in Ningxia, School of Life Science, Ningxia Universisty, Yinchuan, China
| | - Sheng Wang
- Key Laboratory of Ministry of Education for Protection and Utilization of Special Biological Resources in the Western China, School of Life Science, Ningxia Universisty, Yinchuan, China
- Key Laboratory of Modern Molecular Breeding for Dominant and Special Crops in Ningxia, School of Life Science, Ningxia Universisty, Yinchuan, China
- *Correspondence: Sheng Wang,
| |
Collapse
|
408
|
Cheng Y, Liu J, Su Y, Zhao H, Zhao Y, Wen M, Lu S, Zhang W, Wu J. Clinical Impact of Coagulation and Fibrinolysis Markers for Predicting Postoperative Venous Thromboembolism in Total Joint Arthroplasty Patients. Clin Appl Thromb Hemost 2019; 25:1076029619877458. [PMID: 31608652 PMCID: PMC6900621 DOI: 10.1177/1076029619877458] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 08/21/2019] [Accepted: 08/26/2019] [Indexed: 01/08/2023] Open
Abstract
One common complication after joint arthroplasty is venous thromboembolism (VTE). Therefore, it is essential to measure the changes in coagulation and fibrinolysis in order to predict VTE among patients who underwent joint arthroplasty. This study aimed to identify potential useful biomarkers for prognosing to VTE. This was a prospective cohort study enrolling 83 patients who underwent joint arthroplasty. The levels of d-dimer, thrombin-antithrombin complex (TAT), plasmin-α2-antiplasmin complex (PIC), soluble thrombomodulin, and tissue plasminogen activator inhibitor complex were measured on day 0 (before surgery) and days 1, 3, and 6 after surgery. Ultrasound examination was used to diagnose VTE on preoperative day 0 and postoperative day 6. A total of 35 patients developed VTE after surgery. Patients with VTE exhibited significantly higher levels of d-dimer and TAT on postoperative days 3 and 6 (all P < .05). The area under curves (AUC) of receiver operating characteristic (ROC) were 0.65 and 0.68 and 0.68 and 0.74 for d-dimer and TAT levels on postoperative days 3 and 6, respectively. The level of TAT/PIC ratio on postoperative day 6 was significantly increased among patients with VTE compared to non-VTE patients (P < .0001). In addition, the AUC of ROC, cutoff level, sensitivity, specificity, positive-predictive value, and negative-predictive value of TAT/PIC ratio were 0.78, 4.03 ng/TU, 97.14%, 33.33%, 51.52%, and 94.12%, respectively. The high sensitivity and negative predictive value of TAT/PIC ratio make it a potential prognostic index for diagnosing VTE during the early phase of postoperative joint arthroplasty.
Collapse
Affiliation(s)
- Yuying Cheng
- Department of laboratory medicine, Peking University Fourth School of
Clinical Medicine, Beijing, China
| | - Jian Liu
- Adult reconstruction department, Beijing Jishuitan Hospital, Beijing,
China
| | - Yu Su
- Department of laboratory medicine, Beijing Jishuitan Hospital, Beijing,
China
| | - Huiru Zhao
- Department of laboratory medicine, Beijing Jishuitan Hospital, Beijing,
China
| | - Yujing Zhao
- Department of laboratory medicine, Beijing Jishuitan Hospital, Beijing,
China
| | - Meng Wen
- Department of laboratory medicine, Beijing Jishuitan Hospital, Beijing,
China
| | - Shan Lu
- Department of laboratory medicine, Beijing Jishuitan Hospital, Beijing,
China
| | - Wenjie Zhang
- Department of laboratory medicine, Peking University Fourth School of
Clinical Medicine, Beijing, China
| | - Jun Wu
- Department of laboratory medicine, Peking University Fourth School of
Clinical Medicine, Beijing, China
| |
Collapse
|
409
|
Kizawa S, Ito T, Akamatsu K, Ichihara N, Nogi S, Miyamura M, Kanzaki Y, Sohmiya K, Hoshiga M. Chronic Kidney Disease as a Possible Predictor of Left Atrial Thrombogenic Milieu Among Patients with Nonvalvular Atrial Fibrillation. Am J Cardiol 2018; 122:2062-2067. [PMID: 30293657 DOI: 10.1016/j.amjcard.2018.08.058] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/22/2018] [Accepted: 08/28/2018] [Indexed: 11/17/2022]
Abstract
Patients with chronic kidney disease (CKD) experiencing atrial arrhythmia are hypothesized to have elevated CHADS2 and CHA2DS2-VASc scores, thereby predisposed to left atrial (LA) thrombus formation and subsequent thromboembolism. We examined possible association of LA thrombogenic milieu (TM) with CKD in patients with nonvalvular atrial fibrillation. A total of 581 patients (181 women; mean age, 67 years) who underwent transesophageal echocardiography were examined. Patients were divided into 4 groups based on the estimated glomerular filtration rate (eGFR) (ml/min/1.73 m2): eGFR ≥90 (n = 29), 60≤ eGFR <90 (n = 329), 30≤ eGFR <60 (n = 209), and eGFR <30 (n = 14). TM was defined as the presence of LA thrombus, dense spontaneous echo contrast, or LA appendage velocity ≤25 cm/s. Of 581 patients, 147 (25%) had TM. The prevalence of TM increased with decreasing eGFR (4%, 18%, 36%, and 86% for each group, p <0.001). Similar trends were observed for some of the clinical and echocardiographic variables including CHA2DS2-VASc score and LA size. Multivariate logistic regression analysis revealed that every 10 ml/min/1.73 m2 decrement in eGFR was a significant independent correlate of TM (odds ratio 0.80, p = 0.005), along with nonparoxysmal atrial fibrillation (AF) (odds ratio 0.45, p = 0.004), higher CHA2DS2-VASc score (odds ratio 1.24, p = 0.012), every 5 ml/m2 increment in LA volume index (odds ratio 1.57, p <0.001), and every 10% decrement in left ventricular ejection fraction (odds ratio 0.51, p <0.001). In conclusion, CKD may be a significant risk factor for LA thrombus formation in patients with nonvalvular atrial fibrillation.
Collapse
Affiliation(s)
- Shun Kizawa
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Takahide Ito
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan.
| | - Kanako Akamatsu
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Noboru Ichihara
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Shimpei Nogi
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Masatoshi Miyamura
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Yumiko Kanzaki
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Koichi Sohmiya
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| | - Masaaki Hoshiga
- Department of Cardiology, Osaka Medical College, Takatsuki, Osaka, Japan
| |
Collapse
|
410
|
Janićijević Ž, Ninkov M, Kataranovski M, Radovanović F. Poly(DL-Lactide-co-ε-Caprolactone)/Poly(Acrylic Acid) Composite Implant for Controlled Delivery of Cationic Drugs. Macromol Biosci 2018; 19:e1800322. [PMID: 30548776 DOI: 10.1002/mabi.201800322] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 11/02/2018] [Indexed: 11/09/2022]
Abstract
Poly(DL-lactide-co-ε-caprolactone)/poly(acrylic acid) implantable composite reservoirs for cationic drugs are synthesized by sequentially applying photoirradiation and liquid phase inversion. The chemical composition and microstructure of reservoirs are characterized with Fourier transform infrared spectroscopy-attenuated total reflection (FTIR-ATR) and scanning electron microscopy (SEM), respectively. Drug loading and release properties are investigated using methylene blue as the drug model. Biocompatibility of reservoirs is examined through a series of in vitro tests and an in vivo experiment of subcutaneous implantation in Dark Agouti rats. Reservoirs show good ion-exchange capacity, high water content, and fast reversible swelling with retained geometry. Results of drug loading and release reveal excellent loading efficiency and diffusion-controlled release during 2 weeks. Biocompatibility tests in vitro demonstrate the lack of implant proinflammatory potential and hindered adhesion of L929 cells on the implant surface. Implants exhibit low acute toxicity and elicit a normal acute foreign body reaction that reaches the early stages of fibrous capsule formation after 7 days.
Collapse
Affiliation(s)
- Željko Janićijević
- University of Belgrade, School of Electrical Engineering, Bulevar kralja Aleksandra 73, 11120, Belgrade, Serbia.,Institute of Technical Sciences of the Serbian Academy of Sciences and Arts, Knez Mihailova 35/IV, 11000, Belgrade, Serbia
| | - Marina Ninkov
- Department of Ecology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia
| | - Milena Kataranovski
- Department of Ecology, Institute for Biological Research "Siniša Stanković", University of Belgrade, Bulevar despota Stefana 142, 11060, Belgrade, Serbia.,Department of Physiology and Biochemistry, University of Belgrade, Studenstki trg 16, 11000, Belgrade, Serbia
| | - Filip Radovanović
- Institute of Technical Sciences of the Serbian Academy of Sciences and Arts, Knez Mihailova 35/IV, 11000, Belgrade, Serbia
| |
Collapse
|
411
|
Ibrahim A, Walsh LG, Algaali M, Satti Z, Kiernan TJ, Yan BP. Insight into the perioperative management of direct oral anticoagulants: concerns and considerations. Expert Opin Pharmacother 2018; 20:465-472. [PMID: 30521411 DOI: 10.1080/14656566.2018.1551879] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
INTRODUCTION Direct oral anticoagulants (DOACs) have gained momentum in recent years in patients requiring anticoagulation for the prevention and management of venous thromboembolism and thromboembolic events caused by atrial fibrillation. The use of these agents involves potential bleeding complications, particularly during invasive procedures. With increasing use of DOACs, adequate knowledge regarding the perioperative management of patients on DOACs has become indispensable. AREAS COVERED This review covers the indications, mechanism of action, and pharmacokinetics of DOACs and their management in different perioperative settings based on various current guidelines and practices. The role of bridging therapy with heparin and the recently developed reversal agents are also discussed. EXPERT OPINION The perioperative management of DOACs is influenced by drug pharmacokinetics, potential comorbidities of the patient and perioperative thrombotic and bleeding risk. In low bleeding risk and minor procedures, continuing DOACs seems to be safe. Interrupting DOACs in high-risk procedures might be necessary and should be based on the elimination half-life of the drug and renal function of the patient. Further research is needed to better clarify the role of recently developed reversal agents in the perioperative setting and to identify specific laboratory tests to guide the perioperative management of DOACs.
Collapse
Affiliation(s)
- Abdalla Ibrahim
- a Cardiology Department , University Hospital Limerick , Limerick , Ireland
| | - Leon G Walsh
- b Department of Surgery , University Hospital Limerick , Limerick , Ireland
| | - Mohamed Algaali
- a Cardiology Department , University Hospital Limerick , Limerick , Ireland
| | - Zahir Satti
- a Cardiology Department , University Hospital Limerick , Limerick , Ireland
| | - Thomas J Kiernan
- c Division of Cardiology, Department of Medicine , University Hospital Limerick, Graduate Entry Medical School (GEMS), University of Limerick , Limerick , Ireland
| | - Bryan P Yan
- d Division of Cardiology, Department of Medicine and Therapeutics , The Chinese University of Hong Kong, Prince of Wales Hospital , Hong Kong , China
| |
Collapse
|
412
|
Bioengineering the innate vasculature of complex organs: what have we learned so far. Curr Opin Organ Transplant 2018; 23:657-663. [DOI: 10.1097/mot.0000000000000577] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
413
|
Bekendam RH, Iyu D, Passam F, Stopa JD, De Ceunynck K, Muse O, Bendapudi PK, Garnier CL, Gopal S, Crescence L, Chiu J, Furie B, Panicot-Dubois L, Hogg PJ, Dubois C, Flaumenhaft R. Protein disulfide isomerase regulation by nitric oxide maintains vascular quiescence and controls thrombus formation. J Thromb Haemost 2018; 16:2322-2335. [PMID: 30207066 PMCID: PMC6374154 DOI: 10.1111/jth.14291] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Indexed: 12/17/2022]
Abstract
Essentials Nitric oxide synthesis controls protein disulfide isomerase (PDI) function. Nitric oxide (NO) modulation of PDI controls endothelial thrombogenicity. S-nitrosylated PDI inhibits platelet function and thrombosis. Nitric oxide maintains vascular quiescence in part through inhibition of PDI. SUMMARY: Background Protein disulfide isomerase (PDI) plays an essential role in thrombus formation, and PDI inhibition is being evaluated clinically as a novel anticoagulant strategy. However, little is known about the regulation of PDI in the vasculature. Thiols within the catalytic motif of PDI are essential for its role in thrombosis. These same thiols bind nitric oxide (NO), which is a potent regulator of vessel function. To determine whether regulation of PDI represents a mechanism by which NO controls vascular quiescence, we evaluated the effect of NO on PDI function in endothelial cells and platelets, and thrombus formation in vivo. Aim To assess the effect of S-nitrosylation on the regulation of PDI and other thiol isomerases in the vasculature. Methods and results The role of endogenous NO in PDI activity was evaluated by incubating endothelium with an NO scavenger, which resulted in exposure of free thiols, increased thiol isomerase activity, and enhanced thrombin generation on the cell membrane. Conversely, exposure of endothelium to NO+ carriers or elevation of endogenous NO levels by induction of NO synthesis resulted in S-nitrosylation of PDI and decreased surface thiol reductase activity. S-nitrosylation of platelet PDI inhibited its reductase activity, and S-nitrosylated PDI interfered with platelet aggregation, α-granule release, and thrombin generation on platelets. S-nitrosylated PDI also blocked laser-induced thrombus formation when infused into mice. S-nitrosylated ERp5 and ERp57 were found to have similar inhibitory activity. Conclusions These studies identify NO as a critical regulator of vascular PDI, and show that regulation of PDI function is an important mechanism by which NO maintains vascular quiescence.
Collapse
Affiliation(s)
- Roelof H. Bekendam
- Aix Marseille Université, INSERM UMR-S1076, Vascular Research Center Marseille, Marseille, France
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - David Iyu
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
- Departamento de Fisiología. Facultad de Medicina, Instituto Murciano de Investigación Biosanitaria (IMIB), Universidad de Murcia, Murcia, Spain
| | - Freda Passam
- St George Clinical School, University of New South Wales, Kogarah, New South Wales, Australia
| | - Jack D. Stopa
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Karen De Ceunynck
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Oluwatoyosi Muse
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Pavan K. Bendapudi
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Céline L. Garnier
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Srila Gopal
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Lydie Crescence
- Aix Marseille Université, INSERM UMR-S1076, Vascular Research Center Marseille, Marseille, France
| | - Joyce Chiu
- The Centenary Institute, NHMRC Clinical Trials Centre, Sydney Medical School, University of Sydney New South Wales, Australia
| | - Bruce Furie
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| | - Laurence Panicot-Dubois
- Aix Marseille Université, INSERM UMR-S1076, Vascular Research Center Marseille, Marseille, France
| | - Philip J. Hogg
- The Centenary Institute, NHMRC Clinical Trials Centre, Sydney Medical School, University of Sydney New South Wales, Australia
| | - Christophe Dubois
- Aix Marseille Université, INSERM UMR-S1076, Vascular Research Center Marseille, Marseille, France
| | - Robert Flaumenhaft
- Department of Hemostasis and Thrombosis, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, USA
| |
Collapse
|
414
|
Li H, Peng F, Wang D, Qiao Y, Xu D, Liu X. Layered double hydroxide/poly-dopamine composite coating with surface heparinization on Mg alloys: improved anticorrosion, endothelialization and hemocompatibility. Biomater Sci 2018; 6:1846-1858. [PMID: 29789824 DOI: 10.1039/c8bm00298c] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Magnesium (Mg) and its alloys are promising cardiovascular stent materials due to their favourable physical properties and complete degradation in vivo. However, rapid degradation and poor cytocompatibility hinder their clinical applications. To enhance the corrosion resistance and endothelialization of the AZ31 alloy, a layered double hydroxide (LDH)/poly-dopamine (PDA) composite coating (LDH/PDA) was successfully fabricated. Polarization curves and the electrochemical impedance spectroscopy Nyquist spectrum test proved that the corrosion resistance of the LDH/PDA sample was significantly improved in vitro. The LDH/PDA sample greatly improved the adherence process and the proliferation rate of human umbilical vein endothelial cells (HUVECs). After culturing for 10 days, the number of living HUVECs on the LDH/PDA sample was comparable to that on the Ti sample whereas the cells barely survived on the AZ31 or LDH coating. Furthermore, heparin was immobilized on LDH/PDA via a covalent bond (LDH/PDA/HEP). The corrosion resistance and long-term proliferation of HUVECs after the introduction of heparin were mildly decreased compared with the L/P sample, but were still greatly improved compared with AZ31, the LDH coating and the PDA coating. Furthermore, the LDH/PDA/HEP sample greatly improved the HUVEC migration rate compared with the LDH/PDA sample, and inhibited platelet adhesion which was intense on the LDH/PDA sample. Both LDH/PDA and LDH/PDA/HEP samples had a low hemolysis rate (2.52% and 0.65%, respectively) in vitro and eliminated the adverse biocompatible effects of the direct PDA coating on the AZ31 substrate in vivo. Our results suggest that the LDH/PDA composite coating with further heparinization is a promising method to modify the surface of Mg alloys by significantly improving corrosion resistance, endothelialization and hemocompatibility.
Collapse
Affiliation(s)
- Hua Li
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | | | | | | | | | | |
Collapse
|
415
|
Mojiri A, Alavi P, Jahroudi N. Von Willebrand factor contribution to pathophysiology outside of von Willebrand disease. Microcirculation 2018; 26:e12510. [PMID: 30365187 DOI: 10.1111/micc.12510] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/12/2018] [Accepted: 10/19/2018] [Indexed: 12/13/2022]
Abstract
VWF is a procoagulant protein that plays a central role in the initiation of platelets aggregate formation and thrombosis. While von Willebrand disease has long been known to result from qualitative and quantitative deficiencies of VWF, it is recently that contribution of elevated levels of VWF to various pathological conditions including thrombosis, inflammation, angiogenesis, and cancer metastasis has been appreciated. Here, we discuss contribution of elevated levels of VWF to various thrombotic and nonthrombotic pathological conditions.
Collapse
Affiliation(s)
- Anahita Mojiri
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Parnian Alavi
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| | - Nadia Jahroudi
- Department of Medicine, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
416
|
Wang A, Huen SC, Luan HH, Baker K, Rinder H, Booth CJ, Medzhitov R. Glucose metabolism mediates disease tolerance in cerebral malaria. Proc Natl Acad Sci U S A 2018; 115:11042-11047. [PMID: 30291189 PMCID: PMC6205430 DOI: 10.1073/pnas.1806376115] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Sickness behaviors are a conserved set of stereotypic responses to inflammatory diseases. We recently demonstrated that interfering with inflammation-induced anorexia led to metabolic changes that had profound effects on survival of acute inflammatory conditions. We found that different inflammatory states needed to be coordinated with corresponding metabolic programs to actuate tissue-protective mechanisms. Survival of viral inflammation required intact glucose utilization pathways, whereas survival of bacterial inflammation required alternative fuel substrates and ketogenic programs. We thus hypothesized that organismal metabolism would be important in other classes of infectious inflammation and sought to understand its role in the prototypic parasitic disease malaria. Utilizing the cerebral malaria model, Plasmodium berghei ANKA (PbA) infection in C57BL/6J male mice, we unexpectedly found that inhibition of glycolysis using 2-deoxy glucose (2DG) conferred protection from cerebral malaria. Unlike vehicle-treated animals, 2DG-treated animals did not develop cerebral malaria and survived until ultimately succumbing to fatal anemia. We did not find any differences in parasitemia or pathogen load in affected tissues. There were no differences in the kinetics of anemia. We also did not detect differences in immune infiltration in the brain or in blood-brain barrier permeability. Rather, on pathological analyses performed on the entire brain, we found that 2DG prevented the formation of thrombi and thrombotic complications. Using thromboelastography (TEG), we found that 2DG-treated animals formed clots that were significantly less strong and stable. Together, these data suggest that glucose metabolism is involved in inflammation-induced hemostasis and provide a potential therapeutic target in treatment of cerebral malaria.
Collapse
Affiliation(s)
- Andrew Wang
- Rheumatology Unit, Department of Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Sarah C Huen
- Nephrology Unit, Department of Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Harding H Luan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520
| | - Kelly Baker
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Henry Rinder
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520
- Hematology Unit, Department of Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Carmen J Booth
- Department of Comparative Medicine, Yale University School of Medicine, New Haven, CT 06520
| | - Ruslan Medzhitov
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520;
- Howard Hughes Medical Institute, Yale University School of Medicine, New Haven, CT 06520
| |
Collapse
|
417
|
Abstract
The blood vessel is part of the circulatory system, and systemic circulation provides the blood supply to all tissues. Arteries are pathways through which the blood is carried, and the capillaries have a key role in material exchange to maintain the tissue environment. Blood vessels have structures appropriate for their functions, and their sizes and cell types are different. In this review, we introduced recent studies of the microfluidic vascular models. The model structures are classified mainly as poly(dimethylsiloxane) and hydrogel microchannels and self-assembled networks. Basic phenomena and functions were realized in vascular models, including fluid shear stress, cell strain, interstitial flow, endothelial permeation, angiogenesis, and thrombosis. In some models, endothelial cells were co-cultured with smooth muscle cells, pericytes, and fibroblasts in an extracellular matrix. Examples of vascular models involving the brain, lung, liver, kidney, placenta, and cancer were also introduced.
Collapse
Affiliation(s)
- Kae Sato
- Department of Chemical and Biological Sciences, Faculty of Science, Japan Women's University
| | - Kiichi Sato
- Department of Chemistry and Chemical Biology, School of Science and Technology, Gunma University
| |
Collapse
|
418
|
Addi T, Dou L, Burtey S. Tryptophan-Derived Uremic Toxins and Thrombosis in Chronic Kidney Disease. Toxins (Basel) 2018; 10:E412. [PMID: 30322010 PMCID: PMC6215213 DOI: 10.3390/toxins10100412] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 10/09/2018] [Accepted: 10/10/2018] [Indexed: 02/07/2023] Open
Abstract
Patients with chronic kidney disease (CKD) display an elevated risk of thrombosis. Thrombosis occurs in cardiovascular events, such as venous thromboembolism, stroke, and acute coronary syndrome, and is a cause of hemodialysis vascular access dysfunction. CKD leads to the accumulation of uremic toxins, which exerts toxic effects on blood and the vessel wall. Some uremic toxins result from tryptophan metabolization in the gut through the indolic and the kynurenine pathways. An increasing number of studies are highlighting the link between such uremic toxins and thrombosis in CKD. In this review, we describe the thrombotic mechanisms induced by tryptophan-derived uremic toxins (TDUT). These mechanisms include an increase in plasma levels of procoagulant factors, induction of platelet hyperactivity, induction of endothelial dysfunction/ impairment of endothelial healing, decrease in nitric oxide (NO) bioavailability, and production of procoagulant microparticles. We focus on one important prothrombotic mechanism: The induction of tissue factor (TF), the initiator of the extrinsic pathway of the blood coagulation. This induction occurs via a new pathway, dependent on the transcription factor Aryl hydrocarbon receptor (AhR), the receptor of TDUT in cells. A better understanding of the prothrombotic mechanisms of uremic toxins could help to find novel therapeutic targets to prevent thrombosis in CKD.
Collapse
Affiliation(s)
- Tawfik Addi
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France.
- LPNSA, Département de Biologie, Université d'Oran 1 Ahmed Benbella, 31000 Oran, Algérie.
| | - Laetitia Dou
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France.
| | - Stéphane Burtey
- Aix Marseille University, INSERM, INRA, C2VN, 13005 Marseille, France.
- Centre de Néphrologie et Transplantation Rénale, AP-HM, 13005 Marseille, France.
| |
Collapse
|
419
|
Storch AS, Rocha HNM, Garcia VP, Batista GMDS, Mattos JD, Campos MO, Fuly AL, Nóbrega ACLD, Fernandes IA, Rocha NG. Oscillatory shear stress induces hemostatic imbalance in healthy men. Thromb Res 2018; 170:119-125. [DOI: 10.1016/j.thromres.2018.08.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Revised: 07/28/2018] [Accepted: 08/24/2018] [Indexed: 01/27/2023]
|
420
|
In-Ho C, Byung-Woo K, Yun-Jae P, Han-Joo L, Sok P, Namju L. Ginseng berry extract increases nitric oxide level in vascular endothelial cells and improves cGMP expression and blood circulation in muscle cells. J Exerc Nutrition Biochem 2018; 22:6-13. [PMID: 30343553 PMCID: PMC6199483 DOI: 10.20463/jenb.2018.0018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 08/24/2018] [Indexed: 12/19/2022] Open
Abstract
PURPOSE The purpose of this study was to determine whether ginseng berry extract improves blood circulation by regulating vasodilator expression in exposed to tumor necrosis factor alpha (TNF-α)-exposed endothelial cells and muscle cells. METHODS Nitric oxide (NO) and cGMP levels in human umbilical vein endothelial cells (HUVECs) and A7r5 cells exposed to ginseng berry extract were investigated. Based on the in vitro results, healthy participants were treated with ginseng berry extract for 4 weeks and then a non-invasive vascular screening device was used to confirm the improvement of blood circulation. RESULTS When TNF-α-treated cells were exposed to the ginseng berry extract, the expression levels of endothelial nitric oxide synthase (eNOS), NO, and cGMP were recovered to almost normal levels. In addition, TNF-ɑ-induced overexpression of vascular cell adhesion molecule 1 (VCAM-1), intracellular adhesion molecule 1 (ICAM-1), e-selectin, and p-selectin was lowered by ginseng berry extract. The ginseng berry extract significantly increased ankle brachial pressure index compared to placebo (p < 0.05). CONCLUSION This study confirmed that the intake of ginseng berry extract improved blood circulation and therefore, its intake would be helpful for people having problems with blood circulation.
Collapse
|
421
|
Tsukahara R, Haniu H, Matsuda Y, Tsukahara T. The AGP-PPARγ axis promotes oxidative stress and diabetic endothelial cell dysfunction. Mol Cell Endocrinol 2018; 473:100-113. [PMID: 29355589 DOI: 10.1016/j.mce.2018.01.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Revised: 12/21/2017] [Accepted: 01/16/2018] [Indexed: 12/30/2022]
Abstract
Alkyl-glycerophosphate (AGP) accumulates in atherogenic oxidized-LDL and human atherosclerotic plaques and is a potent agonist of peroxisome-proliferator-activated receptor-gamma (PPARγ). Recent studies suggest a potential regulatory role for PPARγ in endothelial nitric oxide synthase (eNOS) expression/activation and nitrogen oxide (NO) generation in the vascular endothelium. Importantly, eNOS-induced NO and advanced glycation end-products (AGEs) are involved in blood-vessel damage, and diabetic patients exhibit high serum NO and AGE levels; however, the effect of AGP on NO- and AGE-mediated endothelium dysfunction remains unknown. Investigation of the AGP-specific effects on NO- and AGE-mediated dysfunction and the underlying molecular mechanisms revealed that AGP upregulated eNOS expression and NO production, and that eNOS silencing and PPARγ antagonism inhibited AGP-mediated eNOS upregulation and NO production. Moreover, AGP-PPARγ-axis-mediated NO production promoted the generation of reactive oxygen species and AGE formation. These results suggested that AGP plays a significant role in the initiation/progression of diabetes-related atherosclerosis through PPARγ activation.
Collapse
Affiliation(s)
- Ryoko Tsukahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan
| | - Hisao Haniu
- Institute for Biomedical Sciences, Shinshu University Interdisciplinary Cluster for Cutting Edge Research 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Yoshikazu Matsuda
- Clinical Pharmacology Educational Center, Nihon Pharmaceutical University, Ina-machi, Saitama 362-0806, Japan
| | - Tamotsu Tsukahara
- Department of Pharmacology and Therapeutic Innovation, Nagasaki University Graduate School of Biomedical Sciences, 1-14 Bunkyo-machi, Nagasaki 852-8521, Japan.
| |
Collapse
|
422
|
Sheng Z, Xu Y, Wang S, Yuan Y, Huang T, Lu P. XPO1-mediated nuclear export of RNF146 protects from angiotensin II-induced endothelial cellular injury. Biochem Biophys Res Commun 2018; 503:1544-1549. [PMID: 30029878 DOI: 10.1016/j.bbrc.2018.07.077] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 01/03/2023]
Abstract
Endothelial cells death induced by angiotensin II (Ang II) plays a role in vascular injury. RNF146 is identified as a E3 ubiquitin ligase, which promotes cell survival under many types of stresses. However, the role of RNF146 in endothelial cellular injury is unknown. In human umbilical vein endothelial cells (HUVECs), Ang II treatment led to cell death by oxidative stress and promoted RNF146 to accumulate in nucleus in time dependent manner. Nuclear export signal was found in the RNF146's sequence. The interaction between RNF146 and XPO1 was further confirmed by co-immunoprecipitation. Inhibition of XPO1 with KPT-185 increased the level of RNF146 in nucleus. The expression of XPO1 was suppressed responding to Ang II treatment. Overexpression of XPO1 facilitated the nuclear shuttling of RNF146, which protected from Ang II-induced cell death. Moreover, overexpression of RNF146 in HUVECs reduced the cell death induced by Ang II, whereas inhibition of XPO1 abolished the protective effect of RNF146. Therefore, our data demonstrated that RNF146 was a protective factor against cell death induced by AngII in human endothelial cells, which was dependent on XPO1-mediated nuclear export.
Collapse
Affiliation(s)
- Zhiyong Sheng
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Yun Xu
- Department of Emergency, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Shu Wang
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Ying Yuan
- Intensive Care Unit, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Tieqiu Huang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China
| | - Peng Lu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, China.
| |
Collapse
|
423
|
Sakamoto A, Torii S, Jinnouchi H, Virmani R, Finn AV. Histopathologic and physiologic effect of overlapping vs single coronary stents: impact of stent evolution. Expert Rev Med Devices 2018; 15:665-682. [DOI: 10.1080/17434440.2018.1515012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
| | - Sho Torii
- CVPath Institute, Gaithersburg, MD, USA
| | | | | | - Aloke V. Finn
- CVPath Institute, Gaithersburg, MD, USA
- School of Medicine, University of Maryland, Baltimore, MD, USA
| |
Collapse
|
424
|
Saito Y, Kitahara H, Matsumiya G, Kobayashi Y. Preoperative endothelial function and long-term cardiovascular events in patients undergoing cardiovascular surgery. Heart Vessels 2018; 34:318-323. [DOI: 10.1007/s00380-018-1248-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2018] [Accepted: 08/17/2018] [Indexed: 01/09/2023]
|
425
|
The histone methyltransferase SETD1A regulates thrombomodulin transcription in vascular endothelial cells. BIOCHIMICA ET BIOPHYSICA ACTA-GENE REGULATORY MECHANISMS 2018; 1861:752-761. [DOI: 10.1016/j.bbagrm.2018.06.004] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 06/12/2018] [Accepted: 06/15/2018] [Indexed: 11/21/2022]
|
426
|
Budnik I, Brill A. Immune Factors in Deep Vein Thrombosis Initiation. Trends Immunol 2018; 39:610-623. [PMID: 29776849 PMCID: PMC6065414 DOI: 10.1016/j.it.2018.04.010] [Citation(s) in RCA: 132] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Revised: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/11/2022]
Abstract
Deep vein thrombosis (DVT) is a major origin of morbidity and mortality. While DVT has long been considered as blood coagulation disorder, several recent lines of evidence demonstrate that immune cells and inflammatory processes are involved in DVT initiation. Here, we discuss these mechanisms, in particular, the role of immune cells in endothelial activation, and the immune cascades leading to expression of adhesion receptors on endothelial cells. We analyze the specific recruitment and functional roles of different immune cells, such as mast cells and leukocytes, in DVT. Importantly, we also speculate how immune modulation could be used for DVT prevention with a lower risk of bleeding complications than conventional therapeutic approaches.
Collapse
Affiliation(s)
- Ivan Budnik
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Alexander Brill
- Department of Pathophysiology, Sechenov First Moscow State Medical University, Moscow, Russia; Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
427
|
Malloy J, Zhuang D, Kim T, Inskeep P, Kim D, Taylor K. Single and multiple dose evaluation of a novel MetAP2 inhibitor: Results of a randomized, double-blind, placebo-controlled clinical trial. Diabetes Obes Metab 2018; 20:1878-1884. [PMID: 29577550 PMCID: PMC6055687 DOI: 10.1111/dom.13305] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Revised: 03/11/2018] [Accepted: 03/20/2018] [Indexed: 11/29/2022]
Abstract
AIMS Methionine aminopeptidase 2 (MetAP2) inhibition has been shown to result in significant weight loss and improved glucose control. This Phase 1 clinical trial assessed the safety and tolerability, pharmacokinetics and preliminary efficacy of a novel MetAP2 inhibitor, ZGN-1061. METHODS This clinical trial included a single ascending dose (SAD) phase in healthy subjects (BMI, 23 to <30 kg/m2 ) and a multiple ascending dose (MAD) phase in otherwise healthy subjects (BMI, 27 to 40 kg/m2 ). SAD phase doses, administered subcutaneously (SC), were 0.2, 0.6, 1.2, 2.4, 3.6 and 4.8 mg and the MAD phase evaluated doses of 0.2, 0.6 and 1.8 mg twice weekly SC for 4 weeks. RESULTS The SAD phase included 39 subjects (ZGN-1061, N = 28; placebo, N = 11); 90% were male and BMI was 26.4 kg/m2 . ZGN-1061 was well tolerated across all doses, with the most frequent adverse events being mild headache and procedural-related irritation. There were no severe or serious adverse events. All doses of ZGN-1061 were rapidly absorbed and cleared, resulting in short duration of exposure that is anticipated to minimize potential off-drug target risks. The MAD phase included 29 subjects (ZGN-1061, N = 22; placebo, N = 7); 76% were male and BMI was 33.5 kg/m2 . Safety observations were consistent with SAD findings. Efficacy measures in the MAD phase indicated trends for weight change (-1.5 kg total ZGN-1061 vs -0.2 kg placebo) and other biomarker changes. CONCLUSIONS ZGN-1061 was well tolerated with no safety signals in all doses tested. In addition, the desired pharmacokinetic profile and preliminary efficacy observations with ZGN-1061 support evaluation in larger and longer clinical trials.
Collapse
|
428
|
Pryzdial ELG, Lee FMH, Lin BH, Carter RLR, Tegegn TZ, Belletrutti MJ. Blood coagulation dissected. Transfus Apher Sci 2018; 57:449-457. [PMID: 30049564 DOI: 10.1016/j.transci.2018.07.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Hemostasis is the physiological control of bleeding and is initiated by subendothelial exposure. Platelets form the primary vascular seal in three stages (localization, stimulation and aggregation), which are triggered by specific interactions between platelet surface receptors and constituents of the subendothelial matrix. As a secondary hemostatic plug, fibrin clot formation is initiated and feedback-amplified to advance the seal and stabilize platelet aggregates comprising the primary plug. Once blood leakage has been halted, the fibrinolytic pathway is initiated to dissolve the clot and restore normal blood flow. Constitutive and induced anticoagulant and antifibrinolytic pathways create a physiological balance between too much and too little clot production. Hemostatic imbalance is a major burden to global healthcare, resulting in thrombosis or hemorrhage.
Collapse
Affiliation(s)
- Edward L G Pryzdial
- Centre for Innovation, Canadian Blood Services, Ottawa, ON, Canada; Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada.
| | - Frank M H Lee
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Bryan H Lin
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Rolinda L R Carter
- Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Tseday Z Tegegn
- Centre for Innovation, Canadian Blood Services, Ottawa, ON, Canada; Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada; Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Mark J Belletrutti
- Pediatric Hematology, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
429
|
Weber M, Steinle H, Golombek S, Hann L, Schlensak C, Wendel HP, Avci-Adali M. Blood-Contacting Biomaterials: In Vitro Evaluation of the Hemocompatibility. Front Bioeng Biotechnol 2018; 6:99. [PMID: 30062094 PMCID: PMC6054932 DOI: 10.3389/fbioe.2018.00099] [Citation(s) in RCA: 380] [Impact Index Per Article: 54.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2018] [Accepted: 06/26/2018] [Indexed: 12/15/2022] Open
Abstract
Hemocompatibility of blood-contacting biomaterials is one of the most important criteria for their successful in vivo applicability. Thus, extensive in vitro analyses according to ISO 10993-4 are required prior to clinical applications. In this review, we summarize essential aspects regarding the evaluation of the hemocompatibility of biomaterials and the required in vitro analyses for determining the blood compatibility. Static, agitated, or shear flow models are used to perform hemocompatibility studies. Before and after the incubation of the test material with fresh human blood, hemolysis, cell counts, and the activation of platelets, leukocytes, coagulation and complement system are analyzed. Furthermore, the surface of biomaterials are evaluated concerning attachment of blood cells, adsorption of proteins, and generation of thrombus and fibrin networks.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Meltem Avci-Adali
- Department of Thoracic and Cardiovascular Surgery, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
430
|
Endothelial Cell Aging: How miRNAs Contribute? J Clin Med 2018; 7:jcm7070170. [PMID: 29996516 PMCID: PMC6068727 DOI: 10.3390/jcm7070170] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 07/04/2018] [Accepted: 07/09/2018] [Indexed: 12/19/2022] Open
Abstract
Endothelial cells (ECs) form monolayers and line the interior surfaces of blood vessels in the entire body. In most mammalian systems, the capacity of endothelial cells to divide is limited and endothelial cells are prone to be senescent. Aging of ECs and resultant endothelial dysfunction lead to a variety of vascular diseases such as atherosclerosis, diabetes mellites, hypertension, and ischemic injury. However, the mechanism by which ECs get old and become senescent and the impact of endothelial senescence on the vascular function are not fully understood. Recent research has unveiled the crucial roles of miRNAs, which are small non-coding RNAs, in regulating endothelial cellular functions, including nitric oxide production, vascular inflammation, and anti-thromboformation. In this review, how senescent-related miRNAs are involved in controlling the functions of ECs will be discussed.
Collapse
|
431
|
Xiao F, Wang D, Kong L, Li M, Feng Z, Shuai B, Wang L, Wei Y, Li H, Wu S, Tan C, Zhao H, Hu X, Liu J, Kang Y, Liao X, Zhou Y, Zhang W. Intermedin protects against sepsis by concurrently re-establishing the endothelial barrier and alleviating inflammatory responses. Nat Commun 2018; 9:2644. [PMID: 29980671 PMCID: PMC6035189 DOI: 10.1038/s41467-018-05062-2] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2017] [Accepted: 06/11/2018] [Indexed: 02/05/2023] Open
Abstract
Sepsis is a life-threatening condition caused by dysregulated host responses to infection. Widespread vascular hyperpermeability and a “cytokine storm” are two pathophysiological hallmarks of sepsis. Here, we show that intermedin (IMD), a member of the calcitonin family, alleviates organ injury and decreases mortality in septic mice by concurrently alleviating vascular leakage and inflammatory responses. IMD promotes the relocation of vascular endothelial cadherin through a Rab11-dependent pathway to dynamically repair the disrupted endothelial junction. Additionally, IMD decreases inflammatory responses by reducing macrophage infiltration via downregulating CCR2 expression. IMD peptide administration ameliorates organ injuries and significantly improves the survival of septic mice, and the experimental results correlate with the clinical data. Patients with high IMD levels exhibit a lower risk of shock, lower severity scores, and greatly improved survival outcomes than those with low IMD levels. Based on our data, IMD may be an important self-protective factor in response to sepsis. Sepsis is a life-threatening condition. Here, the authors show that intermedin alleviates organ injury and decreases mortality in septic mice by concurrently alleviating vascular leakage and inflammatory responses. Patients with high intermedin levels exhibit a low risk of shock, lower severity scores, and greatly improved survival outcomes.
Collapse
Affiliation(s)
- Fei Xiao
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.,Department of Intensive Care Unit of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Denian Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lingmiao Kong
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Min Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhongxue Feng
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Bingxing Shuai
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Lijun Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yong'gang Wei
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hongyu Li
- Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Sisi Wu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Chun Tan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Huan Zhao
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuejiao Hu
- Department of Laboratory Medicine, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jin Liu
- Department of Anesthesiology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Kang
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xuelian Liao
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan Zhou
- Department of Intensive Care Unit, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wei Zhang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
432
|
Abstract
Disseminated intravascular coagulation (DIC) is a serious secondary disease that alters the events in the clotting cascade. DIC leads to microclots in the peripheral vasculature. The uncontrolled formation of these clots consumes the body's clotting factors, which precipitates bleeding. Neonates, defined as newborns zero to one month old, are born with slightly altered hemostasis, also known as developmental coagulopathy. This coagulopathy puts neonates at a greater risk of developing DIC, specifically when additional body systems have been compromised by a primary disease. Because of the serious nature of this disease and its precipitating factors, advanced practitioners should study the nuances of DIC to provide accurate diagnosis and proper treatment. The aim of this article is to discuss the pathophysiology behind DIC along with etiologies, diagnosis, treatment, and prognosis in neonates, along with gaps in current research.
Collapse
|
433
|
|
434
|
Chatterjee S. Endothelial Mechanotransduction, Redox Signaling and the Regulation of Vascular Inflammatory Pathways. Front Physiol 2018; 9:524. [PMID: 29930512 PMCID: PMC5999754 DOI: 10.3389/fphys.2018.00524] [Citation(s) in RCA: 116] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 04/24/2018] [Indexed: 12/13/2022] Open
Abstract
The endothelium that lines the interior of blood vessels is directly exposed to blood flow. The shear stress arising from blood flow is “sensed” by the endothelium and is “transduced” into biochemical signals that eventually control vascular tone and homeostasis. Sensing and transduction of physical forces occur via signaling processes whereby the forces associated with blood flow are “sensed” by a mechanotransduction machinery comprising of several endothelial cell elements. Endothelial “sensing” involves converting the physical cues into cellular signaling events such as altered membrane potential and activation of kinases, which are “transmission” signals that cause oxidant production. Oxidants produced are the “transducers” of the mechanical signals? What is the function of these oxidants/redox signals? Extensive data from various studies indicate that redox signals initiate inflammation signaling pathways which in turn can compromise vascular health. Thus, inflammation, a major response to infection or endotoxins, can also be initiated by the endothelium in response to various flow patterns ranging from aberrant flow to alteration of flow such as cessation or sudden increase in blood flow. Indeed, our work has shown that endothelial mechanotransduction signaling pathways participate in generation of redox signals that affect the oxidant and inflammation status of cells. Our goal in this review article is to summarize the endothelial mechanotransduction pathways that are activated with stop of blood flow and with aberrant flow patterns; in doing so we focus on the complex link between mechanical forces and inflammation on the endothelium. Since this “inflammation susceptible” phenotype is emerging as a trigger for pathologies ranging from atherosclerosis to rejection post-organ transplant, an understanding of the endothelial machinery that triggers these processes is very crucial and timely.
Collapse
Affiliation(s)
- Shampa Chatterjee
- Department of Physiology, Perelman School of Medicine, Institute for Environmental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
435
|
Abstract
PURPOSE OF REVIEW Statins reduce cholesterol synthesis and promote low-density lipoprotein clearance from circulation. Beyond their cholesterol-lowering action, statins may interfere with haemostasis. This review aims to provide an update on the impact of statin treatment on markers of haemostasis and platelet function and on thrombosis-related outcomes. RECENT FINDINGS Different coagulation factors are modulated by statins, leading to inhibition of coagulation and increased fibrinolysis. Also, an impact of statins on platelet function has been documented. From a clinical perspective, several observational studies have revealed a reduced incidence of venous thromboembolism in patients receiving statins, which has been argued in some available studies and meta-analyses. Furthermore, a beneficial effect of early statin initiation following acute coronary syndrome for short-term prevention of thrombosis-related events has been documented, but the available data are still not consistent. SUMMARY Although statins influence the levels of a multitude of haemostatic factors in an antithrombotic direction, data supporting their use for venous thromboembolism prevention are not consistent, and the impact of statins on early vascular events following acute coronary syndrome is still debated. Whether the robust long-term beneficial effects of statins in reducing cardiovascular risk may be also explained by persistent changes in haemostatic factors needs further exploration.
Collapse
|
436
|
Ping S, Qiu X, Gonzalez-Toledo ME, Liu X, Zhao LR. Stem Cell Factor in Combination with Granulocyte Colony-Stimulating Factor reduces Cerebral Capillary Thrombosis in a Mouse Model of CADASIL. Cell Transplant 2018; 27:637-647. [PMID: 29871518 PMCID: PMC6041883 DOI: 10.1177/0963689718766460] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Cerebral autosomal dominant arteriopathy with subcortical infarcts and
leucoencephalopathy (CADASIL) is a cerebral small vascular disease caused by NOTCH3
mutation-induced vascular smooth muscle cell (VSMC) degeneration, leading to ischemic
stroke and vascular dementia. Our previous study has demonstrated that repeated treatment
with a combination of stem cell factor (SCF) and granulocyte colony-stimulating factor
(G-CSF) reduces VSMC degeneration and cerebral endothelial cell (EC) damage and improves
cognitive function in a mouse model of CADASIL (TgNotch3R90C). This study aimed to
determine whether cerebral thrombosis occurs in TgNotch3R90C mice and whether repeated
SCF+G-CSF treatment reduces cerebral thrombosis in TgNotch3R90C mice. Using the approaches
of bone marrow transplantation to track bone marrow-derived cells and confocal imaging, we
observed bone marrow-derived blood cell occlusion in cerebral small vessels and
capillaries (thrombosis). Most thrombosis occurred in the cerebral capillaries (93% of
total occluded vessels), and the thrombosis showed an increased frequency in the regions
of capillary bifurcation. Degenerated capillary ECs were seen inside and surrounding the
thrombosis, and the bone marrow-derived ECs were also found next to the thrombosis. IgG
extravasation was seen in and next to the areas of thrombosis. SCF+G-CSF treatment
significantly reduced cerebral capillary thrombosis and IgG extravasation. These data
suggest that the EC damage is associated with thrombosis and blood–brain barrier leakage
in the cerebral capillaries under the CADASIL-like condition, whereas SCF+G-CSF treatment
diminishes these pathological alterations. This study provides new insight into the
involvement of cerebral capillary thrombosis in the development of CADASIL and potential
approaches to reduce the thrombosis, which may restrict the pathological progression of
CADASIL.
Collapse
Affiliation(s)
- Suning Ping
- 1 Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, New York, NY, USA
| | - Xuecheng Qiu
- 1 Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, New York, NY, USA
| | - Maria E Gonzalez-Toledo
- 2 Departments of Neurology, Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Xiaoyun Liu
- 2 Departments of Neurology, Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Li-Ru Zhao
- 1 Department of Neurosurgery, State University of New York, Upstate Medical University, Syracuse, New York, NY, USA.,2 Departments of Neurology, Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| |
Collapse
|
437
|
Crochemore T, Savioli FA. Thromboelastometry-guided hemostatic therapy for hemorrhagic shock in the postoperative period of vascular surgery: a case report. J Med Case Rep 2018; 12:153. [PMID: 29859130 PMCID: PMC5984768 DOI: 10.1186/s13256-018-1661-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/19/2018] [Indexed: 12/23/2022] Open
Abstract
Background Hemorrhagic shock is a medical emergency that often complicates vascular surgery and can lead to death. Hemorrhagic shock is characterized by hypoperfusion and hemodynamic abnormalities leading to the collapse of homeostasis due to massive blood loss. Early diagnosis is critical for a favorable outcome. Thromboelastometry has been considered an effective tool for bleeding management in critically ill patients. Thromboelastometry can guide transfusion therapy quickly, reducing the need for blood products. Therefore, it could be an alternative test to guide hemostatic therapy in complex cases of hemorrhagic shock as a result of vascular surgeries. We report our successful experience with a case of hemorrhagic shock in postoperative care in vascular surgery, in which bleeding management was guided by thromboelastometry and bleeding control was achieved with hemostatic drugs and coagulation factor concentrates. Case presentation We report a case of an 82-year-old Afro-Brazilian man who presented to the intensive care unit with hemorrhagic shock in the postoperative period of vascular surgery. He underwent surgery for correction of iliac artery aneurysm with endoleak. His laboratory tests revealed severe anemia (hemoglobin 7.4 mg/dL), metabolic acidosis (bicarbonate 10 mEq/L, pH 7.11), acute kidney injury (creatinine 3.1 mg/dL), thrombocytopenia (platelets count 83 × 103/mm3), hypofibrinogenemia (70 mg/dL), international nationalized ratio 1.95, activated partial thromboplastin time 64.5 seconds, and lactate 87 mmol/L. There was active bleeding in surgical site. Bleeding management was guided by thromboelastometry. The first test showed fulminant hyperfibrinolysis, which was corrected with the administration of tranexamic acid. The second thromboelastometry test showed improvement of hyperfibrinolysis but severe hypocoagulability. Fibrinogen concentrate, platelet apheresis, cryoprecipitate, and prothrombin complex concentrate were sequentially administrated. Thromboelastometry was completely corrected after 2 hours. Arteriography to evaluate mechanical cause of bleeding was normal. No more bleeding was identified, and neither was any further transfusion needed. He was discharged from the intensive care unit from the ward 3 days after admission. Conclusions Thromboelastometry may be considered a useful, feasible and safe tool to monitor and manage coagulopathy in patients with hemorrhagic shock. Moreover, it has the potential benefit of allowing rapid diagnosis, goal-directed therapy with hemostatic drugs and coagulation factor concentrates and thus, avoiding unnecessary blood component transfusion.
Collapse
Affiliation(s)
- Tomaz Crochemore
- Hospital Israelita Albert Einstein - Intensive Care Unit, Av. Albert Einstein, 627, Morumbi, São Paulo, SP, CEP: 05651-901, Brazil.
| | - Felicio A Savioli
- Hospital Israelita Albert Einstein - Intensive Care Unit, Av. Albert Einstein, 627, Morumbi, São Paulo, SP, CEP: 05651-901, Brazil
| |
Collapse
|
438
|
Hussain MA, Al-Omran M, Creager MA, Anand SS, Verma S, Bhatt DL. Antithrombotic Therapy for Peripheral Artery Disease. J Am Coll Cardiol 2018; 71:2450-2467. [DOI: 10.1016/j.jacc.2018.03.483] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 03/16/2018] [Accepted: 03/21/2018] [Indexed: 12/18/2022]
|
439
|
Verma S, Mathew V, Farkouh ME. Targeting Inflammation in the Prevention and Treatment of Type 2 Diabetes. J Am Coll Cardiol 2018; 71:2402-2404. [DOI: 10.1016/j.jacc.2018.03.480] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 03/22/2018] [Accepted: 03/26/2018] [Indexed: 11/28/2022]
|
440
|
Burkey BF, Hoglen NC, Inskeep P, Wyman M, Hughes TE, Vath JE. Preclinical Efficacy and Safety of the Novel Antidiabetic, Antiobesity MetAP2 Inhibitor ZGN-1061. J Pharmacol Exp Ther 2018; 365:301-313. [PMID: 29491038 DOI: 10.1124/jpet.117.246272] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 02/23/2018] [Indexed: 01/12/2023] Open
Abstract
Methionine aminopeptidase 2 (MetAP2) inhibition is a promising approach to treating diabetes, obesity, and associated metabolic disorders. Beloranib, a MetAP2 inhibitor previously investigated for treatment of Prader-Willi syndrome, was associated with venous thrombotic adverse events likely resulting from drug effects on vascular endothelial cells (ECs). Here, we report the pharmacological characterization of ZGN-1061, a novel MetAP2 inhibitor being investigated for treatment of diabetes and obesity. Four weeks of subcutaneous administration of ZGN-1061 to diet-induced obese (DIO) insulin-resistant mice produced a 25% reduction in body weight, primarily due to reduced fat mass, that was comparable to beloranib. ZGN-1061 also produced improvements in metabolic parameters, including plasma glucose and insulin, and, in HepG2 cells, initiated gene changes similar to beloranib that support observed in vivo pharmacodynamics. In vitro studies in ECs demonstrated that ZGN-1061 effects on EC proliferation and coagulation proteins were greatly attenuated, or absent, relative to beloranib, due to lower intracellular drug concentrations, shorter half-life of inhibitor-bound MetAP2 complex, and reduced cellular enzyme inhibition. In dogs, ZGN-1061 was more rapidly absorbed and cleared, with a shorter half-life than beloranib. Unlike beloranib, ZGN-1061 did not increase coagulation markers in dogs, and ZGN-1061 had a greatly improved safety profile in rats relative to beloranib. In conclusion, ZGN-1061 and beloranib demonstrated similar efficacy in a mouse model of obesity, while ZGN-1061 had a markedly improved safety profile in multiple in vitro and in vivo models. The lower duration of exposure characteristic of ZGN-1061 is expected to provide a meaningfully enhanced clinical safety profile.
Collapse
Affiliation(s)
- Bryan F Burkey
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Niel C Hoglen
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Philip Inskeep
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Margaret Wyman
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - Thomas E Hughes
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| | - James E Vath
- Zafgen, Inc., Boston, Massachusetts (B.F.B., N.C.H., M.W., T.E.H., J.E.V.) and InskeepDMPK, LLC (P.I.), East Lyme, Connecticut
| |
Collapse
|
441
|
Barrile R, van der Meer AD, Park H, Fraser JP, Simic D, Teng F, Conegliano D, Nguyen J, Jain A, Zhou M, Karalis K, Ingber DE, Hamilton GA, Otieno MA. Organ-on-Chip Recapitulates Thrombosis Induced by an anti-CD154 Monoclonal Antibody: Translational Potential of Advanced Microengineered Systems. Clin Pharmacol Ther 2018; 104:1240-1248. [PMID: 29484632 DOI: 10.1002/cpt.1054] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 01/24/2018] [Accepted: 02/14/2018] [Indexed: 12/18/2022]
Abstract
Clinical development of Hu5c8, a monoclonal antibody against CD40L intended for treatment of autoimmune disorders, was terminated due to unexpected thrombotic complications. These life-threatening side effects were not discovered during preclinical testing due to the lack of predictive models. In the present study, we describe the development of a microengineered system lined by human endothelium perfused with human whole blood, a "Vessel-Chip." The Vessel-Chip allowed us to evaluate key parameters in thrombosis, such as endothelial activation, platelet adhesion, platelet aggregation, fibrin clot formation, and thrombin anti-thrombin complexes in the Chip-effluent in response to Hu5c8 in the presence of soluble CD40L. Importantly, the observed prothrombotic effects were not observed with Hu5c8-IgG2σ designed with an Fc domain that does not bind the FcγRIIa receptor, suggesting that this approach may have a low potential risk for thrombosis. Our results demonstrate the translational potential of Organs-on-Chips, as advanced microengineered systems to better predict human response.
Collapse
Affiliation(s)
| | | | | | | | - Damir Simic
- Janssen Pharmaceutical Research and Development, Discovery & Manufacturing Sciences, Spring House, Pennsylvania, USA
| | - Fang Teng
- Janssen Pharmaceutical Research and Development, Discovery & Manufacturing Sciences, Spring House, Pennsylvania, USA
| | | | | | - Abhishek Jain
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | - Mimi Zhou
- Janssen Pharmaceutical Research and Development, Discovery & Manufacturing Sciences, Spring House, Pennsylvania, USA
| | | | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering at Harvard University, Boston, Massachusetts, USA
| | | | - Monicah A Otieno
- Janssen Pharmaceutical Research and Development, Discovery & Manufacturing Sciences, Spring House, Pennsylvania, USA
| |
Collapse
|
442
|
Nakao T, Yasumoto A, Tokuoka S, Kita Y, Kawahara T, Daimon M, Yatomi Y. The impact of night-shift work on platelet function in healthy medical staff. J Occup Health 2018; 60:324-332. [PMID: 29669967 PMCID: PMC6078842 DOI: 10.1539/joh.2018-0027-fs] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Objectives: Rotating shift work has been reported to increase the risk of cardiovascular diseases. Vascular endothelial dysfunction and platelet activation are among the leading causes of thrombus formation in patients with myocardial infarction or stroke. Endothelial function has been shown to be impaired immediately after night-shift work; however, it is not known whether platelets are also activated. The aim of this study was to investigate the acute impact of night-shift work on platelet function. Methods: This observational study included 11 healthy medical staff members (seven women, median age 32 years). We examined each subject's platelet aggregation rates and the serum concentrations of eicosanoid mediators after night-shift work and on day-shift work without preceding night-shift work (baseline). Results: Platelet aggregation did not differ from baseline levels after night-shift work. However, serum cyclooxygenase (COX)-metabolized eicosanoid mediators, particularly thromboxane (Tx) B2 (a stable metabolite of TxA2 and the most important marker of platelet activation), were significantly higher after the night-shift than at baseline (median 65.3 vs 180.4 ng/ml). Conclusions: Although platelet aggregation did not increase, there was an increase in serum COX-metabolized eicosanoid mediators such as TxB2 in healthy medical staff after night-shift work. This platelet hypersensitivity may be one of the mechanisms underlying the significant association between night-shift work and adverse cardiovascular outcomes.
Collapse
Affiliation(s)
- Tomoko Nakao
- Department of Clinical Laboratory, The University of Tokyo Hospital
| | - Atsushi Yasumoto
- Department of Clinical Laboratory, The University of Tokyo Hospital
| | - Suzumi Tokuoka
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo
| | - Yoshihiro Kita
- Department of Lipidomics, Graduate School of Medicine, The University of Tokyo
| | - Takuya Kawahara
- Biostatistics Division, Clinical Research Support Center, The University of Tokyo Hospital
| | - Masao Daimon
- Department of Clinical Laboratory, The University of Tokyo Hospital
| | - Yutaka Yatomi
- Department of Clinical Laboratory, The University of Tokyo Hospital
| |
Collapse
|
443
|
Wersäll A, Williams CM, Brown E, Iannitti T, Williams N, Poole AW. Mouse Platelet Ral GTPases Control P-Selectin Surface Expression, Regulating Platelet-Leukocyte Interaction. Arterioscler Thromb Vasc Biol 2018; 38:787-800. [PMID: 29437579 DOI: 10.1161/atvbaha.117.310294] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2017] [Accepted: 01/25/2018] [Indexed: 01/28/2023]
Abstract
OBJECTIVE RalA and RalB GTPases are important regulators of cell growth, cancer metastasis, and granule secretion. The purpose of this study was to determine the role of Ral GTPases in platelets with the use of platelet-specific gene-knockout mouse models. APPROACH AND RESULTS This study shows that platelets from double knockout mice, in which both GTPases have been deleted, show markedly diminished (≈85% reduction) P-selectin translocation to the surface membrane, suggesting a critical role in α-granule secretion. Surprisingly, however, there were only minor effects on stimulated release of soluble α- and δ-granule content, with no alteration in granule count, morphology, or content. In addition, their expression was not essential for platelet aggregation or thrombus formation. However, absence of surface P-selectin caused a marked reduction (≈70%) in platelet-leukocyte interactions in blood from RalAB double knockout mice, suggesting a role for platelet Rals in platelet-mediated inflammation. CONCLUSIONS Platelet Ral GTPases primarily control P-selectin surface expression, in turn regulating platelet-leukocyte interaction. Ral GTPases could therefore be important novel targets for the selective control of platelet-mediated immune cell recruitment and inflammatory disease.
Collapse
Affiliation(s)
- Andreas Wersäll
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom (A.W., C.M.W., E.B., A.W.P.); and KWS Biotest, Portishead, Bristol, United Kingdom (T.I., N.W.).
| | - Chris M Williams
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom (A.W., C.M.W., E.B., A.W.P.); and KWS Biotest, Portishead, Bristol, United Kingdom (T.I., N.W.)
| | - Edward Brown
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom (A.W., C.M.W., E.B., A.W.P.); and KWS Biotest, Portishead, Bristol, United Kingdom (T.I., N.W.)
| | - Tommaso Iannitti
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom (A.W., C.M.W., E.B., A.W.P.); and KWS Biotest, Portishead, Bristol, United Kingdom (T.I., N.W.)
| | - Neil Williams
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom (A.W., C.M.W., E.B., A.W.P.); and KWS Biotest, Portishead, Bristol, United Kingdom (T.I., N.W.)
| | - Alastair W Poole
- From the School of Physiology, Pharmacology and Neuroscience, University of Bristol, United Kingdom (A.W., C.M.W., E.B., A.W.P.); and KWS Biotest, Portishead, Bristol, United Kingdom (T.I., N.W.)
| |
Collapse
|
444
|
Shukla K, Sonowal H, Saxena A, Ramana KV. Didymin prevents hyperglycemia-induced human umbilical endothelial cells dysfunction and death. Biochem Pharmacol 2018; 152:1-10. [PMID: 29548811 DOI: 10.1016/j.bcp.2018.03.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/09/2018] [Indexed: 01/01/2023]
Abstract
Although didymin, a flavonoid-O-glycosides compound naturally found in the citrus fruits, has been reported to be a potent anticancer agent in the prevention of various cancers, its role in the prevention of cardiovascular complications is unclear. Most importantly, its effect in the prevention of endothelial dysfunction, a pathological process involved in the atherogenesis, is unknown. We have examined the efficacy of didymin in preventing the high glucose (HG; 25 mM)-induced human umbilical vein endothelial cells (HUVECs) dysfunction. Our results indicate that incubation of HUVECs with HG resulted in the loss of cell viability, and pre-incubation of didymin prevented it. Further, didymin prevented the HG-induced generation of reactive oxygen species (ROS) as well as lipid peroxidation product, malondialdehyde. Pretreatment of HUVECs with didymin also prevented the HG-induced decrease in eNOS and increase in iNOS expressions. Further, didymin prevented the HG-induced monocytes cell adhesion to endothelial cells, expressions of ICAM-1 and VCAM-1 and activation of NF-κB. Didymin also prevented the release of various inflammatory cytokines and chemokines in HG-treated HUVECs. In conclusion, our results demonstrate that didymin with its anti-oxidative and anti-inflammatory actions prevents hyperglycemia-induced endothelial dysfunction and death. Thus, it could be developed as a potential natural therapeutic agent for the prevention of cardiovascular complications in diabetes.
Collapse
Affiliation(s)
- Kirtikar Shukla
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Himangshu Sonowal
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Ashish Saxena
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Kota V Ramana
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
445
|
Obermayer G, Afonyushkin T, Binder CJ. Oxidized low-density lipoprotein in inflammation-driven thrombosis. J Thromb Haemost 2018; 16:418-428. [PMID: 29316215 DOI: 10.1111/jth.13925] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Thrombosis is the defining feature of the most prevalent causes of cardiovascular mortality, such as myocardial infarction, stroke, and pulmonary artery embolism. Although platelet activation and activation of the plasmatic coagulation system are the hallmarks of thrombus formation, inflammatory processes and the cellular responses involved are increasingly being recognized as critical modulators of thrombosis. In the context of many chronic inflammatory diseases that are associated with a high thrombotic risk, oxidized lipoproteins represent a prominent sterile trigger of inflammation. Oxidized low-density lipoprotein and its components play a central role in the initiation and progression of atherosclerotic plaques, but also in other processes that lead to thrombotic events. Moreover, dying cells and microvesicles can be decorated with some of the same oxidized lipid components that are found on oxidized lipoproteins, and thereby similar mechanisms of thromboinflammation may also be active in venous thrombosis. In this review, we summarize the current knowledge on how oxidized lipoproteins and components thereof affect the cells and pathways involved in thrombosis.
Collapse
Affiliation(s)
- G Obermayer
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - T Afonyushkin
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - C J Binder
- Department of Laboratory Medicine, Medical University of Vienna, Vienna, Austria
- Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| |
Collapse
|
446
|
Chinchilla-López P, Cruz-Ramón V, Ramírez-Pérez O, Méndez-Sánchez N. Gastroenteritis in an adult female revealing hemolytic uremic syndrome: Case report. World J Gastroenterol 2018; 24:763-766. [PMID: 29456415 PMCID: PMC5807679 DOI: 10.3748/wjg.v24.i6.763] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 12/15/2017] [Accepted: 12/20/2017] [Indexed: 02/06/2023] Open
Abstract
Nowadays acute gastroenteritis infection caused by Escherichia coli (E. coli) O157:H7 is frequently associated with hemolytic uremic syndrome (HUS), which usually developed after prodromal diarrhea that is often bloody. The abdominal pain accompanied by failure kidney is a suspicious symptom to develop this disorder. Their pathological characteristic is vascular damage which manifested as arteriolar and capillary thrombosis with abnormalities in the endothelium and vessel walls. The major etiological agent of HUS is enterohemorragic (E coli) strain belonging to serotype O157:H7. The lack of papers about HUS associated to gastroenteritis lead us to report this case for explain the symptoms that are uncommon. Furthermore, this report provides some strategies to suspect and make an early diagnosis, besides treatment approach to improving outcomes and prognosis for patients with this disorder.
Collapse
Affiliation(s)
| | - Vania Cruz-Ramón
- Liver Research Unit, Medica Sur Clinic and Foundation, Mexico City 14050, Mexico
| | - Oscar Ramírez-Pérez
- Liver Research Unit, Medica Sur Clinic and Foundation, Mexico City 14050, Mexico
| | - Nahum Méndez-Sánchez
- Liver Research Unit, Medica Sur Clinic and Foundation, Mexico City 14050, Mexico
| |
Collapse
|
447
|
Jia H, Thelwell C, Dilger P, Bird C, Daniels S, Wadhwa M. Endothelial cell functions impaired by interferon in vitro: Insights into the molecular mechanism of thrombotic microangiopathy associated with interferon therapy. Thromb Res 2018; 163:105-116. [PMID: 29407621 DOI: 10.1016/j.thromres.2018.01.039] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2017] [Revised: 12/22/2017] [Accepted: 01/22/2018] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Interferon (IFN)-α and IFN-β approved for treatment of chronic hepatitis C viral infection and multiple sclerosis respectively have been linked to thrombotic microangiopathy (TMA) affecting renal function. Since the molecular mechanisms underlying this severe complication remain largely unclear, we aimed to investigate whether IFN affects directly in vitro endothelial cell functions associated with angiogenesis and blood haemostasis, as well as endothelial cell-derived vasodilators of nitric oxide (NO) and prostacyclin. METHODS Proliferation and survival of human umbilical vein endothelial cells (HUVECs) were measured by BrdU incorporation and alamarBlue assays. Angiogenesis was evaluated in co-cultures of HUVECs and human dermal fibroblasts. Fibrinolysis molecules were measured with ELISA. NO and prostacyclin were measured using a fluorescent NO-specific probe and a competitive enzyme immunoassay, respectively. RESULTS HUVEC proliferation was dose-dependently inhibited by IFN-β1a and IFN-β1b, but not by IFN-α2a and IFN-α2b. Consistently, IFN-β1a and IFN-β1b also reduced survival of HUVECs, but this again was not observed with IFN-α. However, both IFN subtypes inhibited VEGF-induced development of capillary-like structures, but the effect of IFN-α was less potent than IFN-β. In addition, both IFN subtypes upregulated interferon inducible protein 10 production from treated co-cultures while suppressing angiogenesis. Furthermore, intracellular NO generation was reduced by IFN-α2a and IFN-β1a, whereas prostacyclin release from HUVECs was not affected by IFN. Importantly, both IFN-β1a- and IFN-β1b-treated HUVECs showed a marked reduction in urokinase-type plasminogen activator release and a much greater secretion of plasminogen activator inhibitor-1 than tissue-type plasminogen activator compared with untreated cells, suggesting decreased fibrinolytic activity. IFN-α, however was less effective in modulating the fibrinolysis system. CONCLUSIONS We demonstrate the detrimental effects of IFN on endothelial cell functions mediated with angiogenesis and fibrinolysis, which could potentially cause the loss of physiological endothelium thromboresistance and facilitate the development of vascular complications in a clinical setting. Mechanistically, our findings have implications for understanding how IFN therapy can foster the development of TMA.
Collapse
Affiliation(s)
- Haiyan Jia
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom.
| | - Craig Thelwell
- Section of Haemostasis, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Paula Dilger
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Chris Bird
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Sarah Daniels
- Section of Haemostasis, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| | - Meenu Wadhwa
- Section of Cytokines and Growth Factors, Division of Biotherapeutics, National Institute for Biological Standards and Control, United Kingdom
| |
Collapse
|
448
|
Prevention of thrombotic disorders by antithrombotic diet and exercise: evidence by using global thrombosis tests. Future Sci OA 2018; 4:FSO285. [PMID: 29682320 PMCID: PMC5905641 DOI: 10.4155/fsoa-2017-0104] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 01/02/2018] [Indexed: 12/28/2022] Open
Abstract
Prevention of thrombotic disorders has priority over treatment. There are only two pathologically relevant tests which are suitable for measuring the overall thrombotic status both in experimental conditions and in humans. The Global Thrombosis Test (GTT) and the Global Parallel-Plate Thrombosis Test can detect the pathologically relevant global thrombotic status. These tests have been successfully used for monitoring the effect of antithrombotic drugs and for developing novel antithrombotic agents. By using GTT, varieties of fruits, vegetables, and regular physical exercise have been tested for the effect on global thrombotic status. This review discusses the published evidence for the benefit of diet of selected fruit and vegetable varieties and doing regular physical exercise on improving thrombotic status. Future clinical trials monitored by GTT or Global Parallel-Plate Thrombosis Test could decide on the effectiveness of an experimentally proven antithrombotic diet with regular physical exercise in the prevention of thrombotic diseases.
Collapse
|
449
|
Boff D, Fagundes CT, Russo RC, Amaral FA. Innate Immunity and Inflammation: The Molecular Mechanisms Governing the Cross-Talk Between Innate Immune and Endothelial Cells. IMMUNOPHARMACOLOGY AND INFLAMMATION 2018:33-56. [DOI: 10.1007/978-3-319-77658-3_2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
450
|
McCandless SE, Yanovski JA, Miller J, Fu C, Bird LM, Salehi P, Chan CL, Stafford D, Abuzzahab MJ, Viskochil D, Barlow SE, Angulo M, Myers SE, Whitman BY, Styne D, Roof E, Dykens EM, Scheimann AO, Malloy J, Zhuang D, Taylor K, Hughes TE, Kim DD, Butler MG. Effects of MetAP2 inhibition on hyperphagia and body weight in Prader-Willi syndrome: A randomized, double-blind, placebo-controlled trial. Diabetes Obes Metab 2017; 19:1751-1761. [PMID: 28556449 PMCID: PMC5673540 DOI: 10.1111/dom.13021] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 05/23/2017] [Accepted: 05/24/2017] [Indexed: 01/01/2023]
Abstract
AIMS There are no treatments for the extreme hyperphagia and obesity in Prader-Willi syndrome (PWS). The bestPWS clinical trial assessed the efficacy, safety and tolerability of the methionine aminopeptidase 2 (MetAP2) inhibitor, beloranib. MATERIALS AND METHODS Participants with PWS (12-65 years old) were randomly assigned (1:1:1) to biweekly placebo, 1.8 mg beloranib or 2.4 mg beloranib injection for 26 weeks at 15 US sites. Co-primary endpoints were the changes in hyperphagia [measured by Hyperphagia Questionnaire for Clinical Trials (HQ-CT); possible score 0-36] and weight by intention-to-treat. ClinicalTrials.gov registration: NCT02179151. RESULTS One-hundred and seven participants were included in the intention-to-treat analysis: placebo (n = 34); 1.8 mg beloranib (n = 36); or 2.4 mg beloranib (n = 37). Improvement (reduction) in HQ-CT total score was greater in the 1.8 mg (mean difference -6.3, 95% CI -9.6 to -3.0; P = .0003) and 2.4 mg beloranib groups (-7.0, 95% CI -10.5 to -3.6; P = .0001) vs placebo. Compared with placebo, weight change was greater with 1.8 mg (mean difference - 8.2%, 95% CI -10.8 to -5.6; P < .0001) and 2.4 mg beloranib (-9.5%, 95% CI -12.1 to -6.8; P < .0001). Injection site bruising was the most frequent adverse event with beloranib. Dosing was stopped early due to an imbalance in venous thrombotic events in beloranib-treated participants (2 fatal events of pulmonary embolism and 2 events of deep vein thrombosis) compared with placebo. CONCLUSIONS MetAP2 inhibition with beloranib produced statistically significant and clinically meaningful improvements in hyperphagia-related behaviours and weight loss in participants with PWS. Although investigation of beloranib has ceased, inhibition of MetAP2 is a novel mechanism for treating hyperphagia and obesity.
Collapse
Affiliation(s)
- Shawn E McCandless
- UH Cleveland Medical Center and Case Western Reserve University, Cleveland, Ohio
| | - Jack A Yanovski
- Section on Growth and Obesity, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland
| | | | - Cary Fu
- Vanderbilt University Medical Center, Nashville, Tennessee
| | - Lynne M Bird
- UCSD Rady Children's Hospital, San Diego, California
| | - Parisa Salehi
- Division of Endocrinology & Diabetes, Seattle Children's Hospital, Seattle, Washington
| | | | | | | | | | | | | | - Susan E Myers
- Saint Louis University School of Medicine, Saint Louis, Missouri
| | | | - Dennis Styne
- UC Davis Children's Hospital, UC Davis Medical Center, Davis, California
| | - Elizabeth Roof
- Vanderbilt University Medical Center, Nashville, Tennessee
| | | | - Ann O Scheimann
- Baylor College of Medicine and Johns Hopkins School of Medicine, Baltimore, Maryland
| | | | | | | | | | | | - Merlin G Butler
- Departments of Psychiatry, Behavioral Sciences and Pediatrics, University of Kansas Medical Center, Kansas City, Kansas
| |
Collapse
|