1
|
Bao Y, Zhang H, Wang D, Yan P, Shao S, Zhang Z, Liu B, Li N. The Pathological Factors Involved in Current In Vitro Atherosclerotic Models. TISSUE ENGINEERING. PART B, REVIEWS 2024; 30:530-544. [PMID: 38258801 DOI: 10.1089/ten.teb.2023.0272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
Cardiovascular disease stemmed from atherosclerosis (AS) is well recognized to be the predominant cause of global death. To comprehensively clarify the pathogenesis of AS, exploit effective drugs, as well as develop therapeutic solutions, various atherosclerotic models were constructed in vitro and widely utilized by the scientific community. Compared with animal models, the in vitro atherosclerotic models play a prominent role not only in the targeted research of single pathological factor related to AS in the human derived system, but also in the combined study on multipathological factors leading to AS, thereby contributing tremendously to the in-depth elucidation of atherosclerotic pathological process. In the current review, a variety of pathological factors incorporated into the existing atherosclerotic models in vitro are broadly elaborated, including the pathological mechanism, in vitro simulation approaches, and the desired improvement perspectives for reproducing each pathological factor. In addition, this review also summarizes the advantages and disadvantages of current atherosclerotic models as well as their potential functionality. Finally, the promising aspects for future atherosclerotic models in vitro with potential advances are also discussed.
Collapse
Affiliation(s)
- Yuxin Bao
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Hangyu Zhang
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Danbo Wang
- Cancer Hospital of Dalian University of Technology, Shenyang, China
| | - Peishi Yan
- Department of Cardiology, Central Hospital of Dalian University of Technology, Dalian, China
| | - Shuai Shao
- School of Biomedical Engineering, Faculty of Medicine, Dalian University of Technology, Dalian, China
| | - Zhengyao Zhang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Bo Liu
- Cancer Hospital of Dalian University of Technology, Shenyang, China
- School of Basic Medical Sciences, Faculty of Medicine, Dalian University of Technology, Dalian, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, China
| | - Na Li
- Cancer Hospital of Dalian University of Technology, Shenyang, China
- School of Basic Medical Sciences, Faculty of Medicine, Dalian University of Technology, Dalian, China
- Liaoning Key Lab of Integrated Circuit and Biomedical Electronic System, Dalian University of Technology, Dalian, China
| |
Collapse
|
2
|
Nishimura A, Nelke C, Huber M, Mensch A, Roth A, Oberwittler C, Zimmerlein B, Krämer HH, Neuen-Jacob E, Stenzel W, Müller-Ladner U, Ruck T, Schänzer A. Differentiating idiopathic inflammatory myopathies by automated morphometric analysis of MHC-1, MHC-2 and ICAM-1 in muscle tissue. Neuropathol Appl Neurobiol 2024; 50:e12998. [PMID: 39030945 DOI: 10.1111/nan.12998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/22/2024]
Abstract
AIMS Diagnosis of idiopathic inflammatory myopathies (IIM) is based on morphological characteristics and the evaluation of disease-related proteins. However, although broadly applied, substantial bias is imposed by the respective methods, observers and individual staining approaches. We aimed to quantify the protein levels of major histocompatibility complex (MHC)-1, (MHC)-2 and intercellular adhesion molecule (ICAM)-1 using an automated morphometric method to mitigate bias. METHODS Double immunofluorescence staining was performed on whole muscle sections to study differences in protein expression in myofibre and endomysial vessels. We analysed all IIM subtypes including dermatomyositis (DM), anti-synthetase syndrome (ASyS), inclusion body myositis (IBM), immune-mediated-necrotising myopathy (IMNM), dysferlinopathy (DYSF), SARS-CoV-2 infection and vaccination-associated myopathy. Biopsies with neurogenic atrophy (NA) and normal morphology served as controls. Bulk RNA-Sequencing (RNA-Seq) was performed on a subset of samples. RESULTS Our study highlights the significance of MHC-1, MHC-2 and ICAM-1 in diagnosing IIM subtypes and reveals distinct immunological profiles. RNASeq confirmed the precision of our method and identified specific gene pathways in the disease subtypes. Notably, ASyS, DM and SARS-CoV-2-associated myopathy showed increased ICAM-1 expression in the endomysial capillaries, indicating ICAM-1-associated vascular activation in these conditions. In addition, ICAM-1 showed high discrimination between different subgroups with high sensitivity and specificity. CONCLUSIONS Automated morphometric analysis provides precise quantitative data on immune-associated proteins that can be integrated into our pathophysiological understanding of IIM. Further, ICAM-1 holds diagnostic value for the detection of IIM pathology.
Collapse
Affiliation(s)
- Anna Nishimura
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
| | - Christopher Nelke
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Melanie Huber
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig-University, Giessen, Germany
| | - Alexander Mensch
- Department of Neurology, University Medicine Halle, Halle (Saale), Germany
| | - Angela Roth
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
| | | | | | - Heidrun H Krämer
- Department of Neurology, Justus-Liebig-University, Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany
| | - Eva Neuen-Jacob
- Institute of Neuropathology, Heinrich-Heine-University, Düsseldorf, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ulf Müller-Ladner
- Department of Rheumatology and Clinical Immunology, Campus Kerckhoff, Justus-Liebig-University, Giessen, Germany
| | - Tobias Ruck
- Department of Neurology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anne Schänzer
- Institute of Neuropathology, Justus-Liebig University Giessen, Germany
- Translational Neuroscience Network Giessen (TNNG), Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
3
|
Baber U. Stent Failure After Percutaneous Coronary Intervention in Diabetes Mellitus: Does Glycemic Control Matter? J Am Coll Cardiol 2024; 84:273-275. [PMID: 38986669 DOI: 10.1016/j.jacc.2024.04.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 07/12/2024]
Affiliation(s)
- Usman Baber
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA.
| |
Collapse
|
4
|
Ekelund C, Dereke J, Nilsson C, Landin-Olsson M. Are soluble E-selectin, ICAM-1, and VCAM-1 potential predictors for the development of diabetic retinopathy in young adults, 15-34 years of age? A Swedish prospective cohort study. PLoS One 2024; 19:e0304173. [PMID: 38843125 PMCID: PMC11156360 DOI: 10.1371/journal.pone.0304173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Accepted: 05/08/2024] [Indexed: 06/09/2024] Open
Abstract
The aim of this study was to determine plasma levels of three adhesion molecules that may contribute to the development of diabetic retinopathy; soluble endothelial selectin (sE-selectin), soluble intercellular adhesion molecule-1 (sICAM-1), and soluble vascular cell adhesion molecule-1 (sVCAM-1), in young adults, aged 15-34 years at diagnosis of diabetes, to find potential predictors for development of retinopathy, and to evaluate their relation to diabetes associated autoantibodies. Participants with type 1 (n = 169) and type 2 diabetes (n = 83) were selected from the complications trial of the Diabetes Incidence Study in Sweden and classified in two subgroups according to presence (n = 80) or absence (n = 172) of retinopathy as determined by retinal photography at follow-up 8-10 years after diagnosis of diabetes. Blood samples were collected at diagnosis in 1987-88. The levels of sE-selectin, sICAM-1, and sVCAM-1 were analysed by enzyme-linked immunosorbent assay and islet cell antibodies by a prolonged two-colour immunofluorescent assay. Mean HbA1c (p<0.001) and clinical characteristics: mean body mass index (p = 0.019), systolic blood pressure (p = 0.002), diastolic blood pressure (p = 0.003), male gender (p = 0.026), and young age at diagnosis of diabetes (p = 0.015) remained associated with development of retinopathy in type 1 diabetes. However, in a multivariate analysis only HbA1c remained as a risk factor. sE-selectin was significantly higher in the group with type 2 diabetes and retinopathy, compared to the group with type 2 diabetes without retinopathy (p = 0.04). Regarding sE-selectin, sICAM-1, and sVCAM-1 in participants with type 1 diabetes, no differences were observed between the groups with or without retinopathy. This trial confirmed the role of HbA1c and clinical characteristics as predictors for development of retinopathy in type 1 diabetes. sE-selectin stands out as a potential predictor for development of retinopathy in type 2 diabetes, whereas a predictive role for sICAM-1 and sVCAM-1 could not be identified neither for type 1 nor type 2 diabetes.
Collapse
Affiliation(s)
- Charlotte Ekelund
- Diabetes Research Laboratory, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Jonatan Dereke
- Diabetes Research Laboratory, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
| | - Charlotta Nilsson
- Diabetes Research Laboratory, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Pediatrics, Helsingborg Hospital, Helsingborg, Sweden
| | - Mona Landin-Olsson
- Diabetes Research Laboratory, Department of Clinical Sciences, Faculty of Medicine, Lund University, Lund, Sweden
- Department of Endocrinology, Skåne University Hospital, Lund, Sweden
| |
Collapse
|
5
|
Zhu Y, Wang T, Yang Y, Wang Z, Chen X, Wang L, Niu R, Sun Z, Zhang C, Luo Y, Hu Y, Gu W. Low shear stress exacerbates atherosclerosis by inducing the generation of neutrophil extracellular traps via Piezo1-mediated mechanosensation. Atherosclerosis 2024; 391:117473. [PMID: 38412763 DOI: 10.1016/j.atherosclerosis.2024.117473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 01/26/2024] [Accepted: 01/28/2024] [Indexed: 02/29/2024]
Abstract
BACKGROUND AND AIMS Atherosclerosis is a chronic lipid-driven inflammatory disease largely influenced by hemodynamics. Neutrophil extracellular trap (NET)-mediated inflammation plays an important role in atherosclerosis. However, little is known about the relationship between low shear stress (LSS) and NET generation, as well as the underlying mechanism. METHODS We induced LSS by partial ligation of the left carotid artery in high-fat diet-fed male ApoE-/- mice. To further validate the direct relationship between LSS and NET formation invitro, differentiated human promyelocytic leukemia HL-60 cells and bone marrow-derived neutrophils were suspended in fluid flow under normal or low shear stress using a parallel-plate flow chamber system. RESULTS Four weeks after surgery, ligated carotid arteries had more lipid deposition, larger plaque area, and increased NET formation than unligated arteries. Inhibition of NETosis could significantly reduce plaque formation in ApoE-/- mice. Invitro, LSS could promote NET generation directly through downregulation of Piezo1, a mechanosensitive ion channel. Downregulation of Piezol could activate neutrophils and promote NETosis in static conditions. Conversely, Yoda1-evoked activation of Piezo1 attenuated LSS-induced NETosis. Mechanistically, downregulation of Piezo1 resulted in decreased Ca2+ influx and increased histone deacetylase 2 (HDAC2), which increased reactive oxygen species levels and led to NETosis. LSS-induced NET generation also promoted apoptosis and adherence of endothelial cells. CONCLUSION LSS directly promotes NETosis through the Piezo1-HDAC2 axis in atherosclerosis progression. This study uncovers the essential role of Piezo1-mediated mechanical signaling in NET generation and plaque formation, which provides a promising therapeutic strategy for atherosclerosis.
Collapse
Affiliation(s)
- Ying Zhu
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Tian Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400030, China; College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yan Yang
- Department of Cardiovascular Surgery, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Zining Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Xiaohui Chen
- College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Liu Wang
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Ruyan Niu
- College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Zixin Sun
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400030, China
| | - Chong Zhang
- College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yang Luo
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Yijie Hu
- Department of Cardiovascular Surgery, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| | - Wei Gu
- Center of Smart Laboratory and Molecular Medicine, School of Medicine, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
6
|
Calvier L, Alexander A, Marckx AT, Kounnas MZ, Durakoglugil M, Herz J. Safety of Anti-Reelin Therapeutic Approaches for Chronic Inflammatory Diseases. Cells 2024; 13:583. [PMID: 38607022 PMCID: PMC11011630 DOI: 10.3390/cells13070583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 03/18/2024] [Accepted: 03/25/2024] [Indexed: 04/13/2024] Open
Abstract
Reelin, a large extracellular glycoprotein, plays critical roles in neuronal development and synaptic plasticity in the central nervous system (CNS). Recent studies have revealed non-neuronal functions of plasma Reelin in inflammation by promoting endothelial-leukocyte adhesion through its canonical pathway in endothelial cells (via ApoER2 acting on NF-κB), as well as in vascular tone regulation and thrombosis. In this study, we have investigated the safety and efficacy of selectively depleting plasma Reelin as a potential therapeutic strategy for chronic inflammatory diseases. We found that Reelin expression remains stable throughout adulthood and that peripheral anti-Reelin antibody treatment with CR-50 efficiently depletes plasma Reelin without affecting its levels or functionality within the CNS. Notably, this approach preserves essential neuronal functions and synaptic plasticity. Furthermore, in mice induced with experimental autoimmune encephalomyelitis (EAE), selective modulation of endothelial responses by anti-Reelin antibodies reduces pathological leukocyte infiltration without completely abolishing diapedesis. Finally, long-term Reelin depletion under metabolic stress induced by a Western diet did not negatively impact the heart, kidney, or liver, suggesting a favorable safety profile. These findings underscore the promising role of peripheral anti-Reelin therapeutic strategies for autoimmune diseases and conditions where endothelial function is compromised, offering a novel approach that may avoid the immunosuppressive side effects associated with conventional anti-inflammatory therapies.
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Anna Alexander
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Austin T. Marckx
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | | | - Murat Durakoglugil
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas (UT) Southwestern Medical Center, Dallas, TX 75390, USA (A.T.M.); (M.D.); (J.H.)
