151
|
de Oliveira Rech B, Rocha Tenório J, Bertoldi Franco J, Medina JB, Gallottini M, Pérez-Sayáns M, Ortega KL. Risk of bleeding during oral surgery in patients with liver cirrhosis: A systematic review. J Am Dent Assoc 2020; 152:46-54.e2. [PMID: 33250169 DOI: 10.1016/j.adaj.2020.09.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/21/2020] [Accepted: 09/12/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND The liver is responsible for the production of almost all coagulation factors, but does this indicate a risk of developing unusual bleeding in patients with liver cirrhosis during dental surgery? TYPES OF STUDIES REVIEWED In this systemic review, the authors followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses checklist and performed a search of PubMed, Latin American and Caribbean Health Sciences Literature, Web of Science, Scopus, and Cochrane databases. RESULTS Seven studies met the inclusion criteria. The sample sizes ranged from 23 through 318 participants, number of extracted teeth ranged from 62 through 1,183, platelet count ranged from 16,000 through 216,000 per cubic milimeter, and the international normalized ratio was less than 4. The prevalence of hemorrhagic events in the studies ranged from 0% through 8.9%, and almost all were controlled with local hemostatic measures. CONCLUSIONS AND PRACTICAL IMPLICATIONS During dental treatment, patients with liver cirrhosis have a low bleeding risk in spite of the decreased number of platelets and increased international normalized ratio.
Collapse
|
152
|
Davis E, Malig B, Broadwin R, Ebisu K, Basu R, Gold EB, Qi L, Derby CA, Park SK, Wu XM. Association between coarse particulate matter and inflammatory and hemostatic markers in a cohort of midlife women. Environ Health 2020; 19:111. [PMID: 33153486 PMCID: PMC7643259 DOI: 10.1186/s12940-020-00663-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Accepted: 10/12/2020] [Indexed: 05/09/2023]
Abstract
BACKGROUND Exposure to particulate matter air pollution has been associated with cardiovascular disease (CVD) morbidity and mortality; however, most studies have focused on fine particulate matter (PM2.5) exposure and CVD. Coarse particulate matter (PM10-2.5) exposure has not been extensively studied, particularly for long-term exposure, and the biological mechanisms remain uncertain. METHODS We examined the association between ambient concentrations of PM10-2.5 and inflammatory and hemostatic makers that have been linked to CVD. Annual questionnaire and clinical data were obtained from 1694 women (≥ 55 years old in 1999) enrolled in the longitudinal Study of Women's Health Across the Nation (SWAN) at six study sites from 1999 to 2004. Residential locations and the USEPA air monitoring network measurements were used to assign exposure to one-year PM10-2.5, as well as co-pollutants. Linear mixed-effects regression models were used to describe the association between PM10-2.5 exposure and markers, including demographic, health and other covariates. RESULTS Each interquartile (4 μg/m3) increase in one-year PM10-2.5 exposure was associated with a 5.5% (95% confidence interval [CI]: 1.8, 9.4%) increase in levels of plasminogen activator inhibitor-1 (PAI-1) and 4.1% (95% CI: - 0.1, 8.6%) increase in high-sensitivity C-creative Protein (hs-CRP). Stratified analyses suggested that the association with PAI-1 was particularly strong in some subgroups, including women who were peri-menopausal, were less educated, had a body mass index lower than 25, and reported low alcohol consumption. The association between PM10-2.5 and PAI-1 remained unchanged with adjustment for PM2.5, ozone, nitrogen dioxide, and carbon monoxide. CONCLUSIONS Long-term PM10-2.5 exposure may be associated with changes in coagulation independently from PM2.5, and thus, contribute to CVD risk in midlife women.
Collapse
Affiliation(s)
- Emilie Davis
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, 1515 Clay Street, 16th Floor, Oakland, CA, 94612, USA
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT, USA
| | - Brian Malig
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, 1515 Clay Street, 16th Floor, Oakland, CA, 94612, USA
| | - Rachel Broadwin
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, 1515 Clay Street, 16th Floor, Oakland, CA, 94612, USA
| | - Keita Ebisu
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, 1515 Clay Street, 16th Floor, Oakland, CA, 94612, USA
| | - Rupa Basu
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, 1515 Clay Street, 16th Floor, Oakland, CA, 94612, USA
| | - Ellen B Gold
- Department of Public Health Sciences, School of Medicine, University of California, Davis, CA, USA
| | - Lihong Qi
- Department of Public Health Sciences, School of Medicine, University of California, Davis, CA, USA
| | - Carol A Derby
- Department of Neurology, and of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Sung Kyun Park
- Departments of Epidemiology and Environmental Health Sciences, School of of Public Health, University of Michigan, Ann Arbor, MI, USA
| | - Xiangmei May Wu
- Air and Climate Epidemiology Section, Office of Environmental Health Hazard Assessment, California Environmental Protection Agency, 1515 Clay Street, 16th Floor, Oakland, CA, 94612, USA.
| |
Collapse
|
153
|
Spinosa M, Stine JG. Nonalcoholic Fatty Liver Disease-Evidence for a Thrombophilic State? Curr Pharm Des 2020; 26:1036-1044. [PMID: 32003679 DOI: 10.2174/1381612826666200131101553] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023]
Abstract
Nonalcoholic fatty liver disease is the leading cause of liver disease worldwide. It has expansive extrahepatic morbidity and mortality including increased rates of both cardiovascular disease and venous thromboembolism. Derangements in primary, secondary and tertiary hemostasis are found in nonalcoholic fatty liver disease independent of those ascribed to end-stage liver disease. The abnormalities across all stages of hemostasis explain the increased rates of clinically relevant thrombotic events, including pulmonary embolism, deep vein thrombosis and portal vein thrombosis, which on an epidemiologic basis appears to be independent of obesity and other traditional venous thromboembolic risk factors. However, given the complex interaction between obesity, body composition and nonalcoholic fatty liver disease and the potential for exercise to benefit all three, more research is needed to further define the role of each in contributing to the prohemostatic state of nonalcoholic fatty liver disease in order to improve patient oriented outcomes.
Collapse
Affiliation(s)
- Margaret Spinosa
- Department of Medicine, Pennsylvania State University Milton S. Hershey Medical Center, PA 17033, United States
| | - Jonathan G Stine
- Division of Gastroenterology & Hepatology, Department of Medicine, Pennsylvania State University Milton S. Hershey Medical Center, United States.,Department of Public Health Sciences, Pennsylvania State University Milton S. Hershey Medical Center, PA 17033, United States
| |
Collapse
|
154
|
Dastgheib SA, Najafi F, Shajari A, Bahrami R, Asadian F, Sadeghizadeh-Yazdi J, Akbarian E, Emarati SA, Neamatzadeh H. Association of plasminogen activator inhibitor-1 4G5G Polymorphism with risk of diabetic nephropathy and retinopathy: a systematic review and meta-analysis. J Diabetes Metab Disord 2020; 19:2005-2016. [PMID: 33520873 DOI: 10.1007/s40200-020-00675-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Revised: 10/20/2020] [Accepted: 10/27/2020] [Indexed: 12/16/2022]
Abstract
Background The 4G5G polymorphism of Plasminogen activator inhibitor-1 (PAI-1) gene is reported to be associated with diabetes nephropathy and retinopathy (DNR) risk. However, the findings are conflicting. Herein, we conducted a case-control and meta-analysis study to explore the association of PAI-1 4G5G polymorphism with risk of DNR. Methods We retrieved PubMed, EMBASE, Web of Knowledge, and CNKI databases and screened eligible studies up to August 15, 2020. The strength of associations was assessed by odd ratio (OR) and the corresponding 95% confidence interval (95% CI). Results A total of 27 case-control studies including 16 studies with 1,825 cases case and 1,731 controls on DN and eleven studies with 1,397 cases and 1,545 controls on DR were selected. Pooled data showed that the PAI-1 4G5G polymorphism was significantly associated with DN (allele model: OR = 0.674, 95% CI 0.524-0.865, p = 0.002; homozygote model: OR = 0.536, 95% CI 0.351-0.817, p = 0.004; heterozygote model: OR = 0.621, 95% CI 0.427-0.903, p = 0.013; dominant model: OR = 0.575, 95% CI 0.399-0.831, p = 0.003; and recessive model: OR = 0.711, 95% CI 0.515-0.981, p = 0.038) and DR (homozygote model: OR = 0.770, 95% CI 0.621-0.955, p = 0.0.017) risk. Stratified analyses by ethnicity indicated that PAI-1 4G5G polymorphism was associated with DN and DR risk in Asians and Caucasians, respectively. Conclusions The present meta-analysis revealed that the PAI-1 4G5G polymorphism was associated with increased risk of DN and DR risk. However, well-designed large-scale clinical studies are required to further validate our results.
Collapse
Affiliation(s)
- Seyed Alireza Dastgheib
- Department of Medical Genetics, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Farzaneh Najafi
- Department of Internal Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Ahmad Shajari
- Department of Pediatrics, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Reza Bahrami
- Neonatal Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Asadian
- Department of Medical Laboratory Sciences, School of Paramedical Science, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Jalal Sadeghizadeh-Yazdi
- Department of Food Science and Technology, School of Public Health, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Elahe Akbarian
- Children Growth Disorder Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Seyed Alireza Emarati
- Children Growth Disorder Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Hossein Neamatzadeh
- Department of Medical Genetics, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Mother and Newborn Health Research Center, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
155
|
Jin R, Zhong W, Liu S, Wang M, Li G. Inhibition of PI3Kγ by AS605240 plus low-dose tissue plasminogen activator (tPA) combination improves thrombolytic therapy in a rat model of embolic stroke. Neurosci Lett 2020; 738:135339. [PMID: 32882317 PMCID: PMC8171661 DOI: 10.1016/j.neulet.2020.135339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 08/10/2020] [Accepted: 08/27/2020] [Indexed: 11/24/2022]
Abstract
Our previous study showed that PI3Kγ inhibition with AS605240 plus a standard rat-dose tPA (10 mg/kg) combination attenuates delayed tPA-induced brain hemorrhage and ameliorates acute stroke injury 3 days after ischemic stroke in rats. The purpose of this study was to investigate whether combining AS605240 with tPA can enhance thrombolytic efficacy, so that lower doses of tPA can be applied to improve long-term outcome after ischemic stroke. The results showed that AS605240 plus low-dose tPA (5 mg/kg) combination therapy at 4 h after stroke onset significantly reduced infarct volume and neurological deficits at 24 h after stroke compared with saline, AS605240 or low-dose tPA alone group. Importantly, the combination therapy significantly reduced the delayed tPA-associated brain hemorrhage. Moreover, the combination therapy significantly decreased the size of the residual embolus within the middle cerebral artery, which was associated with a decrease in plasma plasminogen activator inhibitor-1 (PAI-1) activity compared with saline and tPA alone. Finally, AS605240 plus low-dose tPA combination improved long-term outcome for at least 35 days after stroke compared with the saline-treated group. Taken together, these findings suggest that PI3Kγ inhibition with AS605240 might act as an adjunct approach for enhancing tPA thrombolytic efficacy in acute ischemic stroke.
Collapse
Affiliation(s)
- Rong Jin
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| | - Wei Zhong
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Shan Liu
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Min Wang
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Guohong Li
- Department of Neurosurgery, the Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA.
| |
Collapse
|
156
|
Berretta AA, Silveira MAD, Cóndor Capcha JM, De Jong D. Propolis and its potential against SARS-CoV-2 infection mechanisms and COVID-19 disease: Running title: Propolis against SARS-CoV-2 infection and COVID-19. Biomed Pharmacother 2020; 131:110622. [PMID: 32890967 PMCID: PMC7430291 DOI: 10.1016/j.biopha.2020.110622] [Citation(s) in RCA: 135] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 08/04/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022] Open
Abstract
Propolis, a resinous material produced by honey bees from plant exudates, has long been used in traditional herbal medicine and is widely consumed as a health aid and immune system booster. The COVID-19 pandemic has renewed interest in propolis products worldwide; fortunately, various aspects of the SARS-CoV-2 infection mechanism are potential targets for propolis compounds. SARS-CoV-2 entry into host cells is characterized by viral spike protein interaction with cellular angiotensin-converting enzyme 2 (ACE2) and serine protease TMPRSS2. This mechanism involves PAK1 overexpression, which is a kinase that mediates coronavirus-induced lung inflammation, fibrosis, and immune system suppression. Propolis components have inhibitory effects on the ACE2, TMPRSS2 and PAK1 signaling pathways; in addition, antiviral activity has been proven in vitro and in vivo. In pre-clinical studies, propolis promoted immunoregulation of pro-inflammatory cytokines, including reduction in IL-6, IL-1 beta and TNF-α. This immunoregulation involves monocytes and macrophages, as well as Jak2/STAT3, NF-kB, and inflammasome pathways, reducing the risk of cytokine storm syndrome, a major mortality factor in advanced COVID-19 disease. Propolis has also shown promise as an aid in the treatment of various of the comorbidities that are particularly dangerous in COVID-19 patients, including respiratory diseases, hypertension, diabetes, and cancer. Standardized propolis products with consistent bioactive properties are now available. Given the current emergency caused by the COVID-19 pandemic and limited therapeutic options, propolis is presented as a promising and relevant therapeutic option that is safe, easy to administrate orally and is readily available as a natural supplement and functional food.
Collapse
Affiliation(s)
- Andresa Aparecida Berretta
- Research, Development and Innovation Department, Apis Flora Indl. Coml. Ltda, Ribeirão Preto, São Paulo, Brazil.
| | | | - José Manuel Cóndor Capcha
- Interdisciplinary Stem Cell Institute at Miller School of Medicine, University of Miami, Miami, Florida, United States.
| | - David De Jong
- Genetics Department, Ribeirão Preto School of Medicine, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| |
Collapse
|
157
|
Recinella L, Orlando G, Ferrante C, Chiavaroli A, Brunetti L, Leone S. Adipokines: New Potential Therapeutic Target for Obesity and Metabolic, Rheumatic, and Cardiovascular Diseases. Front Physiol 2020; 11:578966. [PMID: 33192583 PMCID: PMC7662468 DOI: 10.3389/fphys.2020.578966] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/14/2020] [Indexed: 12/11/2022] Open
Abstract
Besides its role as an energy storage organ, adipose tissue can be viewed as a dynamic and complex endocrine organ, which produces and secretes several adipokines, including hormones, cytokines, extracellular matrix (ECM) proteins, and growth and vasoactive factors. A wide body of evidence showed that adipokines play a critical role in various biological and physiological functions, among which feeding modulation, inflammatory and immune function, glucose and lipid metabolism, and blood pressure control. The aim of this review is to summarize the effects of several adipokines, including leptin, diponectin, resistin, chemerin, lipocalin-2 (LCN2), vaspin, omentin, follistatin-like 1 (FSTL1), secreted protein acidic and rich in cysteine (SPARC), secreted frizzled-related protein 5 (SFRP5), C1q/TNF-related proteins (CTRPs), family with sequence similarity to 19 member A5 (FAM19A5), wingless-type inducible signaling pathway protein-1 (WISP1), progranulin (PGRN), nesfatin-1 (nesfatin), visfatin/PBEF/NAMPT, apelin, retinol binding protein 4 (RPB4), and plasminogen activator inhibitor-1 (PAI-1) in the regulation of insulin resistance and vascular function, as well as many aspects of inflammation and immunity and their potential role in managing obesity-associated diseases, including metabolic, osteoarticular, and cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | | | - Luigi Brunetti
- Department of Pharmacy, Gabriele d’Annunzio University, Chieti, Italy
| | | |
Collapse
|
158
|
Zwaans BMM, Nicolai HE, Chancellor MB, Lamb LE. Prostate cancer survivors with symptoms of radiation cystitis have elevated fibrotic and vascular proteins in urine. PLoS One 2020; 15:e0241388. [PMID: 33119677 PMCID: PMC7595289 DOI: 10.1371/journal.pone.0241388] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/13/2020] [Indexed: 01/22/2023] Open
Abstract
Radiation for pelvic cancers can result in severe bladder damage and radiation cystitis (RC), which is characterized by chronic inflammation, fibrosis, and vascular damage. RC development is poorly understood because bladder biopsies are difficult to obtain. The goal of this study is to gain understanding of molecular changes that drive radiation-induced cystitis in cancer survivors using urine samples from prostate cancer survivors with history of radiation therapy. 94 urine samples were collected from prostate cancer survivors with (n = 85) and without (n = 9) history of radiation therapy. 15 patients with radiation history were officially diagnosed with radiation cystitis. Levels of 47 different proteins were measured using Multiplex Luminex. Comparisons were made between non-irradiated and irradiated samples, and within irradiated samples based on radiation cystitis diagnosis, symptom scores or hematuria. Statistical analysis was performed using Welch’s t-test. In prostate cancer survivors with history of radiation therapy, elevated levels of PAI 1, TIMP1, TIMP2, HGF and VEGF-A were detected in patients that received a radiation cystitis diagnosis. These proteins were also increased in patients suffering from hematuria or high symptom scores. No inflammatory proteins were detected in the urine, except in patients with gross hematuria and end stage radiation cystitis. Active fibrosis and vascular distress is detectable in the urine through elevated levels of associated proteins. Inflammation is only detected in urine of patients with end-stage radiation cystitis disease. These results suggest that fibrosis and vascular damage drive the development of radiation cystitis and could lead to the development of more targeted treatments.
Collapse
Affiliation(s)
- Bernadette M. M. Zwaans
- Department of Urology, William Beaumont Hospital, Royal Oak, Michigan, United States of America
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
| | - Heinz E. Nicolai
- Departamento de Urología, Universidad de Chile, Santiago, Chile
- Hospital Clínico San Borja Arriarán, Santiago, Chile
| | - Michael B. Chancellor
- Department of Urology, William Beaumont Hospital, Royal Oak, Michigan, United States of America
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
| | - Laura E. Lamb
- Department of Urology, William Beaumont Hospital, Royal Oak, Michigan, United States of America
- Oakland University William Beaumont School of Medicine, Rochester, Michigan, United States of America
- * E-mail:
| |
Collapse
|
159
|
Borchert C, Herman A, Roth M, Brooks AC, Friedenberg SG. RNA sequencing of whole blood in dogs with primary immune-mediated hemolytic anemia (IMHA) reveals novel insights into disease pathogenesis. PLoS One 2020; 15:e0240975. [PMID: 33091028 PMCID: PMC7580939 DOI: 10.1371/journal.pone.0240975] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 10/06/2020] [Indexed: 11/29/2022] Open
Abstract
Immune-mediated hemolytic anemia (IMHA) is a life-threatening autoimmune disorder characterized by a self-mediated attack on circulating red blood cells. The disease occurs naturally in both dogs and humans, but is significantly more prevalent in dogs. Because of its shared features across species, dogs offer a naturally occurring model for studying IMHA in people. In this study, we used RNA sequencing of whole blood from treatment-naïve dogs to study transcriptome-wide changes in gene expression in newly diagnosed animals compared to healthy controls. We found many overexpressed genes in pathways related to neutrophil function, coagulation, and hematopoiesis. In particular, the most highly overexpressed gene in cases was a phospholipase scramblase, which mediates the externalization of phosphatidylserine from the inner to the outer leaflet of cell membranes. This family of genes has been shown to be critically important for programmed cell death of erythrocytes as well as the initiation of the clotting cascade. Unexpectedly, we found marked underexpression of many genes related to lymphocyte function. We also identified groups of genes that are highly associated with the inflammatory response and red blood cell regeneration in affected dogs. We did not find any genes that distinguished dogs that lived vs. those that died at 30 days following diagnosis, nor did we find any relevant genomic signatures of microbial organisms in the blood of affected animals. Future studies are warranted to validate these findings and assess their implication in developing novel therapeutic approaches for dogs and humans with IMHA.
Collapse
Affiliation(s)
- Corie Borchert
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Adam Herman
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, Minnesota, United States of America
| | - Megan Roth
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| | - Aimee C. Brooks
- Department of Veterinary Clinical Sciences, Purdue University College of Veterinary Medicine, West Lafayette, Indiana, United States of America
| | - Steven G. Friedenberg
- Department of Veterinary Clinical Sciences, University of Minnesota College of Veterinary Medicine, St. Paul, Minnesota, United States of America
| |
Collapse
|
160
|
Deng P, Wang C, Wahlang B, Sexton T, Morris AJ, Hennig B. Co-exposure to PCB126 and PFOS increases biomarkers associated with cardiovascular disease risk and liver injury in mice. Toxicol Appl Pharmacol 2020; 409:115301. [PMID: 33096110 DOI: 10.1016/j.taap.2020.115301] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/08/2020] [Accepted: 10/18/2020] [Indexed: 02/07/2023]
Abstract
Polychlorinated biphenyl (PCB)126 and perfluorooctane sulfonic acid (PFOS) are halogenated organic pollutants of high concern. Exposure to these chemicals is ubiquitous, and can lead to potential synergistic adverse effects in individuals exposed to both classes of chemicals. The present study was designed to identify interactions between PCB126 and PFOS that might promote acute changes in inflammatory pathways associated with cardiovascular disease and liver injury. Male C57BL/6 mice were exposed to vehicle, PCB126, PFOS, or a mixture of both pollutants. Plasma and liver samples were collected at 48 h after exposure. Changes in the expression of hepatic genes involved in oxidative stress, inflammation, and atherosclerosis were investigated. Plasma and liver samples was analyzed using untargeted lipidomic method. Hepatic mRNA levels for Nqo1, Icam1, and PAI1 were significantly increased in the mixture-exposed mice. Plasma levels of PAI1, a marker of fibrosis and thrombosis, were also significantly elevated in the mixture-exposed group. Liver injury was observed only in the mixture-exposed mice. Lipidomic analysis revealed that co-exposure to the mixture enhanced hepatic lipid accumulation and elevated oxidized phospholipids levels. In summary, this study shows that acute co-exposure to PCB126 and PFOS in mice results in liver injury and increased cardiovascular disease risk.
Collapse
Affiliation(s)
- Pan Deng
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Chunyan Wang
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA
| | - Banrida Wahlang
- Superfund Research Center, University of Louisville, Louisville, KY 40202, USA
| | - Travis Sexton
- Division of Cardiovