101
|
Hendrikson J, Liu Y, Ng WH, Lee JY, Lim AH, Loh JW, Ng CC, Ong WS, Tan JWS, Tan QX, Ng G, Shannon NB, Lim WK, Lim TK, Chua C, Wong JSM, Tan GHC, So JBY, Yeoh KG, Teh BT, Chia CS, Soo KC, Kon OL, Tan IB, Chan JY, Teo MCC, Ong CAJ. Ligand-mediated PAI-1 inhibition in a mouse model of peritoneal carcinomatosis. Cell Rep Med 2022; 3:100526. [PMID: 35243423 PMCID: PMC8861959 DOI: 10.1016/j.xcrm.2022.100526] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 11/22/2021] [Accepted: 01/19/2022] [Indexed: 12/28/2022]
Abstract
Peritoneal carcinomatosis (PC) present a ubiquitous clinical conundrum in all intra-abdominal malignancies. Via functional and transcriptomic experiments of ascites-treated PC cells, we identify STAT3 as a key signaling pathway. Integrative analysis of publicly available databases and correlation with clinical cohorts (n = 7,359) reveal putative clinically significant activating ligands of STAT3 signaling. We further validate a 3-biomarker prognostic panel in ascites independent of clinical covariates in a prospective study (n = 149). Via single-cell sequencing experiments, we uncover that PAI-1, a key component of the prognostic biomarker panel, is largely secreted by fibroblasts and mesothelial cells. Molecular stratification of ascites using PAI-1 levels and STAT3 activation in ascites-treated cells highlight a therapeutic opportunity based on a phenomenon of paracrine addiction. These results are recapitulated in patient-derived ascites-dependent xenografts. Here, we demonstrate therapeutic proof of concept of direct ligand inhibition of a prognostic target within an enclosed biological space.
Collapse
Affiliation(s)
- Josephine Hendrikson
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Ying Liu
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Wai Har Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jing Yi Lee
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Abner Herbert Lim
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jui Wan Loh
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Cedric C.Y. Ng
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Whee Sze Ong
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Joey Wee-Shan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Qiu Xuan Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Gillian Ng
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Nicholas B. Shannon
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Weng Khong Lim
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Tony K.H. Lim
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
| | - Clarinda Chua
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Jolene Si Min Wong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Grace Hwei Ching Tan
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Jimmy Bok Yan So
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
| | - Khay Guan Yeoh
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
| | - Bin Tean Teh
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - Claramae Shulyn Chia
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Khee Chee Soo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Oi Lian Kon
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Iain Beehuat Tan
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| | - Jason Yongsheng Chan
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
| | - Melissa Ching Ching Teo
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
| | - Chin-Ann J. Ong
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
| | - on behalf of the Singapore Peritoneal Oncology Study (SPOS) Group and Singapore Gastric Cancer Consortium (SGCC)
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, National Cancer Centre Singapore, 11 Hospital Cresent, Singapore 169610, Singapore
- Department of Sarcoma, Peritoneal and Rare Tumours (SPRinT), Division of Surgery and Surgical Oncology, Singapore General Hospital, Singapore 169608, Singapore
- Laboratory of Applied Human Genetics, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Cancer Discovery Hub, National Cancer Centre Singapore, Singapore 169610, Singapore
- Laboratory of Cancer Epigenome, Division of Medical Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169609, Singapore
- Cancer and Stem Biology Signature Research Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Anatomical Pathology, Singapore General Hospital, Singapore 169856, Singapore
- Pathology Academic Clinical Program, SingHealth Duke-NUS Academic Medical Centre, Singapore 168753, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore 169610, Singapore
- SingHealth Duke-NUS Oncology Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- SingHealth Duke-NUS Surgery Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
- Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Division of Surgical Oncology, National University Cancer Institute, National University Health System, Singapore 119074, Singapore
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119077, Singapore
- Department of Gastroenterology and Hepatology, National University Hospital, Singapore 119074, Singapore
- Institute of Molecular and Cell Biology, A∗STAR Research Entities, Singapore 138673, Singapore
- Laboratory of Applied Cancer Genomics, Genome Institute of Singapore, A∗STAR Research Entities, Singapore 138672, Singapore
| |
Collapse
|
102
|
Phillippi DT, Daniel S, Pusadkar V, Youngblood VL, Nguyen KN, Azad RK, McFarlin BK, Lund AK. Inhaled diesel exhaust particles result in microbiome-related systemic inflammation and altered cardiovascular disease biomarkers in C57Bl/6 male mice. Part Fibre Toxicol 2022; 19:10. [PMID: 35135577 PMCID: PMC8827295 DOI: 10.1186/s12989-022-00452-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 01/31/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The gut microbiota plays a vital role in host homeostasis and is associated with inflammation and cardiovascular disease (CVD) risk. Exposure to particulate matter (PM) is a known mediator of inflammation and CVD and is reported to promote dysbiosis and decreased intestinal integrity. However, the role of inhaled traffic-generated PM on the gut microbiome and its corresponding systemic effects are not well-characterized. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) alters the gut microbiome and promotes microbial-related inflammation and CVD biomarkers. 4-6-week-old male C57Bl/6 mice on either a low-fat (LF, 10% fat) or high-fat (HF, 45% fat) diet were exposed via oropharyngeal aspiration to 35 μg DEP suspended in 35 μl saline or saline only (CON) 2x/week for 30 days. To determine whether probiotics could prevent diet or DEP exposure mediated alterations in the gut microbiome or systemic outcomes, a subset of animals on the HF diet were treated orally with 0.3 g/day (~ 7.5 × 108 CFU/day) of Winclove Ecologic® Barrier probiotics throughout the study. RESULTS Our results show that inhaled DEP exposure alters gut microbial profiles, including reducing Actinobacteria and expanding Verrucomicrobia and Proteobacteria. We observed increased circulating LPS, altered circulating cytokines (IL-1α, IL-3, IL-13, IL-15, G-CSF, LIF, MIP-2, and TNF-α), and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in DEP-exposed and/or HF diet mice. Furthermore, probiotics attenuated the observed reduction of Actinobacteria and expansion of Proteobacteria in DEP-exposed and HF-diet mice. Probiotics mitigated circulating cytokines (IL-3, IL-13, G-CSF, RANTES, and TNF- α) and CVD biomarkers (siCAM, PAI-1, sP-Selectin, thrombomodulin, and PECAM) in respect to DEP-exposure and/or HF diet. CONCLUSION Key findings of this study are that inhaled DEP exposure alters small intestinal microbial profiles that play a role in systemic inflammation and early CVD biomarkers. Probiotic treatment in this study was fundamental in understanding the role of inhaled DEP on the microbiome and related systemic inflammatory and CVD biomarkers.
Collapse
Affiliation(s)
- Danielle T. Phillippi
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Sarah Daniel
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Vaidehi Pusadkar
- BioDiscovery Institute, Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
| | - Victoria L. Youngblood
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Kayla N. Nguyen
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| | - Rajeev K. Azad
- BioDiscovery Institute, Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
- Department of Mathematics, University of North Texas, Denton, TX 76203 USA
| | - Brian K. McFarlin
- Department of Biological Sciences, University of North Texas, Denton, TX 76203 USA
- UNT Applied Physiology Laboratory, University of North Texas, Denton, TX 76203 USA
| | - Amie K. Lund
- Department of Biological Sciences, Advanced Environmental Research Institute, University of North Texas, EESAT – 215, 1704 W. Mulberry, Denton, TX 76203 USA
| |
Collapse
|
103
|
Ahmad F, Kannan M, Ansari AW. Role of SARS-CoV-2 -induced cytokines and growth factors in coagulopathy and thromboembolism. Cytokine Growth Factor Rev 2022; 63:58-68. [PMID: 34750061 PMCID: PMC8541834 DOI: 10.1016/j.cytogfr.2021.10.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 10/19/2021] [Accepted: 10/20/2021] [Indexed: 01/08/2023]
Abstract
Severe COVID-19 patients frequently present thrombotic complications which commonly lead to multiorgan failure and increase the risk of death. Severe SARS-CoV-2 infection induces the cytokine storm and is often associated with coagulation dysfunction. D-dimer, a hallmark of venous thromboembolism (VTE), is observed at a higher level in the majority of hospitalized COVID-19 patients. The precise molecular mechanism of the disproportionate effect of SARS-CoV-2 infection on the coagulation system is largely undefined. SARS-CoV-2 -induced endotheliopathy and, induction of cytokines and growth factors (GFs) most likely play important roles in platelet activation, coagulopathy, and VTE. Generally, viral infections lead to systemic inflammation and induction of numerous cytokines and GFs and many of them are reported to be associated with increased VTE. Most importantly, platelets play key thromboinflammatory roles linking coagulation to immune mediators in a variety of infections including response to viral infection. Since the pathomechanism of coagulopathy and VTE in COVID-19 is largely undefined, herein we highlight the association of dysregulated inflammatory cytokines and GFs with thrombotic complications and coagulopathy in COVID-19.
Collapse
Affiliation(s)
- Firdos Ahmad
- College of Medicine, University of Sharjah, Sharjah 27272, UAE; Sharjah Institute for Medical Research, University of Sharjah, Sharjah 27272, UAE.
| | - Meganathan Kannan
- Blood and Vascular Biology Research Lab, Department of Life Sciences, Central University of Tamil Nadu, Thiruvarur 610005, India
| | - Abdul W Ansari
- Dermatology Institute, Translational Research Institute, Academic Health Systems, Hamad Medical Corporation, PO Box 3050, Doha, Qatar
| |
Collapse
|
104
|
Sirenko O, Kuryata O, Tykhomyrov A, Yatsenko T. Plasminogen activator inhibitor-1 and circulating ceruloplasmin levels in men with iron-deficiency anemia and heart failure with concomitant prostate cancer and their dynamics after treatment. JOURNAL OF MEDICAL SCIENCES 2022. [DOI: 10.4103/jmedsci.jmedsci_427_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
105
|
Hu MC, Moe OW. Phosphate and Cellular Senescence. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1362:55-72. [PMID: 35288873 PMCID: PMC10513121 DOI: 10.1007/978-3-030-91623-7_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Cellular senescence is one type of permeant arrest of cell growth and one of increasingly recognized contributor to aging and age-associated disease. High phosphate and low Klotho individually and synergistically lead to age-related degeneration in multiple organs. Substantial evidence supports the causality of high phosphate in cellular senescence, and potential contribution to human aging, cancer, cardiovascular, kidney, neurodegenerative, and musculoskeletal diseases. Phosphate can induce cellular senescence both by direct phosphotoxicity, and indirectly through downregulation of Klotho and upregulation of plasminogen activator inhibitor-1. Restriction of dietary phosphate intake and blockage of intestinal absorption of phosphate help suppress cellular senescence. Supplementation of Klotho protein, cellular senescence inhibitor, and removal of senescent cells with senolytic agents are potential novel strategies to attenuate phosphate-induced cellular senescence, retard aging, and ameliorate age-associated, and phosphate-induced disorders.
Collapse
Affiliation(s)
- Ming Chang Hu
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Departments of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
106
|
The role of plasminogen activator inhibitor-1 in gynecological and obstetrical diseases: an update review. J Reprod Immunol 2022; 150:103490. [DOI: 10.1016/j.jri.2022.103490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 01/20/2022] [Accepted: 01/25/2022] [Indexed: 11/21/2022]
|
107
|
Sasidharakurup H, Kumar G, Nair B, Diwakar S. Mathematical Modeling of Severe Acute Respiratory Syndrome Coronavirus 2 Infection Network with Cytokine Storm, Oxidative Stress, Thrombosis, Insulin Resistance, and Nitric Oxide Pathways. OMICS : A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:770-781. [PMID: 34807729 DOI: 10.1089/omi.2021.0155] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a systemic disease affecting not only the lungs but also multiple organ systems. Clinical studies implicate that SARS-CoV-2 infection causes imbalance of cellular homeostasis and immune response that trigger cytokine storm, oxidative stress, thrombosis, and insulin resistance. Mathematical modeling can offer in-depth understanding of the SARS-CoV-2 infection and illuminate how subcellular mechanisms and feedback loops underpin disease progression and multiorgan failure. We report here a mathematical model of SARS-CoV-2 infection pathway network with cytokine storm, oxidative stress, thrombosis, insulin resistance, and nitric oxide (NO) pathways. The biochemical systems theory model shows autocrine loops with positive feedback enabling excessive immune response, cytokines, transcription factors, and interferons, which can imbalance homeostasis of the system. The simulations suggest that changes in immune response led to uncontrolled release of cytokines and chemokines, including interleukin (IL)-1β, IL-6, and tumor necrosis factor α (TNFα), and affect insulin, coagulation, and NO signaling pathways. Increased production of NETs (neutrophil extracellular traps), thrombin, PAI-1 (plasminogen activator inhibitor-1), and other procoagulant factors led to thrombosis. By analyzing complex biochemical reactions, this model forecasts the key intermediates, potential biomarkers, and risk factors at different stages of COVID-19. These insights can be useful for drug discovery and development, as well as precision treatment of multiorgan implications of COVID-19 as seen in systems medicine.
Collapse
Affiliation(s)
- Hemalatha Sasidharakurup
- Amrita Mind Brain Center and Amrita Vishwa Vidyapeetham, Kollam, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
| | - Geetha Kumar
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Tata Institute for Genetics and Society, Kodigehalli, Bengaluru, India
| | - Bipin Nair
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Tata Institute for Genetics and Society, Kodigehalli, Bengaluru, India
| | - Shyam Diwakar
- Amrita Mind Brain Center and Amrita Vishwa Vidyapeetham, Kollam, India
- School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- School of Engineering, Amrita Vishwa Vidyapeetham, Kollam, India
| |
Collapse
|
108
|
Hendley SA, Dimov A, Bhargava A, Snoddy E, Mansour D, Afifi RO, Wool GD, Zha Y, Sammet S, Lu ZF, Ahmed O, Paul JD, Bader KB. Assessment of histological characteristics, imaging markers, and rt-PA susceptibility of ex vivo venous thrombi. Sci Rep 2021; 11:22805. [PMID: 34815441 PMCID: PMC8610976 DOI: 10.1038/s41598-021-02030-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Accepted: 11/08/2021] [Indexed: 12/27/2022] Open
Abstract
Venous thromboembolism is a significant source of morbidity and mortality worldwide. Catheter-directed thrombolytics is the primary treatment used to relieve critical obstructions, though its efficacy varies based on the thrombus composition. Non-responsive portions of the specimen often remain in situ, which prohibits mechanistic investigation of lytic resistance or the development of diagnostic indicators for treatment outcomes. In this study, thrombus samples extracted from venous thromboembolism patients were analyzed ex vivo to determine their histological properties, susceptibility to lytic therapy, and imaging characteristics. A wide range of thrombus morphologies were observed, with a dependence on age and etymology of the specimen. Fibrinolytic inhibitors including PAI-1, alpha 2-antiplasmin, and TAFI were present in samples, which may contribute to the response venous thrombi to catheter-directed thrombolytics. Finally, a weak but significant correlation was observed between the response of the sample to lytic drug and its magnetic microstructure assessed with a quantitative MRI sequence. These findings highlight the myriad of changes in venous thrombi that may promote lytic resistance, and imaging metrics that correlate with treatment outcomes.
Collapse
Affiliation(s)
- Samuel A Hendley
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA
| | - Alexey Dimov
- Department of Radiology, Weill Cornell Medicine, New York, NY, 10021, USA
| | - Aarushi Bhargava
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Erin Snoddy
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Daniel Mansour
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Rana O Afifi
- Department of Cardiothoracic and Vascular Surgery, University of Texas at Houston, Houston, TX, 77030, USA
| | - Geoffrey D Wool
- Department of Pathology, University of Chicago, Chicago, IL, 60637, USA
| | - Yuanyuan Zha
- The Human Immunological Monitoring Facility, University of Chicago, Chicago, IL, 60637, USA
| | - Steffen Sammet
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Zheng Feng Lu
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Osman Ahmed
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA
| | - Jonathan D Paul
- Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Kenneth B Bader
- Committee on Medical Physics, University of Chicago, Chicago, IL, 60637, USA.
- Department of Radiology, University of Chicago, Chicago, IL, 60637, USA.
| |
Collapse
|
109
|
Valsesia A, Egli L, Bosco N, Magkos F, Kong SC, Sun L, Goh HJ, Weiting H, Arigoni F, Leow MKS, Yeo KK, Actis-Goretta L. Clinical- and omics-based models of subclinical atherosclerosis in healthy Chinese adults: a cross-sectional exploratory study. Am J Clin Nutr 2021; 114:1752-1762. [PMID: 34476468 DOI: 10.1093/ajcn/nqab269] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 07/23/2021] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Classical risk factors, such as fasting cholesterol, blood pressure (BP), and diabetes status are used today to predict the risk of developing cardiovascular disease (CVD). However, accurate prediction remains limited, particularly in low-risk groups such as women and younger individuals. Growing evidence suggests that biomarker concentrations following consumption of a meal challenge are better and earlier predictors of disease development than biomarker concentrations. OBJECTIVE To test the hypothesis that postprandial responses of circulating biomarkers differ between healthy subjects with and without subclinical atherosclerosis (SA) in an Asian population at low risk of coronary artery disease (CAD). METHODS One hundred healthy Chinese subjects (46 women, 54 men) completed the study. Subjects consumed a mixed-meal test and 164 blood biomarkers were analyzed over 6 h by using a combination of chemical and NMR techniques. Models were trained using different methodologies (including logistic regression, elastic net, random forest, sparse partial least square) on a random 75% subset of the data, and their performance was evaluated on the remaining 25%. RESULTS We found that models based on baseline clinical parameters or fasting biomarkers could not reliably predict SA. By contrast, an omics model based on magnitude and timing of postprandial biomarkers achieved high performance [receiving operating characteristic (ROC) AUC: 91%; 95% CI: 77, 100). Investigation of key features of this model enabled derivation of a considerably simpler model, solely based on postprandial BP and age, with excellent performance (AUC: 91%; 95% CI: 78, 100). CONCLUSION We report a novel model to detect SA based on postprandial BP and age in a population of Asian subjects at low risk of CAD. The use of this model in large-scale CVD prevention programs should be explored. This trial was registered at ClinicalTrials.gov as NCT03531879.
Collapse
Affiliation(s)
- Armand Valsesia
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Leonie Egli
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Institute of Health Sciences, Nestlé Research, Lausanne, Switzerland
- Nestlé Research Singapore Hub, Singapore
| | | | | | - Lijuan Sun
- Singapore Institute for Clinical Sciences, Singapore
| | - Hui Jen Goh
- Singapore Institute for Clinical Sciences, Singapore
| | | | | | - Melvin Khee-Shing Leow
- Singapore Institute for Clinical Sciences, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
| | - Khung Keong Yeo
- National Heart Center Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
| | | |
Collapse
|
110
|
Serum anti-SERPINE1 antibody as a potential biomarker of acute cerebral infarction. Sci Rep 2021; 11:21772. [PMID: 34741085 PMCID: PMC8571331 DOI: 10.1038/s41598-021-01176-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 10/19/2021] [Indexed: 11/11/2022] Open
Abstract
The presence of disease-specific antigens and autoantibodies in the sera of patients with atherosclerosis-related diseases has been widely reported and is considered to result from inflammation of the arterial wall and the involvement of immune factors. The aim of this study was to identify a novel antibody in patients with ischemic stroke by serological identification of antigens using recombinant cDNA expression cloning from patients who had a transient ischemic attack (TIA). We identified the serpin peptidase inhibitor, clade E member 1 (SERPINE1), as a candidate antigen. The serum anti-SERPINE1 antibody levels quantified using amplified luminescent proximity homogeneous assay-linked immunosorbent assay were significantly higher in patients with ischemic stroke, including those with acute cerebral infarction (aCI), TIA, and chronic cerebral infarction, than in healthy donors. The antibody levels were strongly associated with old age, female sex, and presence of hypertension, diabetes mellitus, and cardiovascular disease. Age and intima-media thickness of the carotid artery were positively correlated with antibody levels, which suggests that SERPINE1 may reflect the progression of atherosclerosis. In a multivariate analysis, SERPINE1 antibody level was an independent predictor of aCI. Thus, the serum levels of anti-SERPINE1 antibody could potentially serve as a biomarker of atherothrombotic infarction.
Collapse
|
111
|
Bidwell JT, Hostinar CE, Higgins MK, Abshire MA, Cothran F, Butts B, Miller AH, Corwin E, Dunbar SB. Caregiver subjective and physiological markers of stress and patient heart failure severity in family care dyads. Psychoneuroendocrinology 2021; 133:105399. [PMID: 34482256 PMCID: PMC8530937 DOI: 10.1016/j.psyneuen.2021.105399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 08/23/2021] [Indexed: 12/20/2022]
Abstract
Greater family caregiver exposure to uncontrolled patient symptoms is predictive of greater caregiver psychological and physiological stress in dementia and other chronic illnesses, but these phenomena have not been well-studied in heart failure (HF) - a disease with high symptom burden. The purpose of this study was to test the hypothesis that worse patient functional status (as reflected by increasing HF symptoms) would be associated with elevated psychological and physiological stress for the caregiver. This was a secondary analysis of data from 125 HF caregivers in the Caregiver Opportunities for Optimizing Lifestyle (COOL) study. Psychological stress was measured on four dimensions: care-related strain/burden (Oberst Caregiving Burden Scale), depression (Center for Epidemiological Studies Depression Scale), anxiety (State-Trait Anxiety Index), and general stress (Perceived Stress Scale). Physiological stress was measured by markers of HPA axis function (elevated cortisol awakening response [CAR]), endothelial dysfunction (increased PAI-1), and inflammation (increased IL-6, hsCRP). HF patient functional status was quantified by caregiver assessment of New York Heart Association (NYHA) Class. Generalized linear models were used to test associations between patient NYHA Class and stress (one model per indicator). NYHA Class (ordinal) was backwards difference coded in each model to examine caregiver stress in relation to increasing levels of HF severity. Caregivers were mostly female and in their mid-fifties, with a slight majority of the sample being African American and the patient's spouse. Overall, patient functional status was associated with greater caregiver psychological and physiological stress. In terms of psychological stress, higher NYHA Class was significantly associated with greater caregiver anxiety and general stress, but not with caregiver burden or depression. In terms of physiological stress, higher NYHA Class was associated with elevated markers in all models (elevated CAR and higher IL-6, hsCRP, and PAI-1). Across models, most associations between NYHA Class and stress were present at relatively early stages of functional limitation (i.e. Class II), while others emerged when functional limitations became more severe. To inform timing and mechanisms for much-needed caregiver interventions, research is needed to determine which aspects of HF symptomatology are most stressful for caregivers across the HF trajectory.
Collapse
Affiliation(s)
- Julie T. Bidwell
- Family Caregiving Institute at the Betty Irene Moore School of Nursing; University of California, Davis; 2570 48th Street, Sacramento, CA 95817, United States of America
| | - Camelia E. Hostinar
- Department of Psychology; University of California, Davis; 1 Shields Avenue, Davis, CA 95616, United States of America
| | - Melinda K. Higgins
- Nell Hodgson Woodruff School of Nursing; Emory University; 1520 Clifton Road, Atlanta, GA 30322, United States of America
| | - Martha A. Abshire
- School of Nursing; Johns Hopkins University; 525 N Wolfe Street, Baltimore, MD 21205, United States of America
| | - Fawn Cothran
- Family Caregiving Institute at the Betty Irene Moore School of Nursing, University of California, Davis, 2570 48th Street, Sacramento, CA 95817, United States.
| | - Brittany Butts
- Nell Hodgson Woodruff School of Nursing, Emory University, 1520 Clifton Road, Atlanta, GA 30322, United States.
| | - Andrew H. Miller
- Department of Psychiatry and Behavioral Sciences, School of Medicine, Emory University, 12 Executive Park Dr, NE. 2nd Floor, Atlanta, GA 30329
| | - Elizabeth Corwin
- School of Nursing, Columbia University, 560 W. 168th St, Room 600, New York, NY 10032, United States.
| | - Sandra B. Dunbar
- Nell Hodgson Woodruff School of Nursing; Emory University; 1520 Clifton Road, Atlanta, GA 30322, United States of America
| |
Collapse
|
112
|
González F, Considine RV, Abdelhadi OA, Xue J, Acton AJ. Saturated fat ingestion stimulates proatherogenic inflammation in polycystic ovary syndrome. Am J Physiol Endocrinol Metab 2021; 321:E689-E701. [PMID: 34632798 PMCID: PMC8782660 DOI: 10.1152/ajpendo.00213.2021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Inflammation and dyslipidemia are often present in polycystic ovary syndrome (PCOS). We determined the effect of saturated fat ingestion on circulating heat shock protein-70 (HSP-70) and mononuclear cell (MNC) toll-like receptor-2 (TLR2) gene expression, activator protein-1 (AP-1) activation, and matrix matalloproteinase-2 (MMP-2) protein in women with PCOS. Twenty reproductive-age women with PCOS (10 lean, 10 with obesity) and 20 ovulatory controls (10 lean, 10 with obesity) participated in the study. HSP-70 was measured in serum and TLR2 mRNA and protein, AP-1 activation, and MMP-2 protein were quantified in MNC from blood drawn while fasting and 2, 3, and 5 h after saturated fat ingestion. Insulin sensitivity was derived from an oral glucose tolerance test (ISOGTT). Androgen secretion was assessed from blood drawn while fasting and 24, 48, and 72 h after human chorionic gonadotropin (HCG) administration. In response to saturated fat ingestion, serum HSP-70, TLR2 gene expression, activated AP-1, and MMP-2 protein were greater in lean women with PCOS compared with lean controls and in women with PCOS and obesity compared with controls with obesity. Both PCOS groups exhibited lower ISOGTT and greater HCG-stimulated androgen secretion compared with control subjects of their respective weight classes. Lipid-stimulated proatherogenic inflammation marker responses were negatively correlated with ISOGTT and positively correlated with abdominal adiposity and HCG-stimulated androgen secretion. In PCOS, saturated fat ingestion stimulates proatherogenic inflammation independent of obesity. This effect is greater when PCOS is combined with obesity compared with obesity alone. Abdominal adiposity and hyperandrogenism may perpetuate proatherogenic inflammation.NEW & NOTEWORTHY This paper demonstrates that in polycystic ovary syndrome (PCOS), ingestion of saturated fat triggers a molecular pathway of inflammation known to drive atherogenesis. This effect is independent of obesity as it occurs in lean women with PCOS and not in lean ovulatory control subjects. Furthermore, the combined effects of PCOS and obesity are greater compared with obesity alone.
Collapse
Affiliation(s)
- Frank González
- Department of Obstetrics and Gynecology, University of Illinois Chicago College of Medicine, Chicago, Illinois
| | - Robert V Considine
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| | - Ola A Abdelhadi
- Department of Obstetrics and Gynecology, Indiana University School of Medicine, Indianapolis, Indiana
| | - Jiaping Xue
- Department of Obstetrics and Gynecology, University of Illinois Chicago College of Medicine, Chicago, Illinois
| | - Anthony J Acton
- Department of Medicine, Indiana University School of Medicine, Indianapolis, Indiana
| |
Collapse
|
113
|
Sfera A, Osorio C, Rahman L, Zapata-Martín del Campo CM, Maldonado JC, Jafri N, Cummings MA, Maurer S, Kozlakidis Z. PTSD as an Endothelial Disease: Insights From COVID-19. Front Cell Neurosci 2021; 15:770387. [PMID: 34776871 PMCID: PMC8586713 DOI: 10.3389/fncel.2021.770387] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 10/11/2021] [Indexed: 12/15/2022] Open
Abstract
SARS-CoV-2 virus, the etiologic agent of COVID-19, has affected almost every aspect of human life, precipitating stress-related pathology in vulnerable individuals. As the prevalence rate of posttraumatic stress disorder in pandemic survivors exceeds that of the general and special populations, the virus may predispose to this disorder by directly interfering with the stress-processing pathways. The SARS-CoV-2 interactome has identified several antigens that may disrupt the blood-brain-barrier by inducing premature senescence in many cell types, including the cerebral endothelial cells. This enables the stress molecules, including angiotensin II, endothelin-1 and plasminogen activator inhibitor 1, to aberrantly activate the amygdala, hippocampus, and medial prefrontal cortex, increasing the vulnerability to stress related disorders. This is supported by observing the beneficial effects of angiotensin receptor blockers and angiotensin converting enzyme inhibitors in both posttraumatic stress disorder and SARS-CoV-2 critical illness. In this narrative review, we take a closer look at the virus-host dialog and its impact on the renin-angiotensin system, mitochondrial fitness, and brain-derived neurotrophic factor. We discuss the role of furin cleaving site, the fibrinolytic system, and Sigma-1 receptor in the pathogenesis of psychological trauma. In other words, learning from the virus, clarify the molecular underpinnings of stress related disorders, and design better therapies for these conditions. In this context, we emphasize new potential treatments, including furin and bromodomains inhibitors.
Collapse
Affiliation(s)
- Adonis Sfera
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
- Patton State Hospital, San Bernardino, CA, United States
| | - Carolina Osorio
- Department of Psychiatry, Loma Linda University, Loma Linda, CA, United States
| | - Leah Rahman
- Patton State Hospital, San Bernardino, CA, United States
| | | | - Jose Campo Maldonado
- Department of Medicine, The University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Nyla Jafri
- Patton State Hospital, San Bernardino, CA, United States
| | | | - Steve Maurer
- Patton State Hospital, San Bernardino, CA, United States
| | - Zisis Kozlakidis
- International Agency For Research On Cancer (IARC), Lyon, France
| |
Collapse
|
114
|
Martins SR, Toledo SLDO, da Silva AJ, Mendes FS, de Oliveira MM, Ferreira LGR, Dusse LMS, Carvalho MDG, Rios DRA, Alpoim PN, Pinheiro MDB. Endothelial dysfunction biomarkers in sickle cell disease: is there a role for ADMA and PAI-1? Ann Hematol 2021; 101:273-280. [PMID: 34665295 DOI: 10.1007/s00277-021-04695-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/10/2021] [Indexed: 01/26/2023]
Abstract
Within the spectrum of sickle cell disease (SCD) are sickle cell anemia (SCA), presence of hemoglobin SS (HbSS), hemoglobin SC disease (HbSC), and sickle cell β-thalassemia (Sβ-thal). Asymmetric dimethylarginine (ADMA) competitively inhibits the binding of arginine to NOS, reducing NO production. In patients with HbSS, increased levels of ADMA have been reported, as well as changes in many hemostatic biomarkers, including the plasminogen activator inhibitor type 1 (PAI-1). We hypothesized that high levels of ADMA and PAI-1 may be associated with more severe SCD. Thus, ADMA and PAI-1 levels were determined in 78 individuals including 38 adult patients with SCD and 40 control subjects. Higher levels of ADMA were shown in HbSS and Sβ-thal patients compared to controls. Concerning PAI-1, all patients showed high levels of PAI-1 compared to controls. As a role of NO in the pathogenesis of SCD has already been established, we concluded that high levels of ADMA should compromise, at least in part, NO synthesis, resulting in endothelial dysfunction. Elevated plasma levels of PAI-1 in all patients may indicate not only endothelial dysfunction but also a hypofibrinolytic state favoring thrombotic complications. Finally, high levels of ADMA and PAI-1 may be associated with more severe SCD.
Collapse
Affiliation(s)
- Suellen Rodrigues Martins
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270901, Brazil
| | - Sílvia Letícia de Oliveira Toledo
- Federal University of São João del-Rei (UFSJ), Campus Centro-Oeste Dona Lindu, Sebastião Gonçalves Coelho Street, 400, Building: D, Room: 308.1, ChanadourDivinópolis, MG, 35501-296, Brazil
| | - Aislander Junio da Silva
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270901, Brazil
| | - Fernanda Santos Mendes
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270901, Brazil
| | - Marina Mendes de Oliveira
- Federal University of São João del-Rei (UFSJ), Campus Centro-Oeste Dona Lindu, Sebastião Gonçalves Coelho Street, 400, Building: D, Room: 308.1, ChanadourDivinópolis, MG, 35501-296, Brazil.,Fundação Centro de Hematologia E Hemoterapia Do Estado de Minas Gerais, (Hemominas), Divinópolis, MG, Brazil
| | - Leticia Gonçalves Resende Ferreira
- Federal University of São João del-Rei (UFSJ), Campus Centro-Oeste Dona Lindu, Sebastião Gonçalves Coelho Street, 400, Building: D, Room: 308.1, ChanadourDivinópolis, MG, 35501-296, Brazil
| | - Luci Maria Sant'Ana Dusse
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270901, Brazil
| | - Maria das Graças Carvalho
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270901, Brazil.,Federal University of São João del-Rei (UFSJ), Campus Centro-Oeste Dona Lindu, Sebastião Gonçalves Coelho Street, 400, Building: D, Room: 308.1, ChanadourDivinópolis, MG, 35501-296, Brazil
| | - Danyelle Romana Alves Rios
- Federal University of São João del-Rei (UFSJ), Campus Centro-Oeste Dona Lindu, Sebastião Gonçalves Coelho Street, 400, Building: D, Room: 308.1, ChanadourDivinópolis, MG, 35501-296, Brazil
| | - Patrícia Nessralla Alpoim
- Department of Clinical and Toxicological Analysis, Faculty of Pharmacy, Federal University of Minas Gerais, Avenida Antonio Carlos, 6627, Pampulha, Belo Horizonte, Minas Gerais, 31270901, Brazil.
| | - Melina de Barros Pinheiro
- Federal University of São João del-Rei (UFSJ), Campus Centro-Oeste Dona Lindu, Sebastião Gonçalves Coelho Street, 400, Building: D, Room: 308.1, ChanadourDivinópolis, MG, 35501-296, Brazil.
| |
Collapse
|
115
|
Wong YK, Tse HF. Circulating Biomarkers for Cardiovascular Disease Risk Prediction in Patients With Cardiovascular Disease. Front Cardiovasc Med 2021; 8:713191. [PMID: 34660715 PMCID: PMC8517145 DOI: 10.3389/fcvm.2021.713191] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Accepted: 09/08/2021] [Indexed: 12/23/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of death globally. Risk assessment is crucial for identifying at-risk individuals who require immediate attention as well as to guide the intensity of medical therapy to reduce subsequent risk of CVD. In the past decade, many risk prediction models have been proposed to estimate the risk of developing CVD. However, in patients with a history of CVD, the current models that based on traditional risk factors provide limited power in predicting recurrent cardiovascular events. Several biomarkers from different pathophysiological pathways have been identified to predict cardiovascular events, and the incorporation of biomarkers into risk assessment may contribute to enhance risk stratification in secondary prevention. This review focuses on biomarkers related to cardiovascular and metabolic diseases, including B-type natriuretic peptide, high-sensitivity cardiac troponin I, adiponectin, adipocyte fatty acid-binding protein, heart-type fatty acid-binding protein, lipocalin-2, fibroblast growth factor 19 and 21, retinol-binding protein 4, plasminogen activator inhibitor-1, 25-hydroxyvitamin D, and proprotein convertase subtilisin/kexin type 9, and discusses the potential utility of these biomarkers in cardiovascular risk prediction among patients with CVD. Many of these biomarkers have shown promise in improving risk prediction of CVD. Further research is needed to assess the validity of biomarker and whether the strategy for incorporating biomarker into clinical practice may help to optimize decision-making and therapeutic management.
Collapse
Affiliation(s)
- Yuen-Kwun Wong
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China
| | - Hung-Fat Tse
- Department of Medicine, The University of Hong Kong, Queen Mary Hospital, Hong Kong, China.,Department of Medicine, Shenzhen Hong Kong University Hospital, Shenzhen, China.,Hong Kong-Guangdong Joint Laboratory on Stem Cell and Regenerative Medicine, The University of Hong Kong, Hong Kong, China.,Shenzhen Institutes of Research and Innovation, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
116
|
Ghosh AK, Soberanes S, Lux E, Shang M, Aillon RP, Eren M, Budinger GRS, Miyata T, Vaughan DE. Pharmacological inhibition of PAI-1 alleviates cardiopulmonary pathologies induced by exposure to air pollutants PM 2.5. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 287:117283. [PMID: 34426376 PMCID: PMC8434953 DOI: 10.1016/j.envpol.2021.117283] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/16/2021] [Accepted: 04/28/2021] [Indexed: 05/09/2023]
Abstract
Numerous studies have established that acute or chronic exposure to environmental pollutants like particulate matter (PM) leads to the development of accelerated aging related pathologies including pulmonary and cardiovascular diseases, and thus air pollution is one of the major global threats to human health. Air pollutant particulate matter 2.5 (PM2.5)-induced cellular dysfunction impairs tissue homeostasis and causes vascular and cardiopulmonary damage. To test a hypothesis that elevated plasminogen activator inhibitor-1 (PAI-1) levels play a pivotal role in air pollutant-induced cardiopulmonary pathologies, we examined the efficacy of a drug-like novel inhibitor of PAI-1, TM5614, in treating PM2.5-induced vascular and cardiopulmonary pathologies. Results from biochemical, histological, and immunohistochemical studies revealed that PM2.5 increases the circulating levels of PAI-1 and thrombin and that TM5614 treatment completely abrogates these effects in plasma. PM2.5 significantly augments the levels of pro-inflammatory cytokine interleukin-6 (IL-6) in bronchoalveolar lavage fluid (BALF), and this also can be reversed by TM5614, indicating its efficacy in amelioration of PM2.5-induced increases in inflammatory and pro-thrombotic factors. TM5614 reduces PM2.5-induced increased levels of inflammatory markers cluster of differentiation 107 b (Mac3) and phospho-signal transducer and activator of transcription-3 (pSTAT3), adhesion molecule vascular cell adhesion molecule 1 (VCAM1), and apoptotic marker cleaved caspase 3. Longer exposure to PM2.5 induces pulmonary and cardiac thrombosis, but TM5614 significantly ameliorates PM2.5-induced vascular thrombosis. TM5614 also reduces PM2.5-induced increased blood pressure and heart weight. In vitro cell culture studies revealed that PM2.5 induces the levels of PAI-1, type I collagen, fibronectin (Millipore), and sterol regulatory element binding protein-1 and 2 (SREBP-1 and SREBP-2), transcription factors that mediate profibrogenic signaling, in cardiac fibroblasts. TM5614 abrogated that stimulation, indicating that it may block PM2.5-induced PAI-1 and profibrogenic signaling through suppression of SREBP-1 and 2. Furthermore, TM5614 blocked PM2.5-mediated suppression of nuclear factor erythroid related factor 2 (Nrf2), a major antioxidant regulator, in cardiac fibroblasts. Pharmacological inhibition of PAI-1 with TM5614 is a promising therapeutic approach to control air pollutant PM2.5-induced cardiopulmonary and vascular pathologies.
Collapse
Affiliation(s)
- Asish K Ghosh
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Saul Soberanes
- Pulmonary and Critical Care Division, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Elizabeth Lux
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Meng Shang
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Raul Piseaux Aillon
- Pulmonary and Critical Care Division, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mesut Eren
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - G R Scott Budinger
- Pulmonary and Critical Care Division, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University, Miyagi, Japan
| | - Douglas E Vaughan
- Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
117
|
Karaman K, Celik A, Aytac S, Bakar-Ates F. Increased endoglin levels correlated with angiogenesis-associated angiopoietin-2 in haemophilia patients. Haemophilia 2021; 27:e747-e753. [PMID: 34614537 DOI: 10.1111/hae.14430] [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: 04/14/2021] [Revised: 09/21/2021] [Accepted: 09/23/2021] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Haemophilia is a bleeding disorder that occurs due to the deficiency of coagulation factors, and the angiogenesis process is an important process underlying the pathophysiology of haemophilic arthropathy. The role of the new adipocytokine endoglin (ENG) in patients with haemophilia is not yet known. AIM The aim of this study is to evaluate the association between ENG protein and angiogenesis-related cytokines in patients with haemophilia for the first time. METHODS Plasma protein levels and mRNA expressions of ENG and various angiogenesis-associated cytokines were compared in blood samples collected from 28 patients with haemophilia A or B and 29 healthy volunteers. The relationship between the cytokines and ENG were determined by correlation analysis. RESULTS Plasma ENG levels and angiogenic markers were found to be significantly higher in patients with haemophilia compared to controls. Real-time PCR studies showed that mRNA expressions of ENG, vascular endothelial growth factor A, hypoxia-inducible factor A, and prostaglandin E2 increased in patients with haemophilia. Correlation analysis showed a significant positive correlation between ENG and angiopoietin-2 levels in the haemophilia group. Besides, a significant decrease in annexin-V binding to platelets in haemophilia patients compared to control was found to be related to the bleeding profiles in the patients. CONCLUSIONS This study determined that ENG protein may be involved in the formation of angiogenesis in haemophilia patients and its effects may be related to angiogenetic marker angiopoietin-2 in this process. Our findings contribute to the literature during the determination of target proteins in haemophilia treatment.
Collapse
Affiliation(s)
- Kubra Karaman
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| | - Aybuke Celik
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| | - Selin Aytac
- Faculty of Medicine, Department of Paediatrics, Division of Paediatric Haematology, Hacettepe University, Ankara, Turkey
| | - Filiz Bakar-Ates
- Ankara University, Faculty of Pharmacy, Department of Biochemistry, Ankara, Turkey
| |
Collapse
|
118
|
Chen J, Zhai C, Wang Z, Li R, Wu W, Hou K, Alzogool M, Wang Y, Cong H. The susceptibility of SERPINE1 rs1799889 SNP in diabetic vascular complications: a meta-analysis of fifty-one case-control studies. BMC Endocr Disord 2021; 21:195. [PMID: 34592988 PMCID: PMC8482645 DOI: 10.1186/s12902-021-00837-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 08/10/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND The serine protease inhibitor-1 (SERPINE1) rs1799889 single nucleotide polymorphism (SNP) has been constantly associated with diabetes mellitus (DM) and its vascular complications. The aim of this meta-analysis was to evaluate this association with combined evidences. METHODS The systematic search was performed for studies published up to March 2021 which assess the associations between SERPINE1 rs1799889 SNP and the risks of DM, diabetic retinopathy (DR), diabetic cardiovascular disease (CVD) and diabetic nephropathy (DN). Only case-control studies were identified, and the linkage between SERPINE1 rs1799889 polymorphism and diabetic vascular risks were evaluated using genetic models. RESULTS 51 comparisons were enrolled. The results revealed a significant association with diabetes risk in overall population (allelic: OR = 1.34, 95 % CI = 1.14-1.57, homozygous: OR = 1.66, 95 % CI = 1.23-2.14, heterozygous: OR = 1.35, 95 % CI = 1.08-1.69, dominant: OR = 1.49, 95 % CI = 1.18-1.88, recessive: OR = 1.30, 95 % CI = 1.06-1.59) as well as in Asian descents (allelic: OR = 1.45, 95 % CI = 1.16-1.82, homozygous: OR = 1.88, 95 % CI = 1.29-2.75, heterozygous: OR = 1.47, 95 % CI = 1.08-2.00, dominant: OR = 1.64, 95 % CI = 1.21-2.24, recessive: OR = 1.46, 95 % CI = 1.09-1.96). A significant association was observed with DR risk (homozygous: OR = 1.25, 95 % CI = 1.01-1.56, recessive: OR = 1.20, 95 % CI = 1.01-1.43) for overall population, as for the European subgroup (homozygous: OR = 1.32, 95 % CI = 1.02-1.72, recessive: OR = 1.38, 95 % CI = 1.11-1.71). A significant association were shown with DN risk for overall population (allelic: OR = 1.48, 95 % CI = 1.15-1.90, homozygous: OR = 1.92, 95 % CI = 1.26-2.95, dominant: OR = 1.41, 95 % CI = 1.01-1.97, recessive: OR = 1.78, 95 % CI = 1.27-2.51) and for Asian subgroup (allelic: OR = 1.70, 95 % CI = 1.17-2.47, homozygous: OR = 2.46, 95 % CI = 1.30-4.66, recessive: OR = 2.24, 95 % CI = 1.40-3.59) after ethnicity stratification. No obvious association was implied with overall diabetic CVD risk in any genetic models, or after ethnicity stratification. CONCLUSIONS SERPINE1 rs1799889 4G polymorphism may outstand for serving as a genetic synergistic factor in overall DM and DN populations, positively for individuals with Asian descent. The association of SERPINE1 rs1799889 SNP and DR or diabetic CVD risks was not revealed.
Collapse
Affiliation(s)
- JingYi Chen
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - ChuanNan Zhai
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| | - ZhiQian Wang
- Department of Optometry, Shenyang Eye Institute, The 4th People’s Hospital of Shenyang, No 20. Huanghe South Avenue, Huanggu District, 110031 Shenyang, Liaoning China
| | - Rui Li
- Tianjin GongAn Hospital, Nanjing Road No. 78, Heping District, 300042 Tianjin, China
| | - WenJing Wu
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - Kai Hou
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| | - Mohammad Alzogool
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - Yan Wang
- School of Medicine, NanKai University, Weijin Road No. 94, Nankai District, 300071 Tianjin, China
- Tianjin Key Lab of Ophthalmology and Visual Science, Tianjin Eye Institute, Tianjin Eye Hospital, Gansu Road No. 4, Heping District, 300020 Tianjin, China
| | - HongLiang Cong
- Department of Cardiology, Tianjin Chest Hospital, Taierzhuang south Road No. 291, Jinnan District, 300350 Tianjin, China
| |
Collapse
|
119
|
SWATH-MS for prospective identification of protein blood biomarkers of rtPA-associated intracranial hemorrhage in acute ischemic stroke: a pilot study. Sci Rep 2021; 11:18765. [PMID: 34548538 PMCID: PMC8455557 DOI: 10.1038/s41598-021-97710-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 08/27/2021] [Indexed: 11/08/2022] Open
Abstract
Intravenous recombinant tissue plasminogen activator (rtPA) is, besides mechanical thrombectomy, the highest class evidence based reperfusion treatment of acute ischemic stroke (AIS). The biggest concern of the therapy is symptomatic intracranial hemorrhage (sICH), which occurs in 3-7% of all treated patients, and is associated with worse functional outcome. Finding a method of the powerful identification of patients at highest risk of sICH, in order to increase the percentage of stroke patients safely treated with rtPA, is one of the most important challenges in stroke research. To address this problem, we designed a complex project to identify blood, neuroimaging, and clinical biomarkers combined for prospective assessment of the risk of rtPA-associated ICH. In this paper we present results of blood proteomic and peptide analysis of pilot 41 AIS patients before rtPA administration (the test ICH group, n = 9 or the controls, without ICH, n = 32). We demonstrated that pre-treatment blood profiles of 15 proteins differ depending on whether the patients develop rtPA-associated ICH or not. SWATH-MS quantification of serum or plasma proteins might allow for robust selection of blood biomarkers to increase the prospective assessment of rtPA-associated ICH over that based solely on clinical and neuroimaging characteristics.
Collapse
|
120
|
Wong A, Chen SQ, Halvorson BD, Frisbee JC. Microvessel Density: Integrating Sex-Based Differences and Elevated Cardiovascular Risks in Metabolic Syndrome. J Vasc Res 2021; 59:1-15. [PMID: 34535606 DOI: 10.1159/000518787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/28/2021] [Indexed: 11/19/2022] Open
Abstract
Metabolic syndrome (MetS) is a complex pathological state consisting of metabolic risk factors such as hypertension, insulin resistance, and obesity. The interconnectivity of cellular pathways within various biological systems suggests that each individual component of MetS may share common pathological sources. Additionally, MetS is closely associated with vasculopathy, including a reduction in microvessel density (MVD) (rarefaction) and elevated risk for various cardiovascular diseases. Microvascular impairments may contribute to perfusion-demand mismatch, where local metabolic needs are insufficiently met due to the lack of nutrient and oxygen supply, thus creating pathological positive-feedback loops and furthering the progression of disease. Sexual dimorphism is evident in these underlying cellular mechanisms, which places males and females at different levels of risk for cardiovascular disease and acute ischemic events. Estrogen exhibits protective effects on the endothelium of pre-menopausal women, while androgens may be antagonistic to cardiovascular health. This review examines MetS and its influences on MVD, as well as sex differences relating to the components of MetS and cardiovascular risk profiles. Finally, translational relevance and interventions are discussed in the context of these sex-based differences.
Collapse
Affiliation(s)
- Angelina Wong
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Shu Qing Chen
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Brayden D Halvorson
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Jefferson C Frisbee
- Department of Medical Biophysics, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| |
Collapse
|
121
|
Yang R, Wu GWY, Verhoeven JE, Gautam A, Reus VI, Kang JI, Flory JD, Abu-Amara D, Hood L, Doyle FJ, Yehuda R, Marmar CR, Jett M, Hammamieh R, Mellon SH, Wolkowitz OM. A DNA methylation clock associated with age-related illnesses and mortality is accelerated in men with combat PTSD. Mol Psychiatry 2021; 26:4999-5009. [PMID: 32382136 DOI: 10.1038/s41380-020-0755-z] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/20/2020] [Accepted: 04/23/2020] [Indexed: 02/02/2023]
Abstract
DNA methylation patterns at specific cytosine-phosphate-guanine (CpG) sites predictably change with age and can be used to derive "epigenetic age", an indicator of biological age, as opposed to merely chronological age. A relatively new estimator, called "DNAm GrimAge", is notable for its superior predictive ability in older populations regarding numerous age-related metrics like time-to-death, time-to-coronary heart disease, and time-to-cancer. PTSD is associated with premature mortality and frequently has comorbid physical illnesses suggestive of accelerated biological aging. This is the first study to assess DNAm GrimAge in PTSD patients. We investigated the acceleration of GrimAge relative to chronological age, denoted "AgeAccelGrim" in combat trauma-exposed male veterans with and without PTSD using cross-sectional and longitudinal data from two independent well-characterized veteran cohorts. In both cohorts, AgeAccelGrim was significantly higher in the PTSD group compared to the control group (N = 162, 1.26 vs -0.57, p = 0.001 and N = 53, 0.93 vs -1.60 Years, p = 0.008), suggesting accelerated biological aging in both cohorts with PTSD. In 3-year follow-up study of individuals initially diagnosed with PTSD (N = 26), changes in PTSD symptom severity were correlated with AgeAccelGrim changes (r = 0.39, p = 0.049). In addition, the loss of CD28 cell surface markers on CD8 + T cells, an indicator of T-cell senescence/exhaustion that is associated with biological aging, was positively correlated with AgeAccelGrim, suggesting an immunological contribution to the accelerated biological aging. Overall, our findings delineate cellular correlates of biological aging in combat-related PTSD, which may help explain the increased medical morbidity and mortality seen in this disease.
Collapse
Affiliation(s)
- Ruoting Yang
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, USA.
| | - Gwyneth W Y Wu
- Weill Institute for Neurosciences and Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Josine E Verhoeven
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Psychiatry, Amsterdam Public Health Research Institute, Amsterdam, The Netherlands
| | - Aarti Gautam
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Victor I Reus
- Weill Institute for Neurosciences and Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| | - Jee In Kang
- Institute of Behavioral Science in Medicine & Department of Psychiatry, Yonsei University College of Medicine, Seoul, South Korea
| | - Janine D Flory
- James J Peters VA Medical Center, Bronx, NY, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Duna Abu-Amara
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | | | - Leroy Hood
- Institute for Systems Biology, Seattle, WA, USA
| | - Francis J Doyle
- John A. Paulson School of Engineering & Applied Sciences, Harvard University, Cambridge, MA, USA
| | - Rachel Yehuda
- James J Peters VA Medical Center, Bronx, NY, USA.,Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles R Marmar
- Department of Psychiatry, New York University School of Medicine, New York, NY, USA
| | - Marti Jett
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Rasha Hammamieh
- Medical Readiness Systems Biology, Walter Reed Army Institute of Research, Silver Spring, MD, USA
| | - Synthia H Mellon
- Department of OB-GYN and Reproductive Sciences, UCSF School of Medicine, San Francisco, CA, USA
| | - Owen M Wolkowitz
- Weill Institute for Neurosciences and Department of Psychiatry, University of California San Francisco (UCSF) School of Medicine, San Francisco, CA, USA
| |
Collapse
|
122
|
Bahrami R, Dastgheib SA, Mirjalili H, Setayesh S, Shaker SH, Mirjalili SR, Noorishadkam M, Neamatzadeh H. Association of SERPINE1 rs1799889 polymorphism with arterial ischemic stroke in children: a systematic review and meta-analysis. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2021; 40:1018-1035. [PMID: 34429017 DOI: 10.1080/15257770.2021.1966798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Inherited thrombophilias are well-established predisposing factors for venous thromboembolism, but their role in arterial ischemic stroke (AIS) in children, remains unclear. The association between SERPINE1 rs1799889 polymorphism and AIS in children was evaluated by several studies, whereas the results were conflicting. Thus, we performed this meta-analysis to combine and analyze the available studies in order to provide a more accurate result on the association. PubMed, Scopus, EMBASE, SciELO, MedRxiv, China Biology Medicine Disk, DeepDyve, CNKI, and Web of Science were used to identify all relevant articles published up to 30 November 2020, without any restrictions on ethnicity. Summary odds ratios (ORs) with 95% confidence intervals (CIs) were used to determine the strength of the associations. A total of eight case-control studies with 600 cases and 2,156 controls were selected. No significant association between SERPINE1 rs1799889 polymorphism and AIS in children susceptibility was noted. In the stratified analyses by ethnicity, source of controls, genotyping methods, and age groups, there was still no significant association between SERPINE1 rs1799889 polymorphism and AIS risk in children. This study suggested that SERPINE1 rs1799889 polymorphism might be not related to etiology of AIS in children. Moreover, well-designed, large-scale and multicenter clinical studies are required to improve and validate these results.Supplemental data for this article is available online at https://doi.org/10.1080/15257770.2021.1966798 .
Collapse
Affiliation(s)
- Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Mirjalili
- Department of Emergency Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Sepideh Setayesh
- School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Hossein Shaker
- Department of Emergency Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyed Reza Mirjalili
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Mahmood Noorishadkam
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Department of Emergency Medicine, Iran University of Medical Sciences, Tehran, Iran.,Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
123
|
Zhang P, Wang W, Mao M, Gao R, Shi W, Li D, Calderone R, Sui B, Tian X, Meng X. Similarities and Differences: A Comparative Review of the Molecular Mechanisms and Effectors of NAFLD and AFLD. Front Physiol 2021; 12:710285. [PMID: 34393826 PMCID: PMC8362097 DOI: 10.3389/fphys.2021.710285] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 06/29/2021] [Indexed: 12/12/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and alcoholic fatty liver disease (AFLD) are the most prevalent metabolic liver diseases globally. Due to the complex pathogenic mechanisms of NAFLD and AFLD, no specific drugs were approved at present. Lipid accumulation, oxidative stress, insulin resistance, inflammation, and dietary habits are all closely related to the pathogenesis of NAFLD and AFLD. However, the mechanism that promotes disease progression has not been fully elucidated. Meanwhile, the gut microbiota and their metabolites also play an important role in the pathogenesis and development of NAFLD and AFLD. This article comparatively reviewed the shared and specific signaling pathways, clinical trials, and potential intervention effectors of NAFLD and AFLD, revealing their similarities and differences. By comparing the shared and specific molecular regulatory mechanisms, this paper provides mutual reference strategies for preventing and treating NAFLD, AFLD, and related metabolic diseases. Furthermore, it provides enlightenment for discovering novel therapies of safe and effective drugs targeting the metabolic liver disease.
Collapse
Affiliation(s)
- Pengyi Zhang
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Weiya Wang
- School of Sports and Health, Shandong Sport University, Jinan, China.,Shandong Academy of Pharmaceutical Science, Jinan, China
| | - Min Mao
- Department of Allied Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Ruolin Gao
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Wenting Shi
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, United States
| | - Bo Sui
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Xuewen Tian
- School of Sports and Health, Shandong Sport University, Jinan, China
| | - Xiangjing Meng
- Shandong Academy of Pharmaceutical Science, Jinan, China
| |
Collapse
|
124
|
Zhang H, Liu B, Jiang S, Wu JF, Qi CH, Mohammadtursun N, Li Q, Li L, Zhang H, Sun J, Dong JC. Baicalin ameliorates cigarette smoke-induced airway inflammation in rats by modulating HDAC2/NF-κB/PAI-1 signalling. Pulm Pharmacol Ther 2021; 70:102061. [PMID: 34314854 DOI: 10.1016/j.pupt.2021.102061] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 07/17/2021] [Accepted: 07/21/2021] [Indexed: 10/20/2022]
Abstract
Chronic obstructive pulmonary disease (COPD) is a chronic inflammatory disease distinguished by airway remodelling and progressive inflammation. PAI-1 is an important regulator of fibrosis. Recent studies have shown that PAI-1 seems to be involved in COPD progression. Elevated levels of PAI-1 have been found in the lungs of patients with acute inflammation. PAI-1 has been shown to regulate the levels of proinflammatory cytokines in the lungs, such as tumour necrosis factor (TNF)-α and interleukin (IL)-6, indicating that PAI-1 may play a fundamental role during inflammation. In the present study, we investigated the anti-inflammatory role of baicalin, the main active component of Scutellaria baicalensis, against cigarette smoke (extract) (CS/CSE)-induced airway inflammation in vivo and in vitro. For the in vivo study, SD rats were exposed to CS for 1 h/day, 6 days/week, for 24 weeks and treated with baicalin (40, 80 and 160 mg/kg) or budesonide (0.2 mg/kg). For this study, HBE cells were pretreated with baicalin (10, 20, 40 μM) or dexamethasone (10-7 M) and then exposed to CSE. We found that baicalin treatment could ameliorate CS-induced airway inflammatory infiltration in rats and decrease PAI-1 expression. The ELISA results showed that baicalin significantly inhibited the levels of TNF-α and IL-1β in CS/CSE-exposed rats and cells. Mechanistic studies showed that baicalin enhanced histone deacetylase 2 (HDAC2) protein expression and inhibited the expression of NF-κB and its downstream target PAI-1, and these effects were reversed by the HDAC2 inhibitor CAY-10683. In conclusion, baicalin ameliorated CS-induced airway inflammation in rats, and these effects were partially attributed to the modulation of HDAC2/NF-κB/PAI-1 signalling.
Collapse
Affiliation(s)
- Hu Zhang
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Baojun Liu
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Shan Jiang
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jin-Feng Wu
- Huashan Hospital, Fudan University, Shanghai, China; Department of Dermatology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Chun-Hui Qi
- Department of Respiratory Medicine, Qingpu District Traditional Chinese Medicine Hospital, Institute of Integrative Medicine, Fudan University, Shanghai, China
| | - Nabijan Mohammadtursun
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Qiuping Li
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Lulu Li
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Hongying Zhang
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jing Sun
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| | - Jing-Cheng Dong
- Huashan Hospital, Fudan University, Shanghai, China; Department of Integrative Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China.
| |
Collapse
|
125
|
Hall RJ, O'Loughlin J, Billington CK, Thakker D, Hall IP, Sayers I. Functional genomics of GPR126 in airway smooth muscle and bronchial epithelial cells. FASEB J 2021; 35:e21300. [PMID: 34165809 DOI: 10.1096/fj.202002073r] [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] [Received: 09/11/2020] [Revised: 12/01/2020] [Accepted: 12/07/2020] [Indexed: 12/12/2022]
Abstract
GPR126 is an adhesion G protein-coupled receptor which lies on chromosome 6q24. Genetic variants in this region are reproducibly associated with lung function and COPD in genome wide association studies (GWAS). The aims of this study were to define the role of GPR126 in the human lung and in pulmonary disease and identify possible casual variants. Online tools (GTEx and LDlink) identified SNPs which may have effects on GPR126 function/ expression, including missense variant Ser123Gly and an intronic variant that shows eQTL effects on GPR126 expression. GPR126 signaling via cAMP-mediated pathways was identified in human structural airway cells when activated with the tethered agonist, stachel. RNA-seq was used to identify downstream genes/ pathways affected by stachel-mediated GPR126 activation in human airway smooth muscle cells. We identified ~350 differentially expressed genes at 4 and 24 hours post stimulation with ~20% overlap. We identified that genes regulated by GPR126 activation include IL33, CTGF, and SERPINE1, which already have known roles in lung biology. Pathways altered by GPR126 included those involved in cell cycle progression and cell proliferation. Here, we suggest a role for GPR126 in airway remodeling.
Collapse
Affiliation(s)
- Robert J Hall
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Jonathan O'Loughlin
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Charlotte K Billington
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Dhruma Thakker
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Ian P Hall
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| | - Ian Sayers
- Division of Respiratory Medicine, National Institute for Health Research, Nottingham Biomedical Research Centre, Biodiscovery Institute, University of Nottingham, Nottingham, UK
| |
Collapse
|
126
|
Karasek D, Spurna J, Macakova D, Cibickova L, Krystynik O, Kucerova V, Ulehlova J, Slavik L. Hypertriglyceridemic Waist in Patients with Type 2 Diabetes: Its Relationship to Selected Markers of Vascular Damage. Metab Syndr Relat Disord 2021; 19:393-400. [PMID: 34096797 DOI: 10.1089/met.2021.0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background: To evaluate the association between hypertriglyceridemic waist (HTGW), a promising marker of visceral adiposity and cardiovascular (CV) risk, and different indicators of vascular damage in type 2 diabetes (T2D) patients. Methods: This case-control study included 161 patients with T2D (91 males, 70 females) and 40 healthy controls (24 males, 16 females). HTWG was defined as waist circumference >90 cm in men or >85 cm in women and triglyceride concentrations >2 mmol/L. In addition to anthropometric and metabolic parameters, markers of endothelial dysfunction, namely von Willebrand factor (vWF) and plasminogen activator inhibitor-1 (PAI-1), were assessed. Arterial stiffness parameters were examined using the SphygmoCor system. Results: Individuals with T2D and HTGW showed the highest elevation of PAI-1 levels and significantly increased vWF levels compared with healthy controls. No significant differences in arterial stiffness markers were observed between T2D individuals. Age and, for several markers, systolic and/or diastolic blood pressure were identified as the main predictors for arterial stiffness, whereas PAI-1 and vWF levels were predicted by metabolic parameters. Conclusions: HTGW represents increased CV risk in T2D patients, mainly due to endothelial damage. The presence of HTGW had no significant effect on arterial stiffness compared with other T2D individuals.
Collapse
Affiliation(s)
- David Karasek
- 3rd Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic, Olomouc, Czech Republic
| | - Jaromira Spurna
- 3rd Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic, Olomouc, Czech Republic
| | - Dominika Macakova
- 3rd Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic, Olomouc, Czech Republic
| | - Lubica Cibickova
- 3rd Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic, Olomouc, Czech Republic
| | - Ondrej Krystynik
- 3rd Department of Internal Medicine-Nephrology, Rheumatology and Endocrinology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic, Olomouc, Czech Republic
| | - Veronika Kucerova
- Department of Clinical Biochemistry, University Hospital Olomouc, Czech Republic, Olomouc, Czech Republic
| | - Jana Ulehlova
- Department of Hemato-Oncology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic, Olomouc, Czech Republic
| | - Ludek Slavik
- Department of Hemato-Oncology, University Hospital Olomouc and Faculty of Medicine and Dentistry, Palacky University Olomouc, Czech Republic, Olomouc, Czech Republic
| |
Collapse
|
127
|
Aleman MN, Díaz EI, Luciardi MC, Mariani AC, Bazán MC, Abregu AV. Hemostatic state of children with type 1 diabetes. Ann Pediatr Endocrinol Metab 2021; 26:99-104. [PMID: 34218631 PMCID: PMC8255861 DOI: 10.6065/apem.2040142.071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
PURPOSE Hyperglycemia is one of the factors responsible for the molecular alterations that modify hemostasis. The aim of this study was to determine the levels of circulating molecules that have a prothrombotic impact on the child and adolescent population with type 1 diabetes mellitus. METHODS There were 35 patients with type 1 diabetes mellitus (11.0±2.5 years of age and a median 3.7±2.0 years of the disease) with no vascular complications and 20 healthy controls with similar age, sex, and body mass index included in the study. The evaluated parameters were fibrinogen, plasminogen activator inhibitor-1 (PAI1), von Willebrand factor antigen, and standard coagulation tests (platelet count, prothrombin time, and activated partial thromboplastin time). Glycemic control was evaluated by hemoglobin A1c and fasting blood glucose tests, and the presence of retinopathy and nephropathy was ruled out. The data obtained were analyzed by IBM SPSS Statistics ver. 20.0 and expressed as mean±standard deviation. The Pearson correlation coefficient was applied to investigate correlations between variables. RESULTS Diabetic patients showed significantly higher levels of fibrinogen (308±66 mg/dL vs. 246±18 mg/dL, P=0.0001), PAI-1 (41.6±12 ng/mL vs. 11.7±1.0 ng/mL, P=0.0001), and von Willebrand factor antigen (284%±55% vs. 121%±19%, P=0.0001). However, standard coagulation tests did not show differences between the 2 groups. PAI-1 was correlated with glycemia, hemoglobin A1c, fibrinogen, and von Willebrand factor antigen. CONCLUSION Elevated levels of fibrinogen, PAI-1, and von Willebrand factor antigen were found in the pediatric and adolescent population with type 1 diabetes mellitus, which suggests a prothrombotic state.
Collapse
Affiliation(s)
- Mariano Nicolás Aleman
- Departamento de Bioquímica Aplicada, Facultad de Bioquímica, Universidad Nacional de Tucumán, Tucumán, Argentina,Address for correspondence: Mariano Nicolás Áleman Departamento de Bioquímica Aplicada, Facultad de Bioquímica, Universidad Nacional de Tucumán, Balcarce 747, San Miguel de Tucumán, Tucumán 4000, Argentina
| | - Elba Irma Díaz
- Departamento de Bioquímica Aplicada, Facultad de Bioquímica, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Maria Constanza Luciardi
- Departamento de Bioquímica Aplicada, Facultad de Bioquímica, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Ana Carolina Mariani
- Departamento de Bioquímica Aplicada, Facultad de Bioquímica, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - Maria Cristina Bazán
- Departamento de Endocrinología, Hospital del Niño Jesús de Tucumán, Tucumán, Argentina
| | - Adela Victoria Abregu
- Departamento de Bioquímica Aplicada, Facultad de Bioquímica, Universidad Nacional de Tucumán, Tucumán, Argentina
| |
Collapse
|
128
|
Breikaa RM, Lilly B. The Notch Pathway: A Link Between COVID-19 Pathophysiology and Its Cardiovascular Complications. Front Cardiovasc Med 2021; 8:681948. [PMID: 34124207 PMCID: PMC8187573 DOI: 10.3389/fcvm.2021.681948] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 05/06/2021] [Indexed: 12/27/2022] Open
Abstract
COVID-19 is associated with a large number of cardiovascular sequelae, including dysrhythmias, myocardial injury, myocarditis and thrombosis. The Notch pathway is one likely culprit leading to these complications due to its direct role in viral entry, inflammation and coagulation processes, all shown to be key parts of COVID-19 pathogenesis. This review highlights links between the pathophysiology of SARS-CoV2 and the Notch signaling pathway that serve as primary drivers of the cardiovascular complications seen in COVID-19 patients.
Collapse
Affiliation(s)
- Randa M. Breikaa
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Molecular, Cellular and Developmental Biology Program, The Ohio State University, Columbus, OH, United States
| | - Brenda Lilly
- Center for Cardiovascular Research and The Heart Center, Nationwide Children's Hospital, Columbus, OH, United States
- Department of Pediatrics, The Ohio State University, Columbus, OH, United States
| |
Collapse
|
129
|
PAI-1 4G/4G Genotype Is Associated with Recurrent Implantation Failure: a Systematic Review and Meta-analysis. Reprod Sci 2021; 28:3051-3060. [PMID: 34019258 DOI: 10.1007/s43032-021-00623-1] [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: 12/09/2020] [Accepted: 05/13/2021] [Indexed: 10/21/2022]
Abstract
To detect the association between PAI-1 -675 4G/5G polymorphism and recurrent implantation failure (RIF). We performed this meta-analysis by searching databases of PubMed, EMBASE, OVID, and CNKI (China National Knowledge Infrastructure) for case-control studies that evaluated the association between PAI 4G/5G polymorphism and RIF. Meta-analysis was performed using the random-effects model. The odds ratios (ORs) with 95% confidence intervals (CIs) were reported to evaluate the association. Meta-regression and subgroup analysis were performed to explore the source of heterogeneity. Sensitivity analysis and trim-and-fill analysis were performed to explore the robustness of the meta-analysis. Eight case-control studies consisted of 1273 women were included in this meta-analysis (including 697 RIF patients and 576 control participants). The combined results showed that the homozygous genotype of PAI-1 -675 4G/4G was significantly associated with RIF (OR 2.79, 95%CI 1.53-5.08, P-value = 0.0008). Meta-regression and subgroup analysis showed that sample origin is the primary source of heterogeneity (P-value for meta-regression: 0.005). Study quality also explains some heterogeneity (P-value for meta-regression: 0.03). Sensitivity analysis showed that the result was not significantly changed after excluding one study each time. Trim-and-fill analysis showed that the result was not significantly changed after filled with three studies. PAI -675 4G/4G genotype may serve as one of the predisposing factors of RIF. Women with PAI-1 4G/4G genotype were at higher risk of RIF. However, more high-quality studies are needed to confirm the conclusion.
Collapse
|
130
|
Wu D, Yang XO. Dysregulation of Pulmonary Responses in Severe COVID-19. Viruses 2021; 13:957. [PMID: 34064104 PMCID: PMC8224314 DOI: 10.3390/v13060957] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/04/2021] [Accepted: 05/19/2021] [Indexed: 12/13/2022] Open
Abstract
Patients with coronavirus disease 2019 (COVID-19) predominantly have a respiratory tract infection with various symptoms and high mortality is associated with respiratory failure second to severe disease. The risk factors leading to severe disease remain unclear. Here, we reanalyzed a published single-cell RNA-Seq (scRNA-Seq) dataset and found that bronchoalveolar lavage fluid (BALF) of patients with severe disease compared to those with mild disease contained decreased TH17-type cells, decreased IFNA1-expressing cells with lower expression of toll-like receptor 7 (TLR7) and TLR8, increased IgA-expressing B cells, and increased hyperactive epithelial cells (and/or macrophages) expressing matrix metalloproteinases (MMPs), hyaluronan synthase 2 (HAS2), and plasminogen activator inhibitor-1 (PAI-1), which may together contribute to the pulmonary pathology in severe COVID-19. We propose IFN-I (and TLR7/TLR8) and PAI-1 as potential biomarkers to predict the susceptibility to severe COVID-19.
Collapse
Affiliation(s)
- Dandan Wu
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA;
- Department of Crop Genetics and Breeding, College of Agronomy, Hunan Agricultural University, Changsha 410128, China
| | - Xuexian O. Yang
- Department of Molecular Genetics and Microbiology, University of New Mexico School of Medicine, Albuquerque, NM 87131, USA;
| |
Collapse
|
131
|
Norooznezhad AH, Mansouri K. Endothelial cell dysfunction, coagulation, and angiogenesis in coronavirus disease 2019 (COVID-19). Microvasc Res 2021; 137:104188. [PMID: 34022205 PMCID: PMC8135191 DOI: 10.1016/j.mvr.2021.104188] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/08/2021] [Accepted: 05/17/2021] [Indexed: 12/14/2022]
Abstract
Coronavirus disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been led to a pandemic emergency. So far, different pathological pathways for SARS-CoV-2 infection have been introduced in which the excess release of pro-inflammatory cytokines (such as interleukin 1 β [IL-1β], IL-6, and tumor necrosis factor α [TNFα]) has earned most of the attentions. However, recent studies have identified new pathways with at least the same level of importance as cytokine storm in which endothelial cell (EC) dysfunction is one of them. In COVID-19, two main pathologic phenomena have been seen as a result of EC dysfunction: hyper-coagulation state and pathologic angiogenesis. The EC dysfunction-induced hypercoagulation state seems to be caused by alteration in the levels of different factors such as plasminogen activator inhibitor 1 (PAI-1), von Willebrand factor (vWF) antigen, soluble thrombomodulin, and tissue factor pathway inhibitor (TFPI). As data have shown, these thromboembolic events are associated with severity of disease severity or even death in COVID-19 patients. Other than thromboembolic events, pathologic angiogenesis is among the recent findings. Furthermore, over-expression/higher levels of different proangiogenic factors such as vascular endothelial growth factor (VEGF), hypoxia-inducible factor 1 α (HIF-1α), IL-6, TNF receptor super family 1A and 12, and angiotensin-converting enzyme 2 (ACE2) have been found in the lung biopsies/sera of both survived and non-survived COVID-19 patients. Also, there are some hypotheses regarding the role of nitric oxide in EC dysfunction and acute respiratory distress syndrome (ARDS) in SARS-CoV-2 infection. It has been demonstrated that different pathways involved in inflammation are generally common with EC dysfunction and angiogenesis. Altogether, considering the common possible upstream pathways in cytokine storm, pathologic angiogenesis, and EC dysfunction, it seems that targeting these molecules (such as nuclear factor κB) could be more effective in the management of patients with COVID-19.
Collapse
Affiliation(s)
- Amir Hossein Norooznezhad
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Kamran Mansouri
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|
132
|
Plasminogen activator inhibitor 1 and venous thrombosis in pancreatic cancer. Blood Adv 2021; 5:487-495. [PMID: 33496742 DOI: 10.1182/bloodadvances.2020003149] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 12/17/2020] [Indexed: 12/25/2022] Open
Abstract
Pancreatic cancer patients have a high risk of venous thromboembolism (VTE). Plasminogen activator inhibitor 1 (PAI-1) inhibits plasminogen activators and increases the risk of thrombosis. PAI-1 is expressed by pancreatic tumors and human pancreatic cell lines. However, to date, there are no studies analyzing the association of active PAI-1 and VTE in pancreatic cancer patients. We investigated the association of active PAI-1 in plasma and VTE in pancreatic cancer patients. In addition, we determined if the presence of human pancreatic tumors expressing PAI-1 impairs venous thrombus resolution in mice. Plasma levels of active PAI-1 in patients with pancreatic cancer and mice bearing human tumors were determined by enzyme-linked immunosorbent assay. We measured PAI-1 expression in 5 different human pancreatic cancer cell lines and found that PANC-1 cells expressed the highest level. PANC-1 tumors were grown in nude mice. Venous thrombosis was induced by complete ligation of the inferior vena cava (IVC). Levels of active PAI-1 were independently associated with increased risk of VTE in patients with pancreatic cancer (subdistribution hazard ratio per doubling of levels: 1.39 [95% confidence interval, 1.09-1.78], P = .007). Mice bearing PANC-1 tumors had increased levels of both active human and active mouse PAI-1 and decreased levels of plasmin activity. Importantly, mice bearing PANC-1 tumors exhibited impaired venous thrombus resolution 8 days after IVC stasis compared with nontumor controls. Our results suggest that PAI-1 contributes to VTE in pancreatic cancer.
Collapse
|
133
|
Lugo-Gavidia LM, Burger D, Matthews VB, Nolde JM, Galindo Kiuchi M, Carnagarin R, Kannenkeril D, Chan J, Joyson A, Herat LY, Azzam O, Schlaich MP. Role of Microparticles in Cardiovascular Disease: Implications for Endothelial Dysfunction, Thrombosis, and Inflammation. HYPERTENSION (DALLAS, TEX. : 1979) 2021; 77:1825-1844. [PMID: 33979187 DOI: 10.1161/hypertensionaha.121.16975] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Leslie Marisol Lugo-Gavidia
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Dylan Burger
- Kidney Research Centre, The Ottawa Hospital Research Institute, Department of Cellular and Molecular Medicine, University of Ottawa (D.B.)
| | - Vance B Matthews
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Janis M Nolde
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Márcio Galindo Kiuchi
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Revathy Carnagarin
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Dennis Kannenkeril
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.).,Department of Nephrology and Hypertension, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Germany (D.K.)
| | - Justine Chan
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Anu Joyson
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Lakshini Y Herat
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.)
| | - Omar Azzam
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.).,Department of Internal Medicine (O.A.), Royal Perth Hospital, Western Australia
| | - Markus P Schlaich
- Dobney Hypertension Centre, School of Medicine, Royal Perth Hospital Unit, Medical Research Foundation, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia (L.M.L.-G., V.B.M., J.M.N., M.G.K., R.C., D.K., J.C., A.J., L.Y.H., O.A., M.P.S.).,Departments of Cardiology and Nephrology (M.P.S.), Royal Perth Hospital, Western Australia.,Neurovascular Hypertension and Kidney Disease Laboratory, Baker Heart and Diabetes Institute, Melbourne, Australia (M.P.S.)
| |
Collapse
|
134
|
Robinson S, Chang J, Parigoris E, Hecker L, Takayama S. Aqueous two-phase deposition and fibrinolysis of fibroblast-laden fibrin micro-scaffolds. Biofabrication 2021; 13. [PMID: 33440354 DOI: 10.1088/1758-5090/abdb85] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 01/13/2021] [Indexed: 11/12/2022]
Abstract
This paper describes printing of microscale fibroblast-laden matrices using an aqueous two-phase approach that controls thrombin-mediated enzymatic crosslinking of fibrin. Optimization of aqueous two-phase formulations enabled polymerization of consistent sub-microliter volumes of cell-laden fibrin. When plasminogen was added to these micro-scaffolds, the primary normal human lung fibroblasts converted it to plasmin, triggering gradual degradation of the fibrin. Time-lapse live-cell imaging and automated image analysis provided readouts of time to degradation of 50% of the scaffold as well as maximum degradation rate. The time required for degradation decreased linearly with cell number while it increased in a dose-dependent manner upon addition of TGF-β1. Fibroblasts isolated from idiopathic pulmonary fibrosis patients showed similar trends with regards to response to TGF-β1 stimulation. Addition of reactive oxygen species (ROS) slowed fibrinolysis but only in the absence of TGF-β1, consistent with published studies demonstrating that pro-fibrotic cellular phenotypes induced by TGF-β1 are mediated, at least in part, through increased production of ROS. FDA-approved and experimental anti-fibrosis drugs were also tested for their effects on fibrinolysis rates. Given the central role of fibrinolysis in both normal and pathogenic wound healing of various tissues, the high-throughput cell-mediated fibrinolysis assay described has broad applicability in the study of many different cell types and diseases. Furthermore, aqueous two-phase printing of fibrin addresses several current limitations of fibrin bio-inks, potentially enabling future applications in tissue engineering andin vitromodels.
Collapse
Affiliation(s)
- Stephen Robinson
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia, United States of America.,The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Jonathan Chang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia, United States of America.,The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Eric Parigoris
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia, United States of America.,The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| | - Louise Hecker
- Division of Pulmonary, Allergy, Critical Care, and Sleep Medicine, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Shuichi Takayama
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory School of Medicine, Atlanta, Georgia, United States of America.,The Parker H. Petit Institute of Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, Georgia, United States of America
| |
Collapse
|
135
|
Bruno G, Ritelli M, Di Pietro A, Cipriano L, Colombi M, Lus G, Puoti G. Clinical and Genetic Heterogeneity in a Large Family with Pseudoxanthoma Elasticum: MTHFR and SERPINE1 Variants as Possible Disease Modifiers in Developing Ischemic Stroke. J Stroke Cerebrovasc Dis 2021; 30:105744. [PMID: 33813081 DOI: 10.1016/j.jstrokecerebrovasdis.2021.105744] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Revised: 03/02/2021] [Accepted: 03/07/2021] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND AND OBJECTIVES Pseudoxanthoma elasticum (PXE) is a rare autosomal recessive disorder caused by pathogenic variants in the ABCC6 gene. The phenotypic spectrum of PXE is highly variable and includes principally three major features: skin lesions, eye and vascular manifestations, while brain manifestations are less common. To date about 400 different PXE associated variants in ABCC6 gene are described without any evident genotype-phenotype correlation. Herein, we report the clinical and molecular findings of a large PXE family with clinical and genetic intra-familial variability with significant cerebrovascular involvement. METHODS The analysis of the ABCC6 gene was performed in the proband and her familiars for the definition of genetic background. Then, in order to determine why some affected individuals had more prominent brain involvement, we investigated classic thrombophilic gene variants. RESULTS Molecular findings disclosed two different ABCC6 mutations, i.e., the recurrent p.(Arg518Gln) and the novel p.(Val1285Met) missense substitution responsible of a pseudo-dominant inheritance. The study of thrombophilic gene variants revealed the presence of 4G/4G SERPINE1 genotype in the proband and in her father, which both developed ischemic stroke. The proband carried also the C677T variant the MTHFR gene. CONCLUSION We argue, for the first time, that the 4G/4G SERPINE1 genotype could represent an additional risk factor in PXE for developing ischemic stroke, which adds up to the already known predisposing conditions. Therapeutic implications are discussed, we also advise that PXE patients should be adequately screened for cerebral vasculopathy, even more if familial history is suggestive of brain complications.
Collapse
Affiliation(s)
- Giorgia Bruno
- Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marco Ritelli
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Andrea Di Pietro
- Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Lorenzo Cipriano
- Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Marina Colombi
- Division of Biology and Genetics, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Giacomo Lus
- Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gianfranco Puoti
- Division of Neurology, Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
136
|
Deinhardt-Emmer S, Wittschieber D, Sanft J, Kleemann S, Elschner S, Haupt KF, Vau V, Häring C, Rödel J, Henke A, Ehrhardt C, Bauer M, Philipp M, Gaßler N, Nietzsche S, Löffler B, Mall G. Early postmortem mapping of SARS-CoV-2 RNA in patients with COVID-19 and the correlation with tissue damage. eLife 2021; 10:e60361. [PMID: 33781385 PMCID: PMC8009677 DOI: 10.7554/elife.60361] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 02/18/2021] [Indexed: 02/07/2023] Open
Abstract
Clinical observations indicate that COVID-19 is a systemic disease. An investigation of the viral distribution within the human body and its correlation with tissue damage can aid in understanding the pathophysiology of SARS-CoV-2 infection. We present a detailed mapping of the viral RNA in 61 tissues and organs of 11 deceased patients with COVID-19. The autopsies were performed within the early postmortem interval (between 1.5 and 15 hr, mean: 5.6 hr) to minimize the bias due to viral RNA and tissue degradation. Very high viral loads (>104copies/ml) were detected in most patients' lungs, and the presence of intact viral particles in the lung tissue could be verified by transmission electron microscopy. Interestingly, viral RNA was detected throughout various extrapulmonary tissues and organs without visible tissue damage. The dissemination of SARS-CoV-2-RNA throughout the body supports the hypothesis that there is a maladaptive host response with viremia and multiorgan dysfunction.
Collapse
Affiliation(s)
| | | | - Juliane Sanft
- Institute of Forensic Medicine, Jena University HospitalJenaGermany
| | - Sandra Kleemann
- Institute of Forensic Medicine, Jena University HospitalJenaGermany
| | - Stefan Elschner
- Institute of Forensic Medicine, Jena University HospitalJenaGermany
| | | | - Vanessa Vau
- Institute of Medical Microbiology, Jena University HospitalJenaGermany
| | - Clio Häring
- Section of Experimental Virology, Institute of Medical Microbiology, Jena University HospitalJenaGermany
| | - Jürgen Rödel
- Institute of Medical Microbiology, Jena University HospitalJenaGermany
| | - Andreas Henke
- Section of Experimental Virology, Institute of Medical Microbiology, Jena University HospitalJenaGermany
| | - Christina Ehrhardt
- Section of Experimental Virology, Institute of Medical Microbiology, Jena University HospitalJenaGermany
| | - Michael Bauer
- Department of Anaesthesiology and Intensive Care Medicine, Jena University HospitalJenaGermany
| | - Mike Philipp
- Department of Anaesthesiology and Intensive Care Medicine, Greiz General HospitalGreizGermany
| | - Nikolaus Gaßler
- Section of Surgical Pathology, Institute of Forensic Medicine, Jena University HospitalJenaGermany
| | - Sandor Nietzsche
- Department of Electron Microscopy, Jena University HospitalJenaGermany
| | - Bettina Löffler
- Institute of Medical Microbiology, Jena University HospitalJenaGermany
| | - Gita Mall
- Institute of Forensic Medicine, Jena University HospitalJenaGermany
| |
Collapse
|
137
|
McKellar J, Rebendenne A, Wencker M, Moncorgé O, Goujon C. Mammalian and Avian Host Cell Influenza A Restriction Factors. Viruses 2021; 13:522. [PMID: 33810083 PMCID: PMC8005160 DOI: 10.3390/v13030522] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/12/2021] [Accepted: 03/15/2021] [Indexed: 12/27/2022] Open
Abstract
The threat of a new influenza pandemic is real. With past pandemics claiming millions of lives, finding new ways to combat this virus is essential. Host cells have developed a multi-modular system to detect incoming pathogens, a phenomenon called sensing. The signaling cascade triggered by sensing subsequently induces protection for themselves and their surrounding neighbors, termed interferon (IFN) response. This response induces the upregulation of hundreds of interferon-stimulated genes (ISGs), including antiviral effectors, establishing an antiviral state. As well as the antiviral proteins induced through the IFN system, cells also possess a so-called intrinsic immunity, constituted of antiviral proteins that are constitutively expressed, creating a first barrier preceding the induction of the interferon system. All these combined antiviral effectors inhibit the virus at various stages of the viral lifecycle, using a wide array of mechanisms. Here, we provide a review of mammalian and avian influenza A restriction factors, detailing their mechanism of action and in vivo relevance, when known. Understanding their mode of action might help pave the way for the development of new influenza treatments, which are absolutely required if we want to be prepared to face a new pandemic.
Collapse
Affiliation(s)
- Joe McKellar
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| | - Antoine Rebendenne
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| | - Mélanie Wencker
- Centre International de Recherche en Infectiologie, INSERM/CNRS/UCBL1/ENS de Lyon, 69007 Lyon, France;
| | - Olivier Moncorgé
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| | - Caroline Goujon
- Institut de Recherche en Infectiologie de Montpellier, CNRS, Université de Montpellier, CEDEX 5, 34293 Montpellier, France; (J.M.); (A.R.)
| |
Collapse
|
138
|
PAI-1 in Diabetes: Pathophysiology and Role as a Therapeutic Target. Int J Mol Sci 2021; 22:ijms22063170. [PMID: 33804680 PMCID: PMC8003717 DOI: 10.3390/ijms22063170] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 12/14/2022] Open
Abstract
Hypofibrinolysis is a key abnormality in diabetes and contributes to the adverse vascular outcome in this population. Plasminogen activator inhibitor (PAI)-1 is an important regulator of the fibrinolytic process and levels of this antifibrinolytic protein are elevated in diabetes and insulin resistant states. This review describes both the physiological and pathological role of PAI-1 in health and disease, focusing on the mechanism of action as well as protein abnormalities in vascular disease with special focus on diabetes. Attempts at inhibiting protein function, using different techniques, are also discussed including direct and indirect interference with production as well as inhibition of protein function. Developing PAI-1 inhibitors represents an alternative approach to managing hypofibrinolysis by targeting the pathological abnormality rather than current practice that relies on profound inhibition of the cellular and/or acellular arms of coagulation, and which can be associated with increased bleeding events. The review offers up-to-date knowledge on the mechanisms of action of PAI-1 together with the role of altering protein function to improve hypofirbinolysis. Developing PAI-1 inhibitors may form for the basis of future new class of antithrombotic agents that reduce vascular complications in diabetes.
Collapse
|
139
|
Roomruangwong C, Sirivichayakul S, Matsumoto AK, Michelin AP, de Oliveira Semeão L, de Lima Pedrão JV, Barbosa DS, Moreira EG, Maes M. Menstruation distress is strongly associated with hormone-immune-metabolic biomarkers. J Psychosom Res 2021; 142:110355. [PMID: 33444909 DOI: 10.1016/j.jpsychores.2020.110355] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To examine the associations between menstruation features and symptoms and hormone-immune-metabolic biomarkers. METHODS Forty-one women completed questionnaires assessing characteristic menstruation symptoms, duration of menstrual cycle and number of pads used/day and completed the Daily Record of Severity of Problems (DRSP) during the consecutive days of their menstrual cycle. Menses-related symptoms (MsRS) were computed from the sum of 10 pre- and post-menses symptoms and the menstruation blood and duration index (MBDI) was computed based on the daily number of pads and duration of menses. We assayed serum levels of various biomarkers at days 7, 14, 21, and 28 of the subjects' menstrual cycle. RESULTS MBDI was significantly associated with a) MsRS including low abdominal cramps, and gastro-intestinal (GI) and pain symptoms (positively); b) plasma levels of haptoglobin (Hp), CCL5, insulin growth factor (IGF)-1, and plasminogen activator inhibitor (PAI)1 (all positively); and c) estradiol and paraoxonase (PON)1 arylesterase activity (both inversely). MsRS were significantly predicted by CCL5 and IGF-1 (both positively) and progesterone (inversely). Low-abdominal cramps, and gastro-intestinal and pain symptoms were associated with lower progesterone levels. The MBDI+MsRS score was significantly predicted by the cumulative effects of (in descending order of importance): Hp, IGF-1, PON1 arylesterase, estradiol and PAI. CONCLUSION Menstruation-related features including estimated blood loss, duration of menses, cramps, pain, and gastro-intestinal symptoms are associated with hormone-immune-metabolic biomarkers, which mechanistically may explain those features. Future research should construct a cross-validated algorithm using MBDI+MsRS features in a larger study group to delineate a useful case-definition of menstruation-related distress.
Collapse
Affiliation(s)
- Chutima Roomruangwong
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Sunee Sirivichayakul
- Department of Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Andressa Keiko Matsumoto
- Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, PR, Brazil
| | - Ana Paula Michelin
- Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, PR, Brazil
| | - Laura de Oliveira Semeão
- Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, PR, Brazil
| | - João Victor de Lima Pedrão
- Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, PR, Brazil
| | - Decio S Barbosa
- Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, PR, Brazil
| | - Estefania G Moreira
- Health Sciences Graduate Program, Health Sciences Center, State University of Londrina, Londrina, PR, Brazil
| | - Michael Maes
- Department of Psychiatry, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Department of Psychiatry, Medical University Plovdiv, Plovdiv, Bulgaria; IMPACT Strategic Research Center, Deakin University, Geelong, Australia.
| |
Collapse
|
140
|
Koca N, Ayar K, Bal Ö, Ersoy C. The evaluation of the role of BMI and insulin resistance on inflammatory markers, PAI-1 levels and arterial stiffness in newly diagnosed type 2 diabetes mellitus patients. Minerva Endocrinol (Torino) 2021; 46:116-123. [PMID: 33779113 DOI: 10.23736/s2724-6507.20.03158-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Increased cardiovascular risk, represented by endothelial inflammation, probably starts with the very first course of type-2 diabetes (T2DM). Almost 85.2% of all T2DM patients are overweight or obese. Thrombosis accounts 80% of all deaths in patients with diabetes. The thrombotic-fibrinolytic equilibrium shifts in favor of thrombosis by plasminogen activator inhibitor-1 (PAI-1). PAI-1 secretion is induced primarily by CRP. PAI-1 overexpression predisposes unstable plaque development. The contribution of obesity and diabetes to this process is not clearly understood. In this study, we aimed to investigate comparison of inflammatory markers, PAI-1 levels and arterial stiffness according to BMI and impaired glucose metabolism in patient with newly diagnosed T2DM. METHODS Newly diagnosed 60 T2DM patients were enrolled. Demographics and measurements were noted. Liver (AST, ALT), kidney (urea, creatinine, albumin/creatinine ratio), metabolic (fasting blood glucose, post-prandial blood glucose, insulin, c-peptide, HbA1c, total cholesterol, low-density lipoprotein [LDL], high-density lipoprotein [HDL], triglyceride) parameters, inflammatory markers [hsCRP, fibrinogen]), PAI-1 levels and pulse wave velocity was measured from all participants. The results were compared. RESULTS Inflammatory markers and PAI-1 levels were significantly elevated in obese group compared to overweight participants. The correlation analysis showed that waist and hip circumferences, high-sensitive CRP, fibrinogen and PAI-1 levels were positively correlated with BMI but not with HbA1c levels. CONCLUSIONS The results of our study showed that lipid levels, glycemic and blood pressure values of the obese and overweight patients were similar. BMI affects inflammatory markers and PAI-1 levels independent of glucose regulation and insulin resistance in newly diagnosed T2DM. According to the current study BMI is found to be more prominent in terms of inflammatory markers and PAI-1 levels compared to insulin resistance and impaired glucose metabolism in newly diagnosed T2DM.
Collapse
Affiliation(s)
- Nizameddin Koca
- Department of Internal Medicine, University of Health Sciences, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey -
| | - Koray Ayar
- Department of Rheumatology, University of Health Sciences, Bursa Yuksek Ihtisas Training and Research Hospital, Bursa, Turkey
| | - Öznur Bal
- Department of Medical Oncology, Ankara Numune Training and Research Hospital, Ankara, Turkey
| | - Canan Ersoy
- Department of Internal Medicine, Division of Endocrinology and Metabolism, Uludag University Hospital, Bursa, Turkey
| |
Collapse
|
141
|
Echeverría LE, Rojas LZ, Gómez-Ochoa SA. Coagulation disorders in Chagas disease: A pathophysiological systematic review and meta-analysis. Thromb Res 2021; 201:73-83. [PMID: 33652329 DOI: 10.1016/j.thromres.2021.02.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/10/2021] [Accepted: 02/16/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Currently, Chagas disease (CD) constitutes one of the main public health problems in Latin America. However, little is known about potential mechanisms of disease different from cardiac or digestive involvement, such as the coagulation disorders elicited by the parasite persistence in the tissues. The aim of this systematic review was to describe and characterize all the published literature that evaluated the pathophysiological aspects of coagulation disorders in CD. METHODS Searches in Medline, EMBASE, and LILACS databases (from inception to July 28th, 2020) were performed. Articles of any language reporting the levels of different coagulation factors/markers or the prevalence of abnormal levels of the mentioned molecules in patients with CD were included. Two reviewers independently selected the studies, extracted the data, and assessed the quality of evidence. Estimates were pooled using random-effects meta-analyses. RESULTS Seven studies evaluating a total of 676 participants fulfilled the criteria and were included, while only six were suitable for meta-analyzing (544 participants, 52% men, mean age: 49 ± 8 years). 57.16% of the patients in the meta-analysis had a serological confirmed diagnosis of CD, while 97% of these were in the indeterminate stage of the disease. Patients in the CD group had higher levels of F 1 + 2 (SMD 5.15. 95% CI 1.92, 8.38), PAI-1 (SMD 0.46. 95% CI 0.07; 0.89), and P-selectin (SMD 1.8; 95% CI 0.13-3.47) compared to healthy controls. Furthermore, benznidazole therapy was associated with a reduction in the levels of these biomarkers after treatment. CONCLUSION The results of the present study suggest that patients with chronic T. cruzi infection are affected by a potential hypercoagulable state irrespective of the development of cardiac or digestive disease. Furthermore, the reduction in the levels of the coagulation markers after benznidazole therapy may suggest a significant role of the parasite load in the development of these coagulation disorders. There is a scarcity of research assessing the molecular and pathophysiological mechanisms of coagulation disorders in Chagas disease. Further research is needed to assess the benefit of benznidazole therapy on this hypercoagulable state in the long-term, along with its impact on the risk of thromboembolic events in CD patients.
Collapse
Affiliation(s)
- Luis E Echeverría
- Heart Failure and Heart Transplant Clinic, Fundación Cardiovascular de Colombia, Floridablanca, Colombia.
| | - Lyda Z Rojas
- Research Group and Development of Nursing Knowledge (GIDCEN-FCV), Research Center, Cardiovascular Foundation of Colombia, Floridablanca, Santander, Colombia
| | | |
Collapse
|
142
|
Chukowry PS, Spittle DA, Turner AM. Small Airways Disease, Biomarkers and COPD: Where are We? Int J Chron Obstruct Pulmon Dis 2021; 16:351-365. [PMID: 33628018 PMCID: PMC7899307 DOI: 10.2147/copd.s280157] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Accepted: 12/11/2020] [Indexed: 11/23/2022] Open
Abstract
The response to treatment and progression of Chronic Obstructive Pulmonary Disease (COPD) varies significantly. Small airways disease (SAD) is being increasingly recognized as a key pathological feature of COPD. Studies have brought forward pathological evidence of small airway damage preceding the development of emphysema and the detection of obstruction using traditional spirometry. In recent years, there has been a renewed interest in the early detection of SAD and this has brought along an increased demand for physiological tests able to identify and quantify SAD. Early detection of SAD allows early targeted therapy and this suggests the potential for altering the course of disease. The aim of this article is to review the evidence available on the physiological testing of small airways. The first half will focus on the role of lung function tests such as maximum mid-expiratory flow, impulse oscillometry and lung clearance index in detecting and quantifying SAD. The role of Computed Tomography (CT) as a radiological biomarker will be discussed as well as the potential of recent CT analysis software to differentiate normal aging of the lungs to pathology. The evidence behind SAD biomarkers sourced from blood as well as biomarkers sourced from sputum and broncho-alveolar lavage (BAL) will be reviewed. This paper focuses on CC-16, sRAGE, PAI-1, MMP-9 and MMP-12.
Collapse
Affiliation(s)
- Priyamvada S Chukowry
- Respiratory Research Department, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Daniella A Spittle
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, B15 2TT, UK
| | - Alice M Turner
- Institute for Applied Health Research, University of Birmingham, Birmingham, B15 2TT, UK
| |
Collapse
|
143
|
Kwak SY, Park S, Kim H, Lee SJ, Jang WS, Kim MJ, Lee S, Jang WI, Kim AR, Kim EH, Shim S, Jang H. Atorvastatin Inhibits Endothelial PAI-1-Mediated Monocyte Migration and Alleviates Radiation-Induced Enteropathy. Int J Mol Sci 2021; 22:ijms22041828. [PMID: 33673196 PMCID: PMC7917640 DOI: 10.3390/ijms22041828] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2021] [Revised: 02/08/2021] [Accepted: 02/09/2021] [Indexed: 01/15/2023] Open
Abstract
Intestinal injury is observed in cancer patients after radiotherapy and in individuals exposed to radiation after a nuclear accident. Radiation disrupts normal vascular homeostasis in the gastrointestinal system by inducing endothelial damage and senescence. Despite advances in medical technology, the toxicity of radiation to healthy tissue remains an issue. To address this issue, we investigated the effect of atorvastatin, a commonly prescribed hydroxy-3-methylglutaryl-coenzyme A reductase inhibitor of cholesterol synthesis, on radiation-induced enteropathy and inflammatory responses. We selected atorvastatin based on its pleiotropic anti-fibrotic and anti-inflammatory effects. We found that atorvastatin mitigated radiation-induced endothelial damage by regulating plasminogen activator inhibitor-1 (PAI-1) using human umbilical vein endothelial cells (HUVECs) and mouse model. PAI-1 secreted by HUVECs contributed to endothelial dysfunction and trans-endothelial monocyte migration after radiation exposure. We observed that PAI-1 production and secretion was inhibited by atorvastatin in irradiated HUVECs and radiation-induced enteropathy mouse model. More specifically, atorvastatin inhibited PAI-1 production following radiation through the JNK/c-Jun signaling pathway. Together, our findings suggest that atorvastatin alleviates radiation-induced enteropathy and supports the investigation of atorvastatin as a radio-mitigator in patients receiving radiotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Sehwan Shim
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| | - Hyosun Jang
- Correspondence: (S.S.); (H.J.); Tel.: +82-2-3399-5873 (S.S.); +82-2-970-1302 (H.J.)
| |
Collapse
|
144
|
Sabouni R, Archer DF, Jacot T. Drospirenone Effects on the Plasminogen Activator System in Immortalized Human Endometrial Endothelial Cells. Reprod Sci 2021; 28:1974-1980. [PMID: 33559058 DOI: 10.1007/s43032-020-00433-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 12/13/2020] [Indexed: 12/09/2022]
Abstract
Drospirenone (DRSP) is a fourth-generation progestin that interacts with the progesterone receptor (PR) and androgen receptor (AR) in addition to uniquely interacting to the mineralocorticoid receptor (MR). The known effects of DRSP via the mineralocorticoid receptor (MR) are limited. This study seeks to determine if DRSP alters plasminogen activator inhibitor-1 (PAI-1) and tissue plasminogen activator (tPA) in human immortalized endometrial endothelial cells (HEEC) and if such changes in the plasminogen activator system (PAS) are mediated through the MR or AR. The in vitro cell culture experiments utilizing an immortalized human endometrial endothelial cell line evaluated two concentrations of DRSP on PAI-1 and tPA levels in the culture media using specific enzyme-linked immunoassays (ELISA). Experiments adding DRSP with an androgen receptor blocker, flutamide, or a mineralocorticoid receptor agonist, aldosterone, were performed to elucidate which receptor(s) mediated the PAS effects. DRSP 10 μM significantly decreased both HEEC levels of PAI-1 and tPA to 0.75 ± 0.04 and 0.82 ± 0.05 of control, respectively. These direct effects were blunted by flutamide, an AR antagonist. PAI-1 and tPA were not changed by the MR agonist, aldosterone. DRSP significantly decreased both PAI-1 and tPA in the HEECs via the androgen receptor.
Collapse
Affiliation(s)
- Reem Sabouni
- Department of Obstetrics and Gynecology, Jones Institute for Reproductive Medicine/Eastern Virginia Medical School, Norfolk, VA, USA.
| | - David F Archer
- Department of Obstetrics and Gynecology, Jones Institute for Reproductive Medicine/Eastern Virginia Medical School, Norfolk, VA, USA
| | - Terry Jacot
- Department of Obstetrics and Gynecology, Jones Institute for Reproductive Medicine/Eastern Virginia Medical School, Norfolk, VA, USA
| |
Collapse
|
145
|
Ntzouvani A, Antonopoulou S, Fragopoulou E, Kontogianni MD, Nomikos T, Mikellidi A, Xanthopoulou Μ, Kalogeropoulos N, Panagiotakos D. Effect of Differently Fed Farmed Gilthead Sea Bream Consumption on Platelet Aggregation and Circulating Haemostatic Markers among Apparently Healthy Adults: A Double-Blind Randomized Crossover Trial. Nutrients 2021; 13:nu13020286. [PMID: 33498445 PMCID: PMC7909403 DOI: 10.3390/nu13020286] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/15/2021] [Accepted: 01/17/2021] [Indexed: 01/21/2023] Open
Abstract
Fish consumption beneficially affects coagulation markers. Few dietary intervention studies have investigated differently fed farmed fish against these cardio-metabolic risk factors in humans. This double-blind randomized crossover trial evaluated differently fed farmed gilthead sea bream consumption against platelet aggregation and circulating haemostatic markers among apparently healthy adults. Subjects aged 30-65 years, with a body mass index 24.0-31.0 kg/m2, consuming less than 150 g cooked fish per week, were recruited in Attica, Greece. Participants were randomized (n = 38, 1:1) to one of two sequences; consumption of fish fed with fish oil diet (conventional fish, CF)/fish fed with olive pomace-enriched diet (enriched fish, EF) versus EF/CF. The primary outcomes were ex vivo human platelet aggregation and circulating plasminogen activator inhibitor-1 (PAI-1) and P-selectin (sP-selectin) concentrations. EF consumption had no significant effect on platelet sensitivity or haemostatic markers compared to CF. Platelet sensitivity to platelet-activating factor (PAF) decreased after CF consumption during the second period (p < 0.01). Plasma PAI-1 and sP-selectin concentrations increased after CF consumption during both periods (p < 0.01 for both). Based on current findings, consumption of enriched farmed gilthead sea bream had no greater effect on coagulation markers in adults compared to the conventionally fed fish.
Collapse
|
146
|
Schaid MD, Zhu Y, Richardson NE, Patibandla C, Ong IM, Fenske RJ, Neuman JC, Guthery E, Reuter A, Sandhu HK, Fuller MH, Cox ED, Davis DB, Layden BT, Brasier AR, Lamming DW, Ge Y, Kimple ME. Systemic Metabolic Alterations Correlate with Islet-Level Prostaglandin E 2 Production and Signaling Mechanisms That Predict β-Cell Dysfunction in a Mouse Model of Type 2 Diabetes. Metabolites 2021; 11:metabo11010058. [PMID: 33467110 PMCID: PMC7830513 DOI: 10.3390/metabo11010058] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/30/2020] [Accepted: 01/07/2021] [Indexed: 12/18/2022] Open
Abstract
The transition from β-cell compensation to β-cell failure is not well understood. Previous works by our group and others have demonstrated a role for Prostaglandin EP3 receptor (EP3), encoded by the Ptger3 gene, in the loss of functional β-cell mass in Type 2 diabetes (T2D). The primary endogenous EP3 ligand is the arachidonic acid metabolite prostaglandin E2 (PGE2). Expression of the pancreatic islet EP3 and PGE2 synthetic enzymes and/or PGE2 excretion itself have all been shown to be upregulated in primary mouse and human islets isolated from animals or human organ donors with established T2D compared to nondiabetic controls. In this study, we took advantage of a rare and fleeting phenotype in which a subset of Black and Tan BRachyury (BTBR) mice homozygous for the Leptinob/ob mutation—a strong genetic model of T2D—were entirely protected from fasting hyperglycemia even with equal obesity and insulin resistance as their hyperglycemic littermates. Utilizing this model, we found numerous alterations in full-body metabolic parameters in T2D-protected mice (e.g., gut microbiome composition, circulating pancreatic and incretin hormones, and markers of systemic inflammation) that correlate with improvements in EP3-mediated β-cell dysfunction.
Collapse
Affiliation(s)
- Michael D. Schaid
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Yanlong Zhu
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; (Y.Z.); (Y.G.)
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Nicole E. Richardson
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Chinmai Patibandla
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Irene M. Ong
- Department of Obstetrics and Gynecology, University of Wisconsin-Madison, Madison, WI 53715, USA;
- Department of Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Rachel J. Fenske
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Joshua C. Neuman
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Erin Guthery
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Austin Reuter
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Harpreet K. Sandhu
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Miles H. Fuller
- Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA; (M.H.F.); (B.T.L.)
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Elizabeth D. Cox
- Department of Pediatrics, University of Wisconsin-Madison, Madison, WI 53792, USA;
| | - Dawn B. Davis
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Brian T. Layden
- Division of Endocrinology, Diabetes, and Metabolism, University of Illinois at Chicago, Chicago, IL 60612, USA; (M.H.F.); (B.T.L.)
- Jesse Brown Veterans Affairs Medical Center, Chicago, IL 60612, USA
| | - Allan R. Brasier
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Institute for Clinical and Translational Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Dudley W. Lamming
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
| | - Ying Ge
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; (Y.Z.); (Y.G.)
- Human Proteomics Program, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Michelle E. Kimple
- Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705, USA; (M.D.S.); (N.E.R.); (C.P.); (E.G.); (A.R.); (H.K.S.); (D.B.D.); (A.R.B.); (D.W.L.)
- Interdepartmental Graduate Program in Nutritional Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (R.J.F.); (J.C.N.)
- Research Service, William S. Middleton Memorial Veterans Hospital, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA; (Y.Z.); (Y.G.)
- Correspondence: ; Tel.: +1-1-608-265-5627
| |
Collapse
|
147
|
Xu C, Liu X, Shen J, Sun Q, Guo X, Yang M, Leng J. Integrative identification of human serpin PAI-1 inhibitors from Dracaena dragon blood and molecular implications for inhibitor-induced PAI-1 allosterism. Biotechnol Appl Biochem 2021; 69:221-229. [PMID: 33433923 DOI: 10.1002/bab.2100] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2020] [Accepted: 01/06/2021] [Indexed: 11/08/2022]
Abstract
Human plasminogen activator inhibitor-1 (PAI-1) is an important component of the coagulation system and has been recognized as a potential therapeutic target of diverse cardiovascular disorders. Previously, it was found that the extracts from the Chinese medicine Dracaena dragon blood have potent inhibitory activity against PAI-1, but it is unclear which constituents directly participate in the inhibition and how do they regulate PAI-1 at molecular level. Here, we describe an integrated strategy to identify the dragon blood's chemical constituents that can directly target PAI-1. With the strategy, five compounds 1-5 are hit as promising PAI-1 inhibitor candidates, from which three are measured to have high or moderate activity against PAI-1. In particular, the compound 3 is determined to exhibit the highest potency; this value is roughly comparable with the widely used PAI-1 inhibitor Tiplaxtinin. We further examine the molecular effect of compound 3 on PAI-1 conformation at structural level. It is supposed that small-molecule inhibitor regulates the reactive center loop (RCL) of PAI-1 through an allosterism, that is, binding of compound 3 to PAI-1 can allosterically stabilize RCL in latent form, thus promoting PAI-1 conformational conversion from metastable active form to the inactive latent form. Long-term atomistic simulations also demonstrate that removal of compound 3 can destabilize the structured β-stranded conformation of RCL in latent form, although the current simulations are still not sufficient to characterize the full conversion dynamics trajectory.
Collapse
Affiliation(s)
- Chong Xu
- Chongqing Academy of Traditional Chinese Medicine, Chongqing, People's Republic of China.,Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| | - Xia Liu
- Chongqing Academy of Traditional Chinese Medicine, Chongqing, People's Republic of China.,Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| | - Jie Shen
- Chongqing Academy of Traditional Chinese Medicine, Chongqing, People's Republic of China.,Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| | - Quan Sun
- Chongqing Academy of Traditional Chinese Medicine, Chongqing, People's Republic of China.,Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| | - Xiaohong Guo
- Chongqing Academy of Traditional Chinese Medicine, Chongqing, People's Republic of China.,Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| | - Min Yang
- Chongqing Academy of Traditional Chinese Medicine, Chongqing, People's Republic of China.,Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| | - Jing Leng
- Chongqing Academy of Traditional Chinese Medicine, Chongqing, People's Republic of China.,Chongqing Traditional Chinese Medicine Hospital, Chongqing, People's Republic of China
| |
Collapse
|
148
|
Lippi G, Sanchis-Gomar F, Favaloro EJ, Lavie CJ, Henry BM. Coronavirus Disease 2019-Associated Coagulopathy. Mayo Clin Proc 2021; 96:203-217. [PMID: 33413819 PMCID: PMC7604017 DOI: 10.1016/j.mayocp.2020.10.031] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 10/09/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
Patients with the severe form of coronavirus disease 2019 (COVID-19) have been frequently found to suffer from both arterial and venous thrombotic events due to the perpetuation of a hypercoagulable state. This phenomenon, termed COVID-19-associated coagulopathy, is now considered a major component of the pathophysiology of this novel infectious disease, leading to widespread thrombosis. While at first, the vascular insults may be limited to the pulmonary microvasculature, as the disease progresses, systemic involvement occurs, culminating in distant organ thrombosis and multiorgan dysfunction syndrome. In this review article, we discuss recent insights into the pathophysiologic mechanisms of COVID-19-associated coagulopathy and review the clinical, histopathologic, and laboratory evidence, which leads us to conclude that COVID-19 is both a pulmonary and vascular disorder.
Collapse
Key Words
- ace2, angiotensin-converting enzyme 2
- ards, acute respiratory distress syndrome
- cc, coronavirus 2019–associated coagulopathy
- covid-19, coronavirus disease 2019
- dic, disseminated intravascular coagulation
- dvt, deep vein thrombosis
- icu, intensive care unit
- il, interleukin
- ly30, lysis at 30 minutes
- no, nitric oxide
- pai-1, plasminogen activator inhibitor 1
- pe, pulmonary embolism
- sars-cov-2, severe acute respiratory syndrome coronavirus 2
- tf, tissue factor
- tma, thrombotic microangiopathy
- tpa, tissue plasminogen activator
- vte, venous thromboembolism
- vwf, von willebrand factor
Collapse
Affiliation(s)
- Giuseppe Lippi
- Section of Clinical Biochemistry, University of Verona, Verona, Italy
| | - Fabian Sanchis-Gomar
- Department of Physiology, Faculty of Medicine, University of Valencia and INCLIVA Biomedical Research Institute, Valencia, Spain.
| | - Emmanuel J Favaloro
- Haematology, Sydney Centers for Thrombosis and Haemostasis, Institute of Clinical Pathology and Medical Research (ICPMR), NSW Health Pathology, Westmead Hospital, Westmead, NSW, Australia
| | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School - The University of Queensland School of Medicine, New Orleans, LA, USA
| | - Brandon M Henry
- Cardiac Intensive Care Unit, The Heart Institute, Cincinnati Children's Hospital Medical Center, Ohio, USA
| |
Collapse
|
149
|
Okeahialam BN. Serine protease inhibitors could be of benefit in the treatment of COVID-19 disease. Ther Adv Infect Dis 2021; 8:20499361211032048. [PMID: 34345429 PMCID: PMC8283214 DOI: 10.1177/20499361211032048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Affiliation(s)
- Basil N. Okeahialam
- Department of Medicine, Jos University Teaching Hospital, Jos, Plateau 930001, Nigeria
| |
Collapse
|
150
|
Chu Y, Bucci JC, Peterson CB. Dissecting molecular details and functional effects of the high-affinity copper binding site in plasminogen activator Inhibitor-1. Protein Sci 2020; 30:597-612. [PMID: 33345392 DOI: 10.1002/pro.4017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 12/01/2020] [Accepted: 12/15/2020] [Indexed: 11/08/2022]
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is the primary inhibitor for plasminogen activators, tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA). As a unique member in the serine protease inhibitor (serpin) family, PAI-1 is metastable and converts to an inactive, latent structure with a half-life of 1-2 hr under physiological conditions. Unusual effects of metals on the rate of the latency conversion are incompletely understood. Previous work has identified two residues near the N-terminus, H2 and H3, which reside in a high-affinity copper-binding site in PAI-1 [Bucci JC, McClintock CS, Chu Y, Ware GL, McConnell KD, Emerson JP, Peterson CB (2017) J Biol Inorg Chem 22:1123-1,135]. In this study, neighboring residues, H10, E81, and H364, were tested as possible sites that participate in Cu(II) coordination at the high-affinity site. Kinetic methods, gel sensitivity assays, and isothermal titration calorimetry (ITC) revealed that E81 and H364 have different roles in coordinating metal and mediating the stability of PAI-1. H364 provides a third histidine in the metal-coordination sphere with H2 and H3. In contrast, E81 does not appear to be required for metal ligation along with histidines; contacts made by the side-chain carboxylate upon metal binding are perturbed and, in turn, influence dynamic fluctuations within the region encompassing helices D, E, and F and the W86 loop that are important in the pathway for the PAI-1 latency conversion. This investigation underscores a prominent role of protein dynamics, noncovalent bonding networks and ligand binding in controlling the stability of the active form of PAI-1.
Collapse
Affiliation(s)
- Yuzhuo Chu
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Joel C Bucci
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Cynthia B Peterson
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, USA
| |
Collapse
|