- Center for Translational Neurodegeneration Research, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neuroscience, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology and Neurotherapeutics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
7
|
Zhang J, Zhang S, Xu S, Zhu Z, Li J, Wang Z, Wada Y, Gatt A, Liu J. Oxidative Stress Induces E-Selectin Expression through Repression of Endothelial Transcription Factor ERG. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:1835-1843. [PMID: 37930129 PMCID: PMC10694031 DOI: 10.4049/jimmunol.2300043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Accepted: 10/12/2023] [Indexed: 11/07/2023]
Abstract
Oxidative stress induces a prothrombotic state through enhancement of adhesion properties of the endothelium. E-selectin, an endothelial cell adhesion molecule, becomes a therapeutic target for venous thrombosis, whereas the regulatory mechanisms of its expression have not been fully understood. In the present study, we report that H2O2 treatment increases expression of E-selectin but decreases expression of the endothelial transcription factor ETS-related gene (ERG) in HUVECs in a dose- and time-dependent manner. In BALB/c mice treated with hypochlorous acid, E-selectin expression is increased and ERG expression is decreased in endothelial cells of the brain and lung. RNA interference of ERG upregulates E-selectin expression, whereas transfection of ERG-expressing plasmid downregulates E-selectin expression in HUVECs. Knockdown or overexpression of ERG comprises H2O2-induced E-selectin expression in HUVECs. Deletion of the Erg gene in mice results in embryonic lethality at embryonic days 10.5-12.5, and E-selectin expression is increased in the Erg-/- embryos. No chromatin loop was found on the E-selectin gene or its promoter region by capture high-throughput chromosome conformation capture. Chromatin immunoprecipitation and luciferase reporter assay determined that the -127 ERG binding motif mediates ERG-repressed E-selectin promoter activity. In addition, ERG decreases H2O2-induced monocyte adhesion. Together, ERG represses the E-selectin gene transcription and inhibits oxidative stress-induced endothelial cell adhesion.
Collapse
Affiliation(s)
- Jinjin Zhang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Shuo Zhang
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Shanhu Xu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zhiying Zhu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Jiang Li
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
| | - Zengjin Wang
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Youichiro Wada
- Isotope Science Center, The University of Tokyo, Tokyo, Japan
| | - Alex Gatt
- Department of Pathology, Faculty of Medicine and Surgery, University of Malta, Tal-Qroqq, Msida, Malta
- Hematology Laboratory, Department of Pathology, Mater Dei Hospital, Msida, Malta
| | - Ju Liu
- Medical Research Center, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, China
- Institute of Microvascular Medicine, Medical Research Center, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
8
|
Angelis D, Jaleel MA, Brion LP. Hyperglycemia and prematurity: a narrative review. Pediatr Res 2023; 94:892-903. [PMID: 37120652 DOI: 10.1038/s41390-023-02628-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 04/11/2023] [Accepted: 04/15/2023] [Indexed: 05/01/2023]
Abstract
Hyperglycemia is commonly encountered in extremely preterm newborns and physiologically can be attributed to immaturity in several biochemical pathways related to glucose metabolism. Although hyperglycemia is associated with a variety of adverse outcomes frequently described in this population, evidence for causality is lacking. Variations in definitions and treatment approaches have further complicated the understanding and implications of hyperglycemia on the immediate and long-term effects in preterm newborns. In this review, we describe the relationship between hyperglycemia and organ development, outcomes, treatment options, and potential gaps in knowledge that need further research. IMPACT: Hyperglycemia is common and less well described than hypoglycemia in extremely preterm newborns. Hyperglycemia can be attributed to immaturity in several cellular pathways involved in glucose metabolism in this age group. Hyperglycemia has been shown to be associated with a variety of adverse outcomes frequently described in this population; however, evidence for causality is lacking. Variations in definitions and treatment approaches have complicated the understanding and the implications of hyperglycemia on the immediate and long-term effects outcomes. This review describes the relationship between hyperglycemia and organ development, outcomes, treatment options, and potential gaps in knowledge that need further research.
Collapse
Affiliation(s)
- Dimitrios Angelis
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Mambarambath A Jaleel
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Luc P Brion
- Division of Neonatal-Perinatal Medicine, Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Yanai H, Adachi H, Hakoshima M, Katsuyama H. Significance of Endothelial Dysfunction Amelioration for Sodium-Glucose Cotransporter 2 Inhibitor-Induced Improvements in Heart Failure and Chronic Kidney Disease in Diabetic Patients. Metabolites 2023; 13:736. [PMID: 37367894 DOI: 10.3390/metabo13060736] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/29/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Beyond lowering plasma glucose levels, sodium-glucose cotransporter 2 inhibitors (SGLT2is) significantly reduce hospitalization for heart failure (HF) and retard the progression of chronic kidney disease (CKD) in patients with type 2 diabetes. Endothelial dysfunction is not only involved in the development and progression of cardiovascular disease (CVD), but is also associated with the progression of CKD. In patients with type 2 diabetes, hyperglycemia, insulin resistance, hyperinsulinemia and dyslipidemia induce the development of endothelial dysfunction. SGLT2is have been shown to improve endothelial dysfunction, as assessed by flow-mediated vasodilation, in individuals at high risk of CVD. Along with an improvement in endothelial dysfunction, SGLT2is have been shown to improve oxidative stress, inflammation, mitochondrial dysfunction, glucotoxicity, such as the advanced signaling of glycation end products, and nitric oxide bioavailability. The improvements in endothelial dysfunction and such endothelium-derived factors may play an important role in preventing the development of coronary artery disease, coronary microvascular dysfunction and diabetic cardiomyopathy, which cause HF, and play a role in retarding CKD. The suppression of the development of HF and the progression of CKD achieved by SGLT2is might have been largely induced by their capacity to improve vascular endothelial function.
Collapse
Affiliation(s)
- Hidekatsu Yanai
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Hiroki Adachi
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Mariko Hakoshima
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| | - Hisayuki Katsuyama
- Department of Diabetes, Endocrinology and Metabolism, National Center for Global Health and Medicine Kohnodai Hospital, 1-7-1 Kohnodai, Chiba 272-8516, Japan
| |
Collapse
|
10
|
Haydinger CD, Ashander LM, Tan ACR, Smith JR. Intercellular Adhesion Molecule 1: More than a Leukocyte Adhesion Molecule. BIOLOGY 2023; 12:biology12050743. [PMID: 37237555 DOI: 10.3390/biology12050743] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/15/2023] [Accepted: 05/16/2023] [Indexed: 05/28/2023]
Abstract
Intercellular adhesion molecule 1 (ICAM-1) is a transmembrane protein in the immunoglobulin superfamily expressed on the surface of multiple cell populations and upregulated by inflammatory stimuli. It mediates cellular adhesive interactions by binding to the β2 integrins macrophage antigen 1 and leukocyte function-associated antigen 1, as well as other ligands. It has important roles in the immune system, including in leukocyte adhesion to the endothelium and transendothelial migration, and at the immunological synapse formed between lymphocytes and antigen-presenting cells. ICAM-1 has also been implicated in the pathophysiology of diverse diseases from cardiovascular diseases to autoimmune disorders, certain infections, and cancer. In this review, we summarize the current understanding of the structure and regulation of the ICAM1 gene and the ICAM-1 protein. We discuss the roles of ICAM-1 in the normal immune system and a selection of diseases to highlight the breadth and often double-edged nature of its functions. Finally, we discuss current therapeutics and opportunities for advancements.
Collapse
Affiliation(s)
- Cameron D Haydinger
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Liam M Ashander
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Alwin Chun Rong Tan
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| | - Justine R Smith
- College of Medicine and Public Health, Flinders University, Adelaide, SA 5042, Australia
| |
Collapse
|
11
|
Shaw DM, Keaney L, Maunder E, Dulson DK. Natural killer cell subset count and antigen-stimulated activation in response to exhaustive running following adaptation to a ketogenic diet. Exp Physiol 2023; 108:706-714. [PMID: 36843281 PMCID: PMC10988467 DOI: 10.1113/ep090729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 02/06/2023] [Indexed: 02/28/2023]
Abstract
NEW FINDINGS What is the central question of this study? Does a ketogenic diet (KD) modulate circulating counts of natural killer (NK) cells, including CD56bright and CD56dim subsets, and their ability to activate (CD69 expression) following in vitro antigen stimulation in response to exhaustive moderate-intensity exercise? What is the main finding and its importance? The KD amplified the biphasic exercise-induced NK cell response due to a greater mobilisation of the cytotoxic CD56dim subset but did not alter NK cell CD69 expression. The KD appears to modulate exercise-induced circulating NK cell mobilisation and egress, but not antigen-stimulated circulating NK cell activation. ABSTRACT We investigated the effect of a 31-day ketogenic diet (KD) compared with a habitual, carbohydrate (CHO)-based diet on total circulating natural killer (NK) CD3- CD56+ , dim and bright subset count, and antigen-stimulated CD3- CD56+ cell activation (CD69+ ) in response to exhaustive running. In a randomised, repeated-measures, cross-over study, eight trained, male endurance athletes ingested a 31-day low-CHO KD or their habitual diet (HD). On day 31, participants ran to exhaustion at 70%V ̇ O 2 max $\dot{V}_{{\rm{O}}_{2}{\rm{max}}}$ (∼3.5-4 h, ∼45-50 km). A low-CHO (<10 g) meal was ingested prior to the KD trial, with fat ingested during exercise. A high-CHO (2 g kg-1 ) meal was ingested prior to the HD trial, with CHO (∼55 g h-1 ) ingested during exercise. Venous blood samples were collected at pre-exercise, post-exercise and 1 h post-exercise. The KD amplified the classical exercise-induced biphasic CD3- CD56+ cell response by increasing the post-exercise counts (P = 0.0004), which appeared to be underpinned by the cytotoxic CD3- CD56dim subset (main effect of time point, P < 0.0001). The KD had no effect on NK cells' expression of CD69 or their geometric mean fluorescence intensity of CD69 expression, either for unstimulated or for antigen-stimulated NK cells (all P > 0.05). In conclusion, adaptation to a KD may alter the number of circulating NK cells but not their ability to activate to an antigenic challenge.
Collapse
Affiliation(s)
- David M. Shaw
- School of Sport, Exercise and NutritionMassey UniversityAucklandNew Zealand
| | - Lauren Keaney
- Sports Performance Research Institute New Zealand (SPRINZ)Auckland University of TechnologyAucklandNew Zealand
| | - Ed Maunder
- Sports Performance Research Institute New Zealand (SPRINZ)Auckland University of TechnologyAucklandNew Zealand
| | - Deborah K. Dulson
- School of Biomedical, Nutritional and Sport Sciences, Faculty of Medical SciencesNewcastle UniversityNewcastle Upon TyneUK
| |
Collapse
|
12
|
Yang Y, Jiang G, Huang R, Liu Y, Chang X, Fu S. Targeting the NLRP3 inflammasome in diabetic retinopathy: From Pathogenesis to Therapeutic Strategies. Biochem Pharmacol 2023; 212:115569. [PMID: 37100255 DOI: 10.1016/j.bcp.2023.115569] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 04/16/2023] [Accepted: 04/18/2023] [Indexed: 04/28/2023]
Abstract
Diabetic retinopathy (DR) is a common diabetic microvascular complication and the main cause of vision loss in working-aged people. The NLRP3 inflammasome is a cytosolic multimeric complex that plays a significant role in innate immunity. After sensing injury, the NLRP3 inflammasome induces inflammatory mediator secretion and triggers a form of inflammatory cell death known as pyroptosis. Studies over the past five years have shown increased expression of NLRP3 and related inflammatory mediators in vitreous samples from DR patients at different clinical stages. Many NLRP3-targeted inhibitors have shown great antiangiogenic and anti-inflammatory effects in diabetes mellitus models, suggesting that the NLRP3 inflammasome is involved in the progression of DR. This review covers the molecular mechanisms of NLRP3 inflammasome activation. Furthermore, we discuss the implications of the NLRP3 inflammasome in DR, including the induction of pyroptosis and inflammation and the promotion of microangiopathy and retinal neurodegeneration. We also summarize the research progress on targeting the NLRP3 inflammasome in DR therapeutics with the expectation of providing new insights into DR progression and treatment.
Collapse
Affiliation(s)
- Yuxuan Yang
- The First Clinical Medical College, Lanzhou University, Lanzhou, The People's Republic of China, 730000
| | - Gengchen Jiang
- The First Clinical Medical College, Lanzhou University, Lanzhou, The People's Republic of China, 730000
| | - Runchun Huang
- The First Clinical Medical College, Lanzhou University, Lanzhou, The People's Republic of China, 730000
| | - Yi Liu
- The First Clinical Medical College, Lanzhou University, Lanzhou, The People's Republic of China, 730000
| | - Xingyu Chang
- The First Clinical Medical College, Lanzhou University, Lanzhou, The People's Republic of China, 730000
| | - Songbo Fu
- Department of Endocrinology, First Hospital of Lanzhou University, Lanzhou, Gansu, The People's Republic of China, 730000; Gansu Province Clinical Research Center for Endocrine Disease, Gansu, The People's Republic of China, 730000.
| |
Collapse
|
13
|
Yao M, Hao Y, Wang T, Xie M, Li H, Feng J, Feng L, Ma D. A review of stress-induced hyperglycaemia in the context of acute ischaemic stroke: Definition, underlying mechanisms, and the status of insulin therapy. Front Neurol 2023; 14:1149671. [PMID: 37025208 PMCID: PMC10070880 DOI: 10.3389/fneur.2023.1149671] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 02/21/2023] [Indexed: 04/08/2023] Open
Abstract
The transient elevation of blood glucose produced following acute ischaemic stroke (AIS) has been described as stress-induced hyperglycaemia (SIH). SIH is common even in patients with AIS who have no previous diagnosis of diabetes mellitus. Elevated blood glucose levels during admission and hospitalization are strongly associated with enlarged infarct size and adverse prognosis in AIS patients. However, insulin-intensive glucose control therapy defined by admission blood glucose for SIH has not achieved the desired results, and new treatment ideas are urgently required. First, we explore the various definitions of SIH in the context of AIS and their predictive value in adverse outcomes. Then, we briefly discuss the mechanisms by which SIH arises, describing the dual effects of elevated glucose levels on the central nervous system. Finally, although preclinical studies support lowering blood glucose levels using insulin, the clinical outcomes of intensive glucose control are not promising. We discuss the reasons for this phenomenon.
Collapse
Affiliation(s)
- Mengyue Yao
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tian Wang
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Meizhen Xie
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Hui Li
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Liangshu Feng
- Stroke Centre, Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin, China
- Liangshu Feng
| | - Di Ma
- Department of Neurology and Neuroscience Centre, The First Hospital of Jilin University, Changchun, Jilin, China
- *Correspondence: Di Ma
| |
Collapse
|
14
|
Norris T, Razieh C, Yates T, Zaccardi F, Gillies CL, Chudasama YV, Rowlands A, Davies MJ, McCann GP, Banerjee A, Docherty AB, Openshaw PJ, Baillie JK, Semple MG, Lawson CA, Khunti K. Admission Blood Glucose Level and Its Association With Cardiovascular and Renal Complications in Patients Hospitalized With COVID-19. Diabetes Care 2022; 45:1132-1140. [PMID: 35275994 PMCID: PMC9174963 DOI: 10.2337/dc21-1709] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 01/30/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVE To investigate the association between admission blood glucose levels and risk of in-hospital cardiovascular and renal complications. RESEARCH DESIGN AND METHODS In this multicenter prospective study of 36,269 adults hospitalized with COVID-19 between 6 February 2020 and 16 March 2021 (N = 143,266), logistic regression models were used to explore associations between admission glucose level (mmol/L and mg/dL) and odds of in-hospital complications, including heart failure, arrhythmia, cardiac ischemia, cardiac arrest, coagulation complications, stroke, and renal injury. Nonlinearity was investigated using restricted cubic splines. Interaction models explored whether associations between glucose levels and complications were modified by clinically relevant factors. RESULTS Cardiovascular and renal complications occurred in 10,421 (28.7%) patients; median admission glucose level was 6.7 mmol/L (interquartile range 5.8-8.7) (120.6 mg/dL [104.4-156.6]). While accounting for confounders, for all complications except cardiac ischemia and stroke, there was a nonlinear association between glucose and cardiovascular and renal complications. For example, odds of heart failure, arrhythmia, coagulation complications, and renal injury decreased to a nadir at 6.4 mmol/L (115 mg/dL), 4.9 mmol/L (88.2 mg/dL), 4.7 mmol/L (84.6 mg/dL), and 5.8 mmol/L (104.4 mg/dL), respectively, and increased thereafter until 26.0 mmol/L (468 mg/dL), 50.0 mmol/L (900 mg/dL), 8.5 mmol/L (153 mg/dL), and 32.4 mmol/L (583.2 mg/dL). Compared with 5 mmol/L (90 mg/dL), odds ratios at these glucose levels were 1.28 (95% CI 0.96, 1.69) for heart failure, 2.23 (1.03, 4.81) for arrhythmia, 1.59 (1.36, 1.86) for coagulation complications, and 2.42 (2.01, 2.92) for renal injury. For most complications, a modifying effect of age was observed, with higher odds of complications at higher glucose levels for patients age <69 years. Preexisting diabetes status had a similar modifying effect on odds of complications, but evidence was strongest for renal injury, cardiac ischemia, and any cardiovascular/renal complication. CONCLUSIONS Increased odds of cardiovascular or renal complications were observed for admission glucose levels indicative of both hypo- and hyperglycemia. Admission glucose could be used as a marker for risk stratification of high-risk patients. Further research should evaluate interventions to optimize admission glucose on improving COVID-19 outcomes.
Collapse
Affiliation(s)
- Tom Norris
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, U.K
| | - Cameron Razieh
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, U.K
| | - Thomas Yates
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, U.K
| | - Francesco Zaccardi
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, U.K
| | - Clare L. Gillies
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, U.K
| | - Yogini V. Chudasama
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, U.K
| | - Alex Rowlands
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, U.K
| | - Melanie J. Davies
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, U.K
| | - Gerry P. McCann
- National Institute for Health Research Leicester Biomedical Research Centre, Leicester General Hospital, Leicester, U.K
- Cardiovascular Sciences Department, University of Leicester, Leicester, U.K
| | - Amitava Banerjee
- Institute of Health Informatics, University College London, London, U.K
| | - Annemarie B. Docherty
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, U.K
- Intensive Care Unit, Royal Infirmary Edinburgh, Edinburgh, U.K
| | | | | | - Malcolm G. Semple
- National Institute for Health Research Health Protection Research Unit in Emerging and Zoonotic Infections, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool, U.K
- Respiratory Medicine, Alder Hey Children’s Hospital, Liverpool, U.K
| | - Claire A. Lawson
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, U.K
| | - Kamlesh Khunti
- Diabetes Research Centre, University of Leicester, Leicester General Hospital, Leicester, U.K
- Leicester Real World Evidence Unit, Diabetes Research Centre, University of Leicester, Leicester, U.K
- National Institute for Health Research Applied Research Collaboration–East Midlands, Leicester General Hospital, Leicester, U.K
| |
Collapse
|
15
|
Perico L, Morigi M, Galbusera M, Pezzotta A, Gastoldi S, Imberti B, Perna A, Ruggenenti P, Donadelli R, Benigni A, Remuzzi G. SARS-CoV-2 Spike Protein 1 Activates Microvascular Endothelial Cells and Complement System Leading to Platelet Aggregation. Front Immunol 2022; 13:827146. [PMID: 35320941 PMCID: PMC8936079 DOI: 10.3389/fimmu.2022.827146] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
Abstract
Microvascular thrombosis is associated with multiorgan failure and mortality in coronavirus disease 2019 (COVID-19). Although thrombotic complications may be ascribed to the ability of SARS-CoV-2 to infect and replicate in endothelial cells, it has been poorly investigated whether, in the complexity of viral infection in the human host, specific viral elements alone can induce endothelial damage. Detection of circulating spike protein in the sera of severe COVID-19 patients was evaluated by ELISA. In vitro experiments were performed on human microvascular endothelial cells from the derma and lung exposed to SARS-CoV-2-derived spike protein 1 (S1). The expression of adhesive molecules was studied by immunofluorescence and leukocyte adhesion and platelet aggregation were assessed under flow conditions. Angiotensin converting enzyme 2 (ACE2) and AMPK expression were investigated by Western Blot analysis. In addition, S1-treated endothelial cells were incubated with anti-ACE2 blocking antibody, AMPK agonist, or complement inhibitors. Our results show that significant levels of spike protein were found in the 30.4% of severe COVID-19 patients. In vitro, the activation of endothelial cells with S1 protein, via ACE2, impaired AMPK signalling, leading to robust leukocyte recruitment due to increased adhesive molecule expression and thrombomodulin loss. This S1-induced pro-inflammatory phenotype led to exuberant C3 and C5b-9 deposition on endothelial cells, along with C3a and C5a generation that further amplified S1-induced complement activation. Functional blockade of ACE2 or complement inhibition halted S1-induced platelet aggregates by limiting von Willebrand factor and P-selectin exocytosis and expression on endothelial cells. Overall, we demonstrate that SARS-CoV-2-derived S1 is sufficient in itself to propagate inflammatory and thrombogenic processes in the microvasculature, amplified by the complement system, recapitulating the thromboembolic complications of COVID-19.
Collapse
Affiliation(s)
- Luca Perico
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Marina Morigi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Miriam Galbusera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Anna Pezzotta
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Sara Gastoldi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Barbara Imberti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Annalisa Perna
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Piero Ruggenenti
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
- Unit of Nephrology and Dialysis, Azienda Socio Sanitaria Territoriale (ASST) Papa Giovanni XXIII, Bergamo, Italy
| | - Roberta Donadelli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Ariela Benigni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| | - Giuseppe Remuzzi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy
| |
Collapse
|
16
|
ETS proto-oncogene 1 modulates PTP1B expression to participate in high glucose-mediated endothelial inflammation. Acta Biochim Biophys Sin (Shanghai) 2022; 54:565-573. [PMID: 35607953 PMCID: PMC9827757 DOI: 10.3724/abbs.2022021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Hyperglycemia-induced endothelial inflammation participates in the pathogenesis of cardiovascular complications in diabetics. Previous studies showed that protein tyrosine phosphatase 1B (PTP1B) and ETS proto-oncogene 1 (ets1) are involved in hyperglycemia-induced endothelial inflammation. In this study, we hypothesized that ets1 modulates PTP1B expression, thus playing a crucial role in hyperglycemia-induced vascular endothelial inflammation. Our results indicated that high glucose increases monocyte/endothelial adhesion, vascular cell adhesion molecule-1 (VCAM-1) expression and p65 phosphorylation in human umbilical vein endothelial cells (HUVECs). Moreover, high glucose-mediated endothelial inflammation is reversed by PTP1B silencing. In addition, high glucose increases ets1 expression in HUVECs. silencing reverses high glucose-mediated endothelial inflammation. Furthermore, the effect of ets1 overexpression is similar to that of high glucose treatment, which is counteracted by si-PTP1B. The results from ChIP assays indicated that ets1 occupies the PTP1B promoter region. Ets1 overexpression enhances PTP1B promoter activity, which is disappeared after specific binding site mutation. experiments demonstrated that the expressions of VCAM-1, PTP1B, and ets1, as well as the phosphorylation of p65 are augmented in the aorta of diabetic rats. In conclusion, ets1 contributes to hyperglycemia-mediated endothelial inflammation via upregulation of PTP1B expression.
Collapse
|
17
|
Yorek M. Treatment for Diabetic Peripheral Neuropathy: What have we Learned from Animal Models? Curr Diabetes Rev 2022; 18:e040521193121. [PMID: 33949936 PMCID: PMC8965779 DOI: 10.2174/1573399817666210504101609] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 01/07/2021] [Accepted: 02/13/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Animal models have been widely used to investigate the etiology and potential treatments for diabetic peripheral neuropathy. What we have learned from these studies and the extent to which this information has been adapted for the human condition will be the subject of this review article. METHODS A comprehensive search of the PubMed database was performed, and relevant articles on the topic were included in this review. RESULTS Extensive study of diabetic animal models has shown that the etiology of diabetic peripheral neuropathy is complex, with multiple mechanisms affecting neurons, Schwann cells, and the microvasculature, which contribute to the phenotypic nature of this most common complication of diabetes. Moreover, animal studies have demonstrated that the mechanisms related to peripheral neuropathy occurring in type 1 and type 2 diabetes are likely different, with hyperglycemia being the primary factor for neuropathology in type 1 diabetes, which contributes to a lesser extent in type 2 diabetes, whereas insulin resistance, hyperlipidemia, and other factors may have a greater role. Two of the earliest mechanisms described from animal studies as a cause for diabetic peripheral neuropathy were the activation of the aldose reductase pathway and increased non-enzymatic glycation. However, continuing research has identified numerous other potential factors that may contribute to diabetic peripheral neuropathy, including oxidative and inflammatory stress, dysregulation of protein kinase C and hexosamine pathways, and decreased neurotrophic support. In addition, recent studies have demonstrated that peripheral neuropathy-like symptoms are present in animal models, representing pre-diabetes in the absence of hyperglycemia. CONCLUSION This complexity complicates the successful treatment of diabetic peripheral neuropathy, and results in the poor outcome of translating successful treatments from animal studies to human clinical trials.
Collapse
Affiliation(s)
- Mark Yorek
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242 USA
- Department of Veterans Affairs Iowa City Health Care System, Iowa City, IA, 52246 USA
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52242 USA
| |
Collapse
|
18
|
c-Myc participates in high glucose-mediated endothelial inflammation via upregulation of IRAK1 expression in diabetic nephropathy. Cell Signal 2022; 92:110263. [DOI: 10.1016/j.cellsig.2022.110263] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 12/18/2021] [Accepted: 01/20/2022] [Indexed: 01/09/2023]
|
19
|
Fang C, Wang L, Qiao J, Chang L, He Q, Zhang X, Liu M. Differential regulation of lipopolysaccharide-induced IL-1β and TNF-α production in macrophages by palmitate via modulating TLR4 downstream signaling. Int Immunopharmacol 2021; 103:108456. [PMID: 34923420 DOI: 10.1016/j.intimp.2021.108456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 12/06/2021] [Accepted: 12/08/2021] [Indexed: 11/25/2022]
Abstract
Diabetic patients are susceptible to infectious diseases. Bacterial invasion activates immune cells such as macrophages through interaction between LPS and TLR4, and induces the expression of inflammatory mediators, including IL-1β and TNF-α, which play key roles in the elimination of infections. Unregulated overproduction or underproduction of these cytokines has been reported as a major factor in the development of septic shock, immune deficiency, and autoimmunity. Recent studies found that metabolic abnormalities of diabetes, such as hyperglycemia and dyslipidemia, played a major role in modulating the immune response. In this study, we studied the effects of palmitic acid (PA) pretreatment on LPS-induced IL-1β and TNF-α production and LPS-TLR4 signaling in macrophages. Compared with control, PA pretreatment significantly increased LPS-induced TNF-α production and secretion in macrophages. In contrast, LPS-induced IL-1β production and secretion was significantly suppressed by PA pretreatment. PA pretreatment did not affect the expression levels of TLR4 or Myd88, or the endocytosis of TLR4 in macrophages. However, PA pretreatment significantly suppressed the phosphorylation level and nuclear translocation of NF-κB, and the phosphorylation level of ERK1/2, whereas increased the phosphorylation levels of p38 and JNK. The activation of IKK which was upstream of NF-κB and ERK1/2 was attenuated, while the activation of TAK1 which was upstream of JNK and p38 was augmented by PA pretreatment. Inhibitors of NF-κB, MEK1/2, and p38 significantly decreased IL-1β expression, while JNK and p38 pathway inhibitors significantly inhibited TNF-α expression. The differential regulation of LPS-induced TNF-α and IL-1β production by PA was associated with cellular metabolism of PA, because inhibiting metabolism of PA with etomoxir or pretreatment with Br-PA which cannot be metabolized reversed these effects. We also showed that PA treatment increased acetylated IKK level which might contribute to the suppressed activation of IKK. The present study showed that LPS-induced production of TNF-α and IL-1β was regulated by different TLR4 downstream pathways in macrophages. PA differentially affected LPS-induced production of TNF-α and IL-1β in macrophages through differentially modulating these pathways. Further experiments will be needed to determine how these phenomena lead to the impaired immune response in patients with diabetes.
Collapse
Affiliation(s)
- Chunyun Fang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Lixia Wang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Jingting Qiao
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Lina Chang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China
| | - Qing He
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China.
| | - Xiaona Zhang
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China.
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
20
|
Rabbani G, Ahn SN. Review: Roles of human serum albumin in prediction, diagnoses and treatment of COVID-19. Int J Biol Macromol 2021; 193:948-955. [PMID: 34673106 PMCID: PMC8520831 DOI: 10.1016/j.ijbiomac.2021.10.095] [Citation(s) in RCA: 64] [Impact Index Per Article: 21.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 10/03/2021] [Accepted: 10/13/2021] [Indexed: 12/17/2022]
Abstract
The severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) keeps on destroying normal social integrity worldwide, bringing about extraordinary medical services, cultural and financial interruption. Individuals with diabetes have been demonstrated to be at higher risk of complications and even death when exposed to SARS-CoV-2. Regardless of pandemic scale infection, there is presently limited comprehension on the potential impact of SARS-CoV-2 on individuals with diabetes. Human serum albumin (HSA) is the most abundant circulating plasma protein in human serum and attracted more interest from researchers because most susceptible to non-enzymatic glycation reactions. Albumin down-regulates the expression of ACE2 that is the target receptor of COVID-19. Hypoalbuminemia, coagulopathy, and vascular disease have been connected in COVID-19 and appear to predict outcomes independent of age and morbidity. This review discusses the most recent evidence that the ACE/ACE2 ratio could influence by human serum albumin both the susceptibility of individuals to SARS-CoV-2 infection and the outcome of the COVID-19 disease.
Collapse
Affiliation(s)
- Gulam Rabbani
- Nano Diagnostics & Devices (NDD), B-312 IT-Medical Fusion Center, 350-27 Gumidae-ro, Gumi-si, Gyeongbuk 39253, Republic of Korea.
| | - Saeyoung Nate Ahn
- Nano Diagnostics & Devices (NDD), B-312 IT-Medical Fusion Center, 350-27 Gumidae-ro, Gumi-si, Gyeongbuk 39253, Republic of Korea; Fuzbien Technology Institute, 13 Taft Court, Rockville, MD 20850, USA.
| |
Collapse
|
21
|
Substrate stiffness modulates endothelial cell function via the YAP-Dll4-Notch1 pathway. Exp Cell Res 2021; 408:112835. [PMID: 34543658 DOI: 10.1016/j.yexcr.2021.112835] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 02/06/2023]
Abstract
Endothelial cells adapt their functions as a consequence of sensing extracellular substrate stiffness; these alterations allow them to maintain their vascular structure and function. Substrate stiffness-mediated yes-associated protein 1 (YAP) activation plays an important role in mechano-transduction and pro-angiogenic phenotype of endothelial cells, and Delta-like ligand 4 (Dll4)-Notch1 signaling is closely related to angiogenesis; however, the impact of substrate stiffness-mediated interrelation of these pathways on endothelial cell functions remains elusive. We confirmed that endothelial cells on softer substrates not only elongate cellular aspects but also attenuate YAP activation compared to cells on stiffer substrates. Endothelial cells on softer substrates also upregulate the vascular endothelial growth factor receptor 1 (VEGFR1) and VEGFR2 mRNA expression that is enhanced by VEGF stimulation. We determined that endothelial cells on softer substrates increased Dll4 expression, but not Notch1 expression, via YAP signaling. Moreover, endothelial cells on soft substrates induced not only VEGFRs upregulation but also suppression of pro-inflammatory interleukin-6 and plasminogen activator inhibitor-1 mRNA expression and the facilitation of anti-coagulant thrombomodulin and pro-coagulant tissue factor mRNA expression. Our results suggest that endothelial cells activate the YAP-Dll4-Notch signaling pathway in response to substrate stiffness and dictate cellular function.
Collapse
|
22
|
Syamsunarno MRA, Safitri R, Kamisah Y. Protective Effects of Caesalpinia sappan Linn. and Its Bioactive Compounds on Cardiovascular Organs. Front Pharmacol 2021; 12:725745. [PMID: 34603037 PMCID: PMC8479160 DOI: 10.3389/fphar.2021.725745] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 08/31/2021] [Indexed: 01/13/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death worldwide. The long-term aim of cardiovascular disease therapy is to reduce the mortality rate and decelerate the progression of cardiovascular organ damage. Current therapies focus on recovering heart function and reducing risk factors such as hyperglycemia and dyslipidemia. However, oxidative stress and inflammation are important causes of further damage to cardiovascular organs. Caesalpinia sappan Linn. (Fabaceae), a flowering tree native to tropical Asia, has antioxidant and anti-inflammatory properties. It is used as a natural dye to color food and beverages and as a traditional treatment for diarrhea, diabetes, and blood stasis. The phytochemical compounds in C. sappan, mainly the homoisoflavonoids brazilin, sappanone A, protosappanin, and hematoxylin, can potentially be used to protect cardiovascular organs. This review aims to provide updates on recent developments in research on C. sappan in relation to treatment of cardiovascular diseases. Many studies have reported protective effects of the plant’s bioactive compounds that reduce cardiac damage and enhance vasorelaxation. For example, brazilin and sappanone A have an impact on molecular and cellular changes in cardiovascular disease pathogenesis, mainly by modulating oxidative, inflammatory, and apoptotic signaling pathways. Therefore, bioactive compounds of C. sappan have the potential to be developed as therapeutic agents to combat cardiovascular diseases like myocardial infarction and vascular disease. This review could help further the understanding of the possible modulatory role of the compounds in cardiovascular diseases, thereby facilitating future studies.
Collapse
Affiliation(s)
- Mas Rizky Aa Syamsunarno
- Department of Biomedical Sciences, Faculty of Medicine, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Ratu Safitri
- Faculty of Mathematics and Natural Sciences, Universitas Padjadjaran, Jatinangor, Indonesia
| | - Yusof Kamisah
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| |
Collapse
|
23
|
Tang L, Zhang C, Yang Q, Xie H, Liu D, Tian H, Lu L, Xu JY, Li W, Xu G, Qiu Q, Liu K, Luo D, Xu GT, Zhang J. Melatonin maintains inner blood-retinal barrier via inhibition of p38/TXNIP/NF-κB pathway in diabetic retinopathy. J Cell Physiol 2021; 236:5848-5864. [PMID: 33432588 DOI: 10.1002/jcp.30269] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 12/15/2020] [Accepted: 12/28/2020] [Indexed: 01/03/2023]
Abstract
The pathophysiology of diabetic retinopathy (DR) was complex. Under hyperglycemic conditions, the release of proinflammatory cytokines and the adhesion of leukocytes to retinal capillaries contribute to endothelial damage and the subsequent increase in vascular permeability resulting in macular edema. Melatonin, produced in the retina to regulate redox reactions and dopamine metabolism, plays protective roles against inflammation and oxidative stress. Considering its anti-inflammatory and antioxidative properties, melatonin was speculated to exert beneficial effects in DR. In this study, we characterized the protective effects of melatonin on the inner blood-retinal barrier (iBRB), as well as the possible mechanisms in experimental DR. Results showed that in diabetic rat retinas, the leakage of iBRB and the expression of inflammatory factors (VEGF, TNF-α, IL-1β, ICAM-1, and MMP9) increased dramatically, while the expression of tight junction proteins (ZO-1, occludin, JAM-A, and claudin-5) decreased significantly. The above changes were largely ameliorated by melatonin. The in vivo data were confirmed in vitro. In addition, the protein expressions of p38 MAPK, NF-κB, and TXNIP were upregulated significantly in diabetes and were downregulated following melatonin treatment. Melatonin could maintain the iBRB integrity by upregulating the expression of tight junction proteins via inhibiting p38/TXNIP/NF-κB pathway, thus decreasing the production of inflammatory factors. This study may shed light on the development of melatonin-based DR therapy.
Collapse
Affiliation(s)
- Lei Tang
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Chaoyang Zhang
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China
| | - Qian Yang
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Hai Xie
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Dandan Liu
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Haibin Tian
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Lixia Lu
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Jing-Ying Xu
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Weiye Li
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Department of Ophthalmology, Drexel University College of Medicine, Philadelphia, Pennsylvania, United States
| | - Guoxu Xu
- Department of Ophthalmology, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qinghua Qiu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Kun Liu
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Dawei Luo
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| | - Guo-Tong Xu
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
| | - Jingfa Zhang
- Department of Regenerative Medicine, and Department of Pharmacology, Tongji Eye Institute, Tongji University School of Medicine, Shanghai, China
- Department of Ophthalmology, Shanghai General Hospital (Shanghai First People's Hospital), Shanghai Jiao Tong University, Shanghai, China
- National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai, China
| |
Collapse
|
24
|
Sharma S, Brown CE. Microvascular basis of cognitive impairment in type 1 diabetes. Pharmacol Ther 2021; 229:107929. [PMID: 34171341 DOI: 10.1016/j.pharmthera.2021.107929] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/23/2021] [Accepted: 05/19/2021] [Indexed: 02/07/2023]
Abstract
The complex computations of the brain require a constant supply of blood flow to meet its immense metabolic needs. Perturbations in blood supply, even in the smallest vascular networks, can have a profound effect on neuronal function and cognition. Type 1 diabetes is a prevalent and insidious metabolic disorder that progressively and heterogeneously disrupts vascular signalling and function in the brain. As a result, it is associated with an array of adverse vascular changes such as impaired regulation of vascular tone, pathological neovascularization and vasoregression, capillary plugging and blood brain barrier disruption. In this review, we highlight the link between microvascular dysfunction and cognitive impairment that is commonly associated with type 1 diabetes, with the aim of synthesizing current knowledge in this field.
Collapse
Affiliation(s)
- Sorabh Sharma
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Craig E Brown
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Psychiatry, University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
25
|
Jiao X, Guo H, Zhang G, Yin X, Li H, Chen Y. In-hospital fasting hyperglycemia and increased risk of mortality after acute coronary syndrome: a systematic overview and meta-analysis. Int J Diabetes Dev Ctries 2021. [DOI: 10.1007/s13410-020-00850-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
26
|
Diabetic retinopathy and diabetic macular oedema pathways and management: UK Consensus Working Group. Eye (Lond) 2021; 34:1-51. [PMID: 32504038 DOI: 10.1038/s41433-020-0961-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The management of diabetic retinopathy (DR) has evolved considerably over the past decade, with the availability of new technologies (diagnostic and therapeutic). As such, the existing Royal College of Ophthalmologists DR Guidelines (2013) are outdated, and to the best of our knowledge are not under revision at present. Furthermore, there are no other UK guidelines covering all available treatments, and there seems to be significant variation around the UK in the management of diabetic macular oedema (DMO). This manuscript provides a summary of reviews the pathogenesis of DR and DMO, including role of vascular endothelial growth factor (VEGF) and non-VEGF cytokines, clinical grading/classification of DMO vis a vis current terminology (of centre-involving [CI-DMO], or non-centre involving [nCI-DMO], systemic risks and their management). The excellent UK DR Screening (DRS) service has continued to evolve and remains world-leading. However, challenges remain, as there are significant variations in equipment used, and reproducible standards of DMO screening nationally. The interphase between DRS and the hospital eye service can only be strengthened with further improvements. The role of modern technology including optical coherence tomography (OCT) and wide-field imaging, and working practices including virtual clinics and their potential in increasing clinic capacity and improving patient experiences and outcomes are discussed. Similarly, potential roles of home monitoring in diabetic eyes in the future are explored. The role of pharmacological (intravitreal injections [IVT] of anti-VEGFs and steroids) and laser therapies are summarised. Generally, IVT anti-VEGF are offered as first line pharmacologic therapy. As requirements of diabetic patients in particular patient groups may vary, including pregnant women, children, and persons with learning difficulties, it is important that DR management is personalised in such particular patient groups. First choice therapy needs to be individualised in these cases and may be intravitreal steroids rather than the standard choice of anti-VEGF agents. Some of these, but not all, are discussed in this document.
Collapse
|
27
|
Yamagata K. Prevention of Endothelial Dysfunction and Cardiovascular Disease by n-3 Fatty Acids-Inhibiting Action on Oxidative Stress and Inflammation. Curr Pharm Des 2021; 26:3652-3666. [PMID: 32242776 DOI: 10.2174/1381612826666200403121952] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/11/2020] [Indexed: 01/04/2023]
Abstract
BACKGROUND Prospective cohort studies and randomized controlled trials have shown the protective effect of n-3 fatty acids against cardiovascular disease (CVD). The effect of n-3 fatty acids on vascular endothelial cells indicates their possible role in CVD prevention. OBJECTIVE Here, we describe the effect of docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) on endothelial dysfunction-caused by inflammation and oxidative stress-and their role in the development of CVD. METHODS We reviewed epidemiological studies done on n-3 fatty acids in CVD. The effect of DHA and EPA on vascular endothelial cells was examined with regard to changes in various markers, such as arteriosclerosis, inflammation, and oxidative stress, using cell and animal models. RESULTS Epidemiological studies revealed that dietary intake of EPA and DHA was associated with a reduced risk of various CVDs. EPA and DHA inhibited various events involved in arteriosclerosis development by preventing oxidative stress and inflammation associated with endothelial cell damage. In particular, EPA and DHA prevented endothelial cell dysfunction mediated by inflammatory responses and oxidative stress induced by events related to CVD. DHA and EPA also increased eNOS activity and induced nitric oxide production. CONCLUSION The effects of DHA and EPA on vascular endothelial cell damage and dysfunction may involve the induction of nitric oxide, in addition to antioxidant and anti-inflammatory effects. n-3 fatty acids inhibit endothelial dysfunction and prevent arteriosclerosis. Therefore, the intake of n-3 fatty acids may prevent CVDs, like myocardial infarction and stroke.
Collapse
Affiliation(s)
- Kazuo Yamagata
- College of Bioresource Science, Nihon University (UNBS), Kanagawa, Japan
| |
Collapse
|
28
|
Bellis A, Mauro C, Barbato E, Ceriello A, Cittadini A, Morisco C. Stress-Induced Hyperglycaemia in Non-Diabetic Patients with Acute Coronary Syndrome: From Molecular Mechanisms to New Therapeutic Perspectives. Int J Mol Sci 2021; 22:E775. [PMID: 33466656 PMCID: PMC7828822 DOI: 10.3390/ijms22020775] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Revised: 01/08/2021] [Accepted: 01/10/2021] [Indexed: 01/08/2023] Open
Abstract
Stress-induced hyperglycaemia (SIH) at hospital admission for acute coronary syndrome is associated with poor outcome, especially in patients without known diabetes. Nevertheless, insulin treatment in these subjects was not correlated with the reduction of mortality. This is likely due to the fact that SIH in the context of an acute coronary syndrome, compared to that in known diabetes, represents an epiphenomenon of other pathological conditions, such as adrenergic and renin-angiotensin system over-activity, hyperglucagonaemia, increase of circulating free fatty acids and pancreatic beta-cell dysfunction, which are not completely reversed by insulin therapy and so worsen the prognosis. Thus, SIH may be considered not only as a biomarker of organ damage, but also as an indicator of a more complex therapeutic strategy in these subjects. The aim of this review is to analyse the molecular mechanisms by which SIH may favour a worse prognosis in non-diabetic patients with acute coronary syndrome and identify new therapeutic strategies, in addition to insulin therapy, for a more appropriate treatment and improved outcomes.
Collapse
Affiliation(s)
- Alessandro Bellis
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica-Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy; (A.B.); (C.M.)
| | - Ciro Mauro
- Unità Operativa Complessa Cardiologia con UTIC ed Emodinamica-Dipartimento Emergenza Accettazione, Azienda Ospedaliera “Antonio Cardarelli”, 80131 Napoli, Italy; (A.B.); (C.M.)
| | - Emanuele Barbato
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, 80131 Napoli, Italy;
| | - Antonio Ceriello
- Department of Cardiovascular and Metabolic Diseases, IRCCS Multimedica, Sesto San Giovanni, 20099 Milan, Italy;
| | - Antonio Cittadini
- Dipartimento di Scienze Mediche Traslazionali, Università Federico II, 80131 Napoli, Italy;
| | - Carmine Morisco
- Dipartimento di Scienze Biomediche Avanzate, Università Federico II, 80131 Napoli, Italy;
| |
Collapse
|
29
|
Rajpal A, Rahimi L, Ismail‐Beigi F. Factors leading to high morbidity and mortality of COVID-19 in patients with type 2 diabetes. J Diabetes 2020; 12:895-908. [PMID: 32671936 PMCID: PMC7405270 DOI: 10.1111/1753-0407.13085] [Citation(s) in RCA: 95] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/03/2020] [Accepted: 07/09/2020] [Indexed: 01/08/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is a recent pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a novel coronavirus. Diabetes (mostly type 2 diabetes mellitus, T2DM) and hyperglycemia are among the major comorbidities in patients with COVID-19 leading to poor outcomes. Reports show that patients with diabetes and COVID-19 are at an increased risk for developing severe complications including acute respiratory distress syndrome, multi-organ failure, and death. Here we explore potential mechanistic links that could explain the observed higher morbidity and mortality in this patient population. Patients with T2DM have an underlying increased level of inflammation associated with obesity and insulin resistance in addition to other comorbidities including hypertension, obesity, cardiovascular disease, dyslipidemia, and being older. We review evidence that T2DM with hyperglycemia are among factors that lead to elevated expression of angiotensin-converting enzyme 2 (ACE2) in lungs and other tissues; ACE2 is the cellular "receptor" and port of viral entry. The preexisting chronic inflammation with augmented inflammatory response to the infection and the increasing viral load leads to extreme systemic immune response ("cytokine storm") that is strongly associated with increased severity of COVID-19. Based on the available evidence, it is recommended by a panel of experts that safe but stringent control of blood glucose, blood pressure, and lipids be carried out in patients with T2DM, measures that could potentially serve to decrease the severity of COVID-19 should these patients contract the viral infection. Once the infection occurs, then attention should be directed to proper glycemic control with use of insulin and frequent monitoring of blood glucose levels.
Collapse
Affiliation(s)
- Aman Rajpal
- Department of MedicineCase Western Reserve University and Cleveland VA Medical CenterClevelandOhioUSA
| | - Leili Rahimi
- Department of MedicineCase Western Reserve University and University Hospitals Cleveland Medical CenterClevelandOhioUSA
| | - Faramarz Ismail‐Beigi
- Department of MedicineCase Western Reserve University and Cleveland VA Medical CenterClevelandOhioUSA
- Department of MedicineCase Western Reserve University and University Hospitals Cleveland Medical CenterClevelandOhioUSA
| |
Collapse
|
30
|
Lou XQ, Wang DW, Wang JF, Du B. New thoughts on the diagnosis and treatment of patients with diabetes mellitus in relation to coronavirus disease. World J Diabetes 2020; 11:481-488. [PMID: 33269060 PMCID: PMC7672793 DOI: 10.4239/wjd.v11.i11.481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/16/2020] [Accepted: 10/05/2020] [Indexed: 02/06/2023] Open
Abstract
The coronavirus disease 2019 (COVID-19) outbreak that occurred in late 2019 has posed a huge threat to the health of all humans, especially for individuals who already have diabetes mellitus (DM). DM is one of the most serious diseases that affect human health, with high morbidity and rates of complications. Medical scientists worldwide have been working to control blood sugar levels and the complications associated with sugar level alterations, with an aim to reduce the adverse consequences of acute and chronic complications caused by DM. Patients with DM face great challenges during the pandemic owing to not only changes in the allocation of medical resources but also their abnormal autoimmune status, which reduces their resistance to infections. This increases the difficulty in treatment and the risk of mortality. This review presents, from an epidemiological viewpoint, information on the susceptibility of patients with DM to COVID-19 and the related treatment plans and strategies used in this population.
Collapse
Affiliation(s)
- Xiao-Qian Lou
- Department of Endocrinology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Da-Wei Wang
- Department of Emergency Medicine, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Jun-Feng Wang
- Department of Cardiology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Bing Du
- Department of Cardiology, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| |
Collapse
|
31
|
SET8 suppression mediates high glucose-induced vascular endothelial inflammation via the upregulation of PTEN. Exp Mol Med 2020; 52:1715-1729. [PMID: 33028948 PMCID: PMC8080680 DOI: 10.1038/s12276-020-00509-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/27/2020] [Accepted: 08/10/2020] [Indexed: 11/19/2022] Open
Abstract
Hyperglycemia-mediated endothelial inflammation participates in the pathogenesis of cardiovascular complications in subjects with diabetes. Previous studies reported that phosphatase and tensin homolog deleted on chromosome ten (PTEN) and SET8 participate in high glucose-mediated endothelial inflammation. In this study, we hypothesize that SET8 regulates PTEN expression, thus contributing to high glucose-mediated vascular endothelial inflammation. Our data indicated that plasma soluble intercellular adhesion molecule-1 (sICAM-1) and endothelial selectin (e-selectin) were increased in patients with diabetes and diabetic rats. PTEN expression was augmented in the peripheral blood mononuclear cells of patients with diabetes and in the aortic tissues of diabetic rats. Our in vitro study indicated that high glucose increased monocyte/endothelial adhesion, endothelial adhesion molecule expression and p65 phosphorylation in human umbilical vein endothelial cells (HUVECs). Moreover, high glucose led to endothelial inflammation via upregulation of PTEN. Furthermore, high glucose inhibited SET8 expression and histone H4 lysine 20 methylation (H4K20me1), a downstream target of SET8. SET8 overexpression reversed the effects of high-glucose treatment. shSET8-mediated endothelial inflammation was counteracted by siPTEN. Furthermore, SET8 was found to interact with FOXO1. siFOXO1 attenuated high glucose-mediated endothelial inflammation. FOXO1 overexpression-mediated endothelial inflammation was counteracted by siPTEN. H4K20me1 and FOXO1 were enriched in the PTEN promoter region. shSET8 increased PTEN promoter activity and augmented the positive effect of FOXO1 overexpression on PTEN promoter activity. Our in vivo study indicated that SET8 was downregulated and FOXO1 was upregulated in the peripheral blood mononuclear cells of patients with diabetes and the aortic tissues of diabetic rats. In conclusion, SET8 interacted with FOXO1 to modulate PTEN expression in vascular endothelial cells, thus contributing to hyperglycemia-mediated endothelial inflammation. High glucose levels in patients with diabetes trigger vascular inflammation by affecting the expression of key proteins in blood vessel linings. Elevated glucose causes inflammation of the endothelium, a thin layer of cells that lines blood and lymph vessels, leading to cardiovascular complications. The phosphatase and tensin homolog protein (PTEN) contributes to endothelial inflammation, but the precise mechanisms are unclear. Xuefang Shen at Fudan University in Shanghai, China, and co-workers demonstrated that elevated glucose increases PTEN expression, with increased levels of the protein found in peripheral blood cells of diabetic patients and aortic tissues of diabetic rats. In further experiments on rats, the researchers found that glucose also suppressed another protein called SET8, which contributed to increased PTEN levels. This suggests that SET8 is involved in PTEN modulation, and that both proteins influence vascular inflammation.
Collapse
|
32
|
Bennett RE, Hu M, Fernandes A, Perez-Rando M, Robbins A, Kamath T, Dujardin S, Hyman BT. Tau reduction in aged mice does not impact Microangiopathy. Acta Neuropathol Commun 2020; 8:137. [PMID: 32811565 PMCID: PMC7436970 DOI: 10.1186/s40478-020-01014-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 08/01/2020] [Indexed: 11/30/2022] Open
Abstract
Microangiopathy, including proliferation of small diameter capillaries, increasing vessel tortuosity, and increased capillary blockage by leukocytes, was previously observed in the aged rTg4510 mouse model. Similar gene expression changes related to angiogenesis were observed in both rTg4510 and Alzheimer's disease (AD). It is uncertain if tau is directly responsible for these vascular changes by interacting directly with microvessels, and/or if it contributes indirectly via neurodegeneration and concurrent neuronal loss and inflammation. To better understand the nature of tau-related microangiopathy in human AD and in tau mice, we isolated capillaries and observed that bioactive soluble tau protein could be readily detected in association with vasculature. To examine whether this soluble tau is directly responsible for the microangiopathic changes, we made use of the tetracycline-repressible gene expression cassette in the rTg4510 mouse model and measured vascular pathology following tau reduction. These data suggest that reduction of tau is insufficient to alter established microvascular complications including morphological alterations, enhanced expression of inflammatory genes involved in leukocyte adherence, and blood brain barrier compromise. These data imply that 1) soluble bioactive tau surprisingly accumulates at the blood brain barrier in human brain and in mouse models, and 2) the morphological and molecular phenotype of microvascular disturbance does not resolve with reduction of whole brain soluble tau. Additional consideration of vascular-directed therapies and strategies that target tau in the vascular space may be required to restore normal function in neurodegenerative disease.
Collapse
Affiliation(s)
- Rachel E Bennett
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| | - Miwei Hu
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Analiese Fernandes
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Marta Perez-Rando
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ashley Robbins
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Tarun Kamath
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Simon Dujardin
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| |
Collapse
|
33
|
Sacks LJ, Pham CT, Fleming N, Neoh SL, Ekinci EI. Considerations for people with diabetes during the Coronavirus Disease (COVID-19) pandemic. Diabetes Res Clin Pract 2020; 166:108296. [PMID: 32623041 PMCID: PMC7332442 DOI: 10.1016/j.diabres.2020.108296] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/24/2020] [Accepted: 06/29/2020] [Indexed: 01/08/2023]
Abstract
INTRODUCTION The severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) continues to cause havoc globally, resulting in unprecedented healthcare, societal and economic disruption. People with diabetes have been shown to be at higher risk of complications and death when exposed to pneumonia, influenza and other coronaviruses. Despite pandemic scale infection, there is currently limited understanding on the potential impact of coronavirus disease (COVID-19) on people with diabetes. AIMS (1) To characterise the outcomes of COVID-19 for people with diabetes and (2) add value to current recommendations for healthcare providers and people with diabetes to encourage optimal management. METHODS A search of PubMed, Embase and MEDLINE to March 2020 was undertaken, using search terms pertaining to diabetes, coronavirus and acute respiratory distress syndrome (ARDS). We briefly reviewed the epidemiology and pathophysiology of SARS-CoV-2 in the context of diabetes. CONCLUSION People with diabetes are at greater risk of severe infection and death with COVID-19. COVID-19 has significantly impacted the daily lives of individuals living with diabetes through financial implications, food and medication scarcity and its burden on mental health. In Australia, delivery of medical care has been adapted to reduce the risk of transmission, with a particular emphasis on telehealth and remote monitoring.
Collapse
Affiliation(s)
- Lori J Sacks
- Department of Medicine, Austin Health, Melbourne, Victoria, Australia
| | - Cecilia T Pham
- Department of Medicine, Austin Health, Melbourne, Victoria, Australia; Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia
| | - Nicola Fleming
- Department of Surgery, Austin Health, Melbourne, Victoria, Australia
| | - Sandra L Neoh
- Department of Medicine, Austin Health, Melbourne, Victoria, Australia; Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia; Department of Endocrinology, Northern Health, Melbourne, Victoria, Australia
| | - Elif I Ekinci
- Department of Medicine, Austin Health, Melbourne, Victoria, Australia; Department of Endocrinology, Austin Health, Melbourne, Victoria, Australia; Department of Medicine, The University of Melbourne, Melbourne, Victoria, Australia.
| |
Collapse
|
34
|
Gutsaeva DR, Shalaby L, Powell FL, Thounaojam MC, Abouhish H, Wetzstein SA, Jadeja RN, Kwok HF, Martin PM, Bartoli M. Inactivation of Endothelial ADAM17 Reduces Retinal Ischemia-Reperfusion Induced Neuronal and Vascular Damage. Int J Mol Sci 2020; 21:E5379. [PMID: 32751103 PMCID: PMC7432237 DOI: 10.3390/ijms21155379] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/24/2020] [Accepted: 07/27/2020] [Indexed: 12/12/2022] Open
Abstract
Retinal ischemia contributes to visual impairment in ischemic retinopathies. A disintegrin and metalloproteinase ADAM17 is implicated in multiple vascular pathologies through its ability to regulate inflammatory signaling via ectodomain shedding. We investigated the role of endothelial ADAM17 in neuronal and vascular degeneration associated with retinal ischemia reperfusion (IR) injury using mice with conditional inactivation of ADAM17 in vascular endothelium. ADAM17Cre-flox and control ADAM17flox mice were subjected to 40 min of pressure-induced retinal ischemia, with the contralateral eye serving as control. Albumin extravasation and retinal leukostasis were evaluated 48 h after reperfusion. Retinal morphometric analysis was conducted 7 days after reperfusion. Degenerate capillaries were assessed by elastase digest and visual function was evaluated by optokinetic test 14 and 7 days following ischemia, respectively. Lack of ADAM17 decreased vascular leakage and reduced retinal thinning and ganglion cell loss in ADAM17Cre-flox mice. Further, ADAM17Cre-flox mice exhibited a remarkable reduction in capillary degeneration following IR. Decrease in neurovascular degeneration in ADAM17Cre-flox mice correlated with decreased activation of caspase-3 and was associated with reduction in oxidative stress and retinal leukostasis. In addition, knockdown of ADAM17 resulted in decreased cleavage of p75NTR, the process known to be associated with retinal cell apoptosis. A decline in visual acuity evidenced by decrease in spatial frequency threshold observed in ADAM17flox mice was partially restored in ADAM17-endothelial deficient mice. The obtained results provide evidence that endothelial ADAM17 is an important contributor to IR-induced neurovascular damage in the retina and suggest that interventions directed at regulating ADAM17 activity can be beneficial for alleviating the consequences of retinal ischemia.
Collapse
Affiliation(s)
- Diana R Gutsaeva
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| | - Lamiaa Shalaby
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| | - Folami L Powell
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (F.L.P.); (R.N.J.); (P.M.M.)
| | - Menaka C Thounaojam
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| | - Hossameldin Abouhish
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
- Department of Clinical Pharmacology, Faculty of Medicine, Mansoura University, 35516 Mansoura, Egypt
| | | | - Ravirajsinh N Jadeja
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (F.L.P.); (R.N.J.); (P.M.M.)
| | - Hang Fai Kwok
- Institute of Translational Medicine, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Macau 999078;
| | - Pamela M Martin
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (F.L.P.); (R.N.J.); (P.M.M.)
| | - Manuela Bartoli
- Department of Ophthalmology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; (L.S.); (M.C.T.); (H.A.); (M.B.)
| |
Collapse
|
35
|
Omentin-1 Ameliorated Free Fatty Acid-Induced Impairment in Proliferation, Migration, and Inflammatory States of HUVECs. Cardiol Res Pract 2020; 2020:3054379. [PMID: 32300482 PMCID: PMC7140148 DOI: 10.1155/2020/3054379] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 01/20/2020] [Accepted: 02/28/2020] [Indexed: 02/05/2023] Open
Abstract
Objectives Endothelial cell injury is a critical pathological change during the development of atherosclerosis. Here, we explored the effect of omentin-1 on free fatty acid- (FFA-) induced endothelial cell injury. Methods An FFA-induced endothelial cell injury model was established to investigate the role of omentin-1 in this process. Cell proliferation was analyzed with the Cell Counting Kit assay and flow cytometry. Scratch and transwell assays were used to evaluate cell migration. Factors secreted by endothelial cells after injury were detected by western blotting, reverse-transcription quantitative polymerase chain reaction, and cellular fluorescence assay. Results Omentin-1 rescued the FFA-induced impaired proliferation and migration capabilities of human umbilical vein endothelial cells (HUVECs). It decreased the number of THP-1 cells attached to HUVECs in response to injury and inhibited the FFA-induced proinflammatory state of HUVECs. Conclusion Omentin-1 could partly ameliorate FFA-induced endothelial cell injury.
Collapse
|
36
|
Zareie M, Van Lambalgen AA, De Vriese AS, Van Gelderop E, Lameire N, Ter Wee PM, Beelen RH, van den Born J, Tangelder GJ. Increased Leukocyte Rolling in Newly Formed Mesenteric Vessels in the Rat during Peritoneal Dialysis. Perit Dial Int 2020. [DOI: 10.1177/089686080202200603] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Objective Long-term peritoneal dialysis (PD) is associated with the development of functional and structural alterations of the peritoneal membrane. The present study reports the effects of chronic exposure to PD fluid on mesenteric leukocyte–endothelium interactions, using intravital video microscopy. Methods Rats ( n = 7) received 10 mL lactate-buffered 3.86% glucose-containing PD fluid daily during a 5-week period via a subcutaneously implanted mini access port that was connected via a catheter to the peritoneal cavity. In a first control group ( n = 8), catheters were implanted but no fluid was instilled; a second control group ( n = 8) remained untreated. The number of rolling and adherent leukocytes as well as blood flow and other fluid dynamic variables were analyzed in mesenteric postcapillary (diameter 10 – 25 μ) and collecting (diameter 26 – 40 μ) venules. Neovascularization was semiquantitatively assessed after inspection of video images and by light and electron microscopy. Using FITC-labeled albumin, microvascular leakage was examined. Results Rats exposed to PD fluid showed a more than twofold increase in the number of rolling leukocytes ( p < 0.01); the number of adherent leukocytes was not changed. Furthermore, exposure to PD fluid induced severe neovascularization in rat mesentery. No microvascular leakage was observed in the various groups. The observed differences could not be explained by differences in systemic or local hemodynamic parameters or peripheral leukocyte counts, but is most likely associated with new vessel formation. Conclusions Exposure of rat peritoneal membrane to conventional PD fluid for 5 weeks affected local leukocyte–endothelium interactions. In addition, severe angiogenesis was induced, whereas microvascular permeability remained unaltered.
Collapse
Affiliation(s)
- Mohammad Zareie
- Departments of Molecular Cell Biology, University Hospital, Gent, Belgium
| | | | - An S. De Vriese
- VU University Medical Center, Amsterdam, The Netherlands; Renal Unit, University Hospital, Gent, Belgium
| | - Edwin Van Gelderop
- Departments of Molecular Cell Biology, University Hospital, Gent, Belgium
| | - Norbert Lameire
- VU University Medical Center, Amsterdam, The Netherlands; Renal Unit, University Hospital, Gent, Belgium
| | | | - Robert H.J. Beelen
- Departments of Molecular Cell Biology, University Hospital, Gent, Belgium
| | - Jacob van den Born
- Departments of Molecular Cell Biology, University Hospital, Gent, Belgium
| | | |
Collapse
|
37
|
Wang T, Zhu H, Hou Y, Duan W, Meng F, Liu Y. Ketamine attenuates high-glucose-mediated endothelial inflammation in human umbilical vein endothelial cells. Can J Physiol Pharmacol 2020; 98:156-161. [PMID: 32078386 DOI: 10.1139/cjpp-2019-0185] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hyperglycemia mediates oxidative stress, thus inducing transcription factor nuclear factor kappa B (NF-κB) activation, increasing endothelial adhesion molecule expression and monocyte/endothelial interaction, and resulting in endothelial injury. Ketamine was reported to attenuate oxidative stress in many cases. In this research, we determined whether and how ketamine protects against high-glucose-mediated augmentation of monocyte/endothelial interaction and endothelial adhesion molecule expression in human umbilical vein endothelial cells. High glucose augmented monocyte/endothelial adhesion and endothelial adhesion molecule expression. High glucose induced reactive oxygen species (ROS) production and augmented phospho-protein kinase C (p-PKC) βII expression and PKC activity. Moreover, high glucose inhibited the inhibitory subunit of nuclear factor-κBα (IκBα) expression in the cytoplasm and induced NF-κB nuclear translocation. Importantly, the effects induced by high glucose were counteracted by ketamine treatment. Further, CGP53353, a PKC βII inhibitor, inhibited high-glucose-mediated NF-κB nuclear translocation, attenuated adhesion molecule expression, and reduced monocyte/endothelial interaction. Further, these effects of ketamine against high-glucose-induced endothelial injury were inhibited by phorbol 12-myristate 13-acetate, a PKC βII activator. In conclusion, ketamine, via reducing ROS accumulation, inhibited PKC βII Ser660 phosphorylation and PKC and NF-κB activation and reduced high-glucose-induced expression of endothelial adhesion molecules and monocyte/endothelial interaction.
Collapse
Affiliation(s)
- Tianhai Wang
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Hongge Zhu
- Department of Second Pulmonary Medicine, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Yanshen Hou
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Wenming Duan
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Fufen Meng
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| | - Yahua Liu
- Department of Anaesthesiology, Xinjiang Medical University, Affiliated Tumour Hospital, Xinjiang, PR China
| |
Collapse
|
38
|
Role of Endothelial ADAM17 in Early Vascular Changes Associated with Diabetic Retinopathy. J Clin Med 2020; 9:jcm9020400. [PMID: 32024241 PMCID: PMC7073770 DOI: 10.3390/jcm9020400] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 01/25/2020] [Accepted: 01/30/2020] [Indexed: 02/06/2023] Open
Abstract
ADAM17, a disintegrin and metalloproteinase 17, is a transmembrane metalloproteinase that regulates bioavailability of multiple membrane-bound proteins via ectodomain shedding. ADAM17 activity was shown to contribute to a number of vascular pathologies, but its role in the context of diabetic retinopathy (DR) is not determined. We found that expression and enzymatic activity of ADAM17 are upregulated in human diabetic postmortem retinas and a mouse model of streptozotocin-induced diabetes. To further investigate the contribution of ADAM17 to vascular alterations associated with DR, we used human retinal endothelial cells (HREC) treated with ADAM17 neutralizing antibodies and exposed to glucidic stress and streptozotocin-induced endothelial ADAM17 knockout mice. Evaluation of vascular permeability, vascular inflammation, and oxidative stress was performed. Loss of ADAM17 in endothelial cells markedly reduced oxidative stress evidenced by decreased levels of superoxide, 3-nitrotyrosine, and 4-hydroxynonenal and decreased leukocyte-endothelium adhesive interactions in vivo and in vitro. Reduced leukostasis was associated with decreased vascular permeability and was accompanied by downregulation of intercellular adhesion molecule-1 expression. Reduction in oxidative stress in HREC was associated with downregulation of NAD(P)H oxidase 4 (Nox4) expression. Our data suggest a role for endothelial ADAM17 in DR pathogenesis and identify ADAM17 as a potential new therapeutic target for DR.
Collapse
|
39
|
Targosz-Korecka M, Malek-Zietek KE, Kloska D, Rajfur Z, Stepien EŁ, Grochot-Przeczek A, Szymonski M. Metformin attenuates adhesion between cancer and endothelial cells in chronic hyperglycemia by recovery of the endothelial glycocalyx barrier. Biochim Biophys Acta Gen Subj 2020; 1864:129533. [PMID: 31953127 DOI: 10.1016/j.bbagen.2020.129533] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 12/30/2019] [Accepted: 01/13/2020] [Indexed: 12/26/2022]
Abstract
BACKGROUND Epidemiologic studies suggest that diabetes is associated with an increased risk of cancer. Concurrently, clinical trials have shown that metformin, which is a first-line antidiabetic drug, displays anticancer activity. The underlying mechanisms for these effects are, however, still not well recognized. METHODS Methods based on atomic force microscopy (AFM) were used to directly evaluate the influence of metformin on the nanomechanical and adhesive properties of endothelial and cancer cells in chronic hyperglycemia. AFM single-cell force spectroscopy (SCFS) was used to measure the total adhesion force and the work of detachment between EA.hy926 endothelial cells and A549 lung carcinoma cells. Nanoindentation with a spherical AFM probe provided information about the nanomechanical properties of cells, particularly the length and grafting density of the glycocalyx layer. Fluorescence imaging was used for glycocalyx visualization and monitoring of E-selectin and ICAM-1 expression. RESULTS SCFS demonstrated that metformin attenuates adhesive interactions between EA.hy926 endothelial cells and A549 lung carcinoma cells in chronic hyperglycemia. Nanoindentation experiments, confirmed by confocal microscopy imaging, revealed metformin-induced recovery of endothelial glycocalyx length and density. The recovery of endothelial glycocalyx was correlated with a decrease in the surface expression of E-selectin and ICAM-1. CONCLUSION Our results identify metformin-induced endothelial glycocalyx restoration as a key factor responsible for the attenuation of adhesion between EA.hy926 endothelial cells and A549 lung carcinoma cells. GENERAL SIGNIFICANCE Metformin-induced glycocalyx restoration and the resulting attenuation of adhesive interactions between the endothelium and cancer cells may account for the antimetastatic properties of this drug.
Collapse
Affiliation(s)
- Marta Targosz-Korecka
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland.
| | - Katarzyna Ewa Malek-Zietek
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Damian Kloska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Zenon Rajfur
- Department of Molecular and Interfacial Biophysics, Faculty of Physics, Astronomy and Applied Computer Science, Krakow, Poland
| | - Ewa Łucja Stepien
- Department of Medical Physics, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| | - Marek Szymonski
- Department of Physics of Nanostructures and Nanotechnology, Faculty of Physics, Astronomy and Applied Computer Science, Jagiellonian University, Krakow, Poland
| |
Collapse
|
40
|
Pang B, Li M, Song J, Li QW, Wang J, Di S, Tong XL, Ni Q. Luo Tong formula attenuates retinal inflammation in diabetic rats via inhibition of the p38MAPK/NF-κB pathway. Chin Med 2020; 15:5. [PMID: 31956338 PMCID: PMC6958683 DOI: 10.1186/s13020-019-0284-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 12/27/2019] [Indexed: 12/20/2022] Open
Abstract
Background Diabetic retinopathy (DR) is a serious microvascular complication of diabetes and remains the leading cause of blindness in adults. Retinal inflammation is playing a crucial role in the development of DR, and targeting inflammatory mediators is a promising strategy for controlling DR. Here, we investigated compound Chinese medicine Luo Tong formula (LTF) alleviated retinal inflammatory responses in a STZ-induced diabetic rat model. Methods Sprague–Dawley rats were divided into four groups: control, streptozotocin-induced diabetic, LTF-treated diabetic, and calcium dobesilate (CaD)-treated diabetic rats. Blood samples were collected for blood glucose examination. Hematoxylin–eosin and periodic acid-Schiff staining were conducted for light microscopy observations. Retinal cell apoptosis was detected using the TUNEL assay. Proteins expression was quantified by Western blotting and/or immunohistochemistry, and gene expression was assessed by real-time PCR. Results Diabetic rats showed significant increases in the expression of tumor necrosis factor α (TNF-α), interleukin-1β (IL-1β), monocyte chemotactic protein-1 (MCP-1), intercellular adhesion molecule-1 (ICAM-1), nuclear factor-κB (NF-κB), and the phospho-p38 mitogen-activated protein kinase (p-p38-MAPK)/p38 MAPK ratio compared to control rats. LTF treatment significantly improved both retinal and pancreatic pathological injury, LTF treatment also inhibited inducible the p-p38 MAPK/p38 MAPK ratio and NF-κB activation and decreased the subsequent induction of the retinal expression of proinflammatory mediators TNF-α, IL-1β, MCP-1 and ICAM-1 compared to diabetic rats. LTF also exhibited a protective effect on islet function. Conclusions LTF before the onset of DR can alleviate retinal pathological injury, LTF may play an anti-inflammatory role by inhibiting p38-MAPK and then inhibiting NF-κB pathway. But further studies are needed to confirm this conclusion. Trial registration This is an animal experiment, trial registration is not necessary.
Collapse
Affiliation(s)
- Bing Pang
- 1Department of Endocrinology, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, 6 Floors of Inpatients Building, 5 Beixiange Street, Xicheng District, Beijing, 100053 China
| | - Min Li
- 2Molecular Biology Laboratory, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Jun Song
- 1Department of Endocrinology, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, 6 Floors of Inpatients Building, 5 Beixiange Street, Xicheng District, Beijing, 100053 China
| | - Qing-Wei Li
- 1Department of Endocrinology, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, 6 Floors of Inpatients Building, 5 Beixiange Street, Xicheng District, Beijing, 100053 China
| | - Jia Wang
- 3General Department, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, Beijing, 100053 China
| | - Sha Di
- 1Department of Endocrinology, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, 6 Floors of Inpatients Building, 5 Beixiange Street, Xicheng District, Beijing, 100053 China
| | - Xiao-Lin Tong
- 4Department of Endocrinology, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, Room 432, Administration Building, 5 Beixiange Street, Xicheng District, Beijing, 100053 China
| | - Qing Ni
- 1Department of Endocrinology, Guang'anmen Hospital of China Academy of Chinese Medical Sciences, 6 Floors of Inpatients Building, 5 Beixiange Street, Xicheng District, Beijing, 100053 China
| |
Collapse
|
41
|
Antinozzi C, Sgrò P, Di Luigi L. Advantages of Phosphodiesterase Type 5 Inhibitors in the Management of Glucose Metabolism Disorders: A Clinical and Translational Issue. Int J Endocrinol 2020; 2020:7078108. [PMID: 32774364 PMCID: PMC7407035 DOI: 10.1155/2020/7078108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/06/2020] [Accepted: 07/14/2020] [Indexed: 12/17/2022] Open
Abstract
Among metabolic diseases, carbohydrate metabolism disorders are the most widespread. The most common glucose pathological conditions are acquired and may increase the risk of type 2 diabetes, obesity, heart diseases, stroke, and kidney insufficiency. Phosphodiesterase type 5 inhibitors (PDE5i) have long been used as an effective therapeutic option for the treatment of erectile dysfunction (ED). Different studies have demonstrated that PDE5i, by sensitizing insulin target tissues to insulin, play an important role in controlling the action of insulin and glucose metabolism, highlighting the protective action of these drugs against metabolic diseases. In this review, we report the latest knowledge about the role of PDE5i in the metabolic diseases of insulin resistance and type 2 diabetes, highlighting clinical aspects and potential treatment approaches. Although various encouraging data are available, further in vivo and in vitro studies are required to elucidate the mechanism of action and their clinical application in humans.
Collapse
Affiliation(s)
- Cristina Antinozzi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy
| | - Paolo Sgrò
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy
| | - Luigi Di Luigi
- Unit of Endocrinology, Department of Movement, Human and Health Sciences, University of Rome “Foro Italico”, 00135 Rome, Italy
| |
Collapse
|
42
|
Inflammation and Vascular Ageing: From Telomeres to Novel Emerging Mechanisms. High Blood Press Cardiovasc Prev 2019; 26:321-329. [DOI: 10.1007/s40292-019-00331-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 07/12/2019] [Indexed: 12/20/2022] Open
|
43
|
Hitsumoto T. Clinical Significance of Skin Autofluorescence in Elderly Patients With Long-Standing Persistent Atrial Fibrillation. Cardiol Res 2019; 10:181-187. [PMID: 31236181 PMCID: PMC6575111 DOI: 10.14740/cr885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Accepted: 05/27/2019] [Indexed: 11/17/2022] Open
Abstract
Background Recent clinical studies have demonstrated the importance of skin autofluorescence as a cardiovascular risk factor. However, data regarding the relationship between skin autofluorescence and atrial fibrillation are limited. The aim of this study was to clarify the clinical significance of skin autofluorescence in elderly patients with long-standing persistent atrial fibrillation. Methods This cross-sectional study enrolled 112 elderly patients with long-standing persistent atrial fibrillation who were treated medically (46 men and 66 women; mean age, 81 ± 9 years). The association between skin autofluorescence and various clinical parameters was examined. Results Significant relationships were observed between skin autofluorescence and CHADS2 score (r = 0.53, P < 0.001), high-sensitivity cardiac troponin T level (r = 0.43, P < 0.001), reactive oxygen metabolite levels (r = 0.52, P < 0.001), and whole blood passage time (r = 0.45, P < 0.001). Furthermore, multiple regression analyses showed that these clinical parameters were independent variables when skin autofluorescence was used as a subordinate factor. Receiver-operating characteristic curve analysis indicated that the risk values of skin autofluorescence for high CHADS2 scores (≥ 2) or elevated high-sensitivity cardiac troponin T levels (> 0.014ng/mL) were 2.6 arbitrary units (AU) and 2.7 AU, respectively. Conclusions The findings of this study indicated that skin autofluorescence may be a prognostic factor in elderly patients with long-standing persistent atrial fibrillation. The risk value of skin autofluorescence was considered as 2.6 AU or 2.7 AU.
Collapse
Affiliation(s)
- Takashi Hitsumoto
- Hitsumoto Medical Clinic, 2-7-7, Takezakicyou, Shimonoseki City, Yamaguchi 750-0025, Japan.
| |
Collapse
|
44
|
Wang X, Sun Y, Shao X. Predictive value of procalcitonin for infection of patients with type-2 diabetes mellitus. Exp Ther Med 2019; 18:722-728. [PMID: 31258707 DOI: 10.3892/etm.2019.7611] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2018] [Accepted: 04/26/2019] [Indexed: 12/11/2022] Open
Abstract
The aim of the present study was to investigate the predictive value of procalcitonin (PCT) for infection in patients with type 2 diabetes mellitus (T2DM). A retrospective analysis of 178 patients with T2DM who were divided into non-infection, local infection and sepsis groups was conducted; in conjunction with 33 healthy control patients. Clinicopathological characteristics and inflammatory indicators were compared between the four groups. Patients in the non-infection group exhibited significantly higher PCT levels compared with healthy controls (P=0.002). In addition, PCT, C-reactive protein (CRP), white blood cell count and neutrophil percent were significantly different amongst patients with T2MD across different infection groups (all P<0.001) with the following rank order: Sepsis group > local infection group > non-infection group (all P<0.05). In addition, the following observations were made: i) PCT and CRP demonstrated larger areas under the curve (AUC) for predicting local infection (0.804 and 0.741, respectively); ii) PCT displayed lower sensitivity of only 21.8% at its classical cutoff value (0.500 ng/ml) whereas CRP exhibited higher sensitivity and specificity at 64.1 and 76.1%, respectively, at its classical cutoff value (10.0 mg/l); and iii) PCT exhibited the largest AUC (0.914) for predicting sepsis with high sensitivity and specificity (86.4 and 84.5%, respectively) at its cutoff value (0.990 ng/ml). Patients with T2DM without infection demonstrated higher baseline PCT levels. The present study clarified the value of PCT in predicting infection of T2DM patients. The application of PCT to predict local infection in patients with T2DM was identified to be inferior to CRP, but its ability to predict sepsis was concluded to be the best when compared with CRP, white blood cell count and neutrophil percent.
Collapse
Affiliation(s)
- Xinshui Wang
- Department of Endocrinology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Yanbei Sun
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Xiaonan Shao
- Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
45
|
Hitsumoto T. Clinical Significance of Low Blood Testosterone Concentration in Men as a Cardiovascular Risk Factor From the Perspective of Blood Rheology. Cardiol Res 2019; 10:106-113. [PMID: 31019640 PMCID: PMC6469906 DOI: 10.14740/cr858] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 04/19/2019] [Indexed: 01/08/2023] Open
Abstract
Background Recent clinical studies have indicated the importance of low blood testosterone concentration or whole blood passage time (WBPT) which reflects blood rheology as a cardiovascular risk factor. On the contrary, there are no reports regarding the association of blood testosterone concentrations and WBPT. This cross-sectional study aimed to elucidate the clinical significance of low blood testosterone concentration in men as a cardiovascular risk factor from the perspective of blood rheology using WBPT. Methods In total, 382 male patients with traditional cardiovascular risk factor and no history of cardiovascular disease (age (mean ± standard deviation (SD)), 64 ± 10 years) were enrolled. Serum total testosterone concentration (T-T) was measured as a marker of testosterone level in vivo, and WBPT was also measured using microchannel array flow analyzer as a commercial device. The relationship between T-T and WBPT was evaluated. Results There was a significantly negative correlation between T-T and WBPT (r = -0.45; P < 0.001). Furthermore, multiple regression analysis revealed that T-T (β = -0.25; P < 0.001) could be selected as an independent variable when WBPT was used as a subordinate factor. According to receiver operating characteristic curve analysis and the result of the previous report that determined WBPT of > 72.4 s as a risk for incidence of primary cardiovascular disease, T-T of < 551.4 ng/dL is the optimal cut-off point for discriminating high WBPT. Conclusions The study results showed that T-T is independently and inversely associated with WBPT in male patients with traditional cardiovascular risk factor and no history of cardiovascular disease. In addition, this study suggests that the incidence of primary cardiovascular events can be prevented by maintaining T-T at approximately ≥ 550 ng/dL from the perspective of blood rheology.
Collapse
Affiliation(s)
- Takashi Hitsumoto
- Hitsumoto Medical Clinic, 2-7-7, Takezakicyou, Shimonoseki City, Yamaguchi, 750-0025, Japan.
| |
Collapse
|
46
|
Hijmans JG, Bammert TD, Stockelman KA, Reiakvam WR, Greiner JJ, DeSouza CA. High glucose-induced endothelial microparticles increase adhesion molecule expression on endothelial cells. Diabetol Int 2019; 10:143-147. [PMID: 31139533 PMCID: PMC6506489 DOI: 10.1007/s13340-018-0375-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 09/07/2018] [Indexed: 12/17/2022]
Abstract
The experimental aim of this study was to determine, in vitro, the effects of glucose-induced EMPs on endothelial cell expression of E-selectin, intercellular cell adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 and platelet cell adhesion molecule-1 (PECAM-1). Human umbilical vein endothelial cells (HUVECs) were cultured (3rd passage) and plated in 6-well plates at a density of 5.0 × 105 cells/condition. HUVECs were incubated with media containing either 25 mM d-glucose (concentration representing a hyperglycemic state) or 5 mM d-glucose (normoglycemic condition) for 48 h to generate EMPs. EMP identification (CD144+) and concentration were determined by flow cytometry. HUVECs (3 × 106 cells/condition) were treated with either high glucose-derived EMPs (hgEMPs) or normal glucose-derived (ngEMPs) for 24 h and surface expression of E-selectin (CD62E-PE), ICAM-1 (CD54-FITC), VCAM-1 (CD106-APC) and PECAM-1 (CD31-BV) was assessed by flow cytometry and reported as mean fluorescent intensity (MFI). Hyperglycemic-derived EMPs induced significantly higher surface expression of E-selectin (2614 ± 132 vs. 2010 ± 204 MFI), ICAM-1 (2110 ± 81 vs. 1688 ± 152 MFI), VCAM-1 (3589 ± 431 vs. 2134 ± 386) and PECAM-1 (4237 ± 395 vs. 2525 ± 269 MFI) on endothelial cells than EMPs from normoglycemic conditions. Microparticle-induced cell adhesion molecule expression provides potential novel mechanistic insight regarding the accelerated risk of atherosclerotic vascular disease associated with hyperglycemia.
Collapse
Affiliation(s)
- Jamie G. Hijmans
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, 354 UCB, 1725 Pleasant St, Boulder, CO 80309 USA
| | - Tyler D. Bammert
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, 354 UCB, 1725 Pleasant St, Boulder, CO 80309 USA
| | - Kelly A. Stockelman
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, 354 UCB, 1725 Pleasant St, Boulder, CO 80309 USA
| | - Whitney R. Reiakvam
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, 354 UCB, 1725 Pleasant St, Boulder, CO 80309 USA
| | - Jared J. Greiner
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, 354 UCB, 1725 Pleasant St, Boulder, CO 80309 USA
| | - Christopher A. DeSouza
- Integrative Vascular Biology Laboratory, Department of Integrative Physiology, University of Colorado, 354 UCB, 1725 Pleasant St, Boulder, CO 80309 USA
| |
Collapse
|
47
|
Hitsumoto T. Clinical Impact of Hemorheology on Subclinical Myocardial Injury in Patients with Hypertension. J Clin Med Res 2018; 10:928-935. [PMID: 30425766 PMCID: PMC6225855 DOI: 10.14740/jocmr3652] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 12/21/2022] Open
Abstract
Background The blood concentration of high-sensitivity cardiac troponin T (hs-cTnT) is a useful biomarker for myocardial injury or the pathogenesis of hypertension. Little is known about the relationship between hemorheology and myocardial injury in patients with hypertension. This cross-sectional study aimed to clarify the clinical impact of hemorheology on subclinical myocardial injury assessed with a microchannel array flow analyzer (MC-FAN) and its impact on hs-cTnT in patients with hypertension. Methods A total of 447 outpatients (men: 181; women: 266; mean age: 65 ± 13 years), with no history of cardiovascular disease, including admission for heart failure, who were undergoing treatment for hypertension, were enrolled. Whole blood passage time (WBPT) as a marker of hemorheology was measured with a MC-FAN, and the relationship between hs-cTnT levels and various clinical parameters, including WBPT, was examined. Results hs-cTnT levels were detected in 400 patients (89.5%). WBPT was significantly higher in patients with detectable hs-cTnT levels than in those with undetectable hs-cTnT levels (60.5 ± 16.8 s versus 50.2 ± 14.2 s, P < 0.001). In patients with detectable hs-cTnT levels, there was a significant positive correlation between WBPT and hs-cTnT level (r = 0.33; P < 0.001). Multiple regression analysis revealed that WBPT was an independent variable when hs-cTnT was a subordinate factor (β = 0.15; P < 0.01). Receiver-operating characteristic curve analysis indicated that a cutoff value for WBPT of 55.6 s yielded the largest area under the curve (0.744; P < 0.001) for discriminating high hs-cTnT levels as ≥ 0.014 ng/mL. Conclusion The results indicate that WBPT is independently associated with hs-cTnT in hypertensive patients with no history of cardiovascular events, suggesting that impairment of hemorheology in small cardiac vessels causes subclinical myocardial injury. In addition, the study suggests that progression of myocardial injury can be prevented by maintaining WBPT at approximately ≤ 55 s.
Collapse
|
48
|
Wang Z, Zhao H, Guan W, Kang X, Tai X, Shen Y. Metabolic memory in mitochondrial oxidative damage triggers diabetic retinopathy. BMC Ophthalmol 2018; 18:258. [PMID: 30249212 PMCID: PMC6154827 DOI: 10.1186/s12886-018-0921-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2018] [Accepted: 09/11/2018] [Indexed: 01/09/2023] Open
Abstract
Background Diabetic retinopathy (DR) is a microvascular complication induced by high blood glucose. This study was conducted to investigate the effect of metabolic memory on mitochondrial oxidative damage-induced DR. Methods Rat retinal endothelial cells (rRECs) were isolated from SD rats and treated with high glucose (20 mM) for various times and then cultured in normal glucose (5.6 mM) medium for 2 days. The cells were assayed for the expression of respiratory chain complexes cytochrome c oxidase subunit 1 (CO1) and NADPH-1 using RT-PCR, mitochondrial membrane potentials and reactive oxygen species (ROS) production using flow cytometry and apoptosis using Annexin V/PI flow cytometry. Results rRECs displayed like short spindles after cultured for 9–10 days and reached 100% confluency. Compared with the control grown in normal glucose (5.6 mM) medium, rRECs exposed to high glucose medium for 3, 12 and 24 h had significantly increased mRNA levels of CO1 and NAPDH-1 even after being shifted back to normal glucose medium. They also had lower mitochondrial membrane potential (89.13% vs 78.21%, p < 0.05), cytochrome C level (1 in control vs 0.25 after 24 h exposure to high glucose, p < 0.05 and higher ROS production (2.77% in control vs 9.00% after 12 h exposure to high glucose, p < 0.05) and apoptosis (7.15% in control vs and 29.91% after 24 h exposure to high glucose, p < 0.05). Conclusion It is likely that mitochondrial oxidative damage triggers metabolic memory via ROS overproduction, leading to diabetic retinopathy.
Collapse
Affiliation(s)
- Zhaoge Wang
- Center of Myopia, the Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, China
| | - Haixia Zhao
- Center of Myopia, the Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, China
| | - Wenying Guan
- Center of Myopia, the Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, China
| | - Xin Kang
- Center of Myopia, the Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, China
| | - Xue Tai
- Center of Myopia, the Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, China
| | - Ying Shen
- Center of Myopia, the Affiliated Hospital of Inner Mongolia Medical University, 1 Tongdao North Street, Hohhot, 010050, China.
| |
Collapse
|
49
|
Homme RP, Singh M, Majumder A, George AK, Nair K, Sandhu HS, Tyagi N, Lominadze D, Tyagi SC. Remodeling of Retinal Architecture in Diabetic Retinopathy: Disruption of Ocular Physiology and Visual Functions by Inflammatory Gene Products and Pyroptosis. Front Physiol 2018; 9:1268. [PMID: 30233418 PMCID: PMC6134046 DOI: 10.3389/fphys.2018.01268] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 08/21/2018] [Indexed: 02/06/2023] Open
Abstract
Diabetic patients suffer from a host of physiological abnormalities beyond just those of glucose metabolism. These abnormalities often lead to systemic inflammation via modulation of several inflammation-related genes, their respective gene products, homocysteine metabolism, and pyroptosis. The very nature of this homeostatic disruption re-sets the overall physiology of diabetics via upregulation of immune responses, enhanced retinal neovascularization, upregulation of epigenetic events, and disturbances in cells' redox regulatory system. This altered pathophysiological milieu can lead to the development of diabetic retinopathy (DR), a debilitating vision-threatening eye condition with microvascular complications. DR is the most prevalent cause of irreversible blindness in the working-age adults throughout the world as it can lead to severe structural and functional remodeling of the retina, decreasing vision and thus diminishing the quality of life. In this manuscript, we attempt to summarize recent developments and new insights to explore the very nature of this intertwined crosstalk between components of the immune system and their metabolic orchestrations to elucidate the pathophysiology of DR. Understanding the multifaceted nature of the cellular and molecular factors that are involved in DR could reveal new targets for effective diagnostics, therapeutics, prognostics, preventive tools, and finally strategies to combat the development and progression of DR in susceptible subjects.
Collapse
Affiliation(s)
- Rubens P. Homme
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Mahavir Singh
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Avisek Majumder
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY, United States
| | - Akash K. George
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Kavya Nair
- Eye and Vision Science Laboratory, Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Harpal S. Sandhu
- Department of Ophthalmology and Visual Sciences, University of Louisville School of Medicine, Louisville, KY, United States
- Kentucky Lions Eye Center, University of Louisville School of Medicine, Louisville, KY, United States
| | - Neetu Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - David Lominadze
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States
| |
Collapse
|
50
|
Ramos-Prol A, Hervás-Marín D, Rodríguez-Medina B, Rubio-Almanza M, Berenguer M, Moya-Herraiz Á, Merino-Torres JF. Intensified blood glucose treatment in diabetic patients undergoing a liver transplant: impact on graft evolution at 3 months and at 5 years. J Endocrinol Invest 2018; 41:821-829. [PMID: 29289983 DOI: 10.1007/s40618-017-0810-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 12/14/2017] [Indexed: 01/08/2023]
Abstract
PURPOSE The debate about the impact of intensified hyperglycemia treatment is still ranging. The main objective was to assess whether intensive glycemic control in hospitalized diabetic patients undergoing a liver transplant is associated with a lower rate of graft rejection at 3 months and at 5 years post-transplant. METHODS Cross-sectional study comparing a cohort of patients undergoing liver transplant in 2010 and 2011, in whom an intensive insulin protocol was applied, with a retrospective group of patients undergoing a liver transplant in 2005 and 2006, in whom a conventional insulin protocol was applied. Both diabetics and non-diabetics were compared. As intensive insulin therapy is applied mainly in diabetic patients, it is expected that, when comparing both periods, the treatment would only benefit those patients. RESULTS The logistic regression model showed a statistically significant interaction between the treatment group and the presence of diabetes for the rejection rate 3 months and 5 years post-transplant. At both time points, the intensive insulin treatment group had lower rejection rates in the case of diabetic patients, which did not occur in non-diabetic patients. CONCLUSIONS Our study shows a decrease in the rate of liver graft rejection in diabetic patients undergoing intensive insulin treatment.
Collapse
Affiliation(s)
- A Ramos-Prol
- Endocrinology and Nutrition Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Unidad Mixta de Investigación de Endocrinología, Nutrición y Dietética, Instituto de Investigación Sanitaria La Fe (Health Research Institute La Fe), Valencia, Spain
- Department of Internal Medicine (Endocrinology and Nutrition), Hospital Francesc de Borja, Gandía, Spain
| | - D Hervás-Marín
- Biostatistics Unit, Health Research Institute La Fe, Valencia, Spain
| | - B Rodríguez-Medina
- Liver Transplantation and Hepatology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - M Rubio-Almanza
- Endocrinology and Nutrition Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Unidad Mixta de Investigación de Endocrinología, Nutrición y Dietética, Instituto de Investigación Sanitaria La Fe (Health Research Institute La Fe), Valencia, Spain
| | - M Berenguer
- Liver Transplantation and Hepatology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - Á Moya-Herraiz
- Liver Transplantation and Hepatology Unit, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | - J F Merino-Torres
- Endocrinology and Nutrition Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain.
- Unidad Mixta de Investigación de Endocrinología, Nutrición y Dietética, Instituto de Investigación Sanitaria La Fe (Health Research Institute La Fe), Valencia, Spain.
| |
Collapse
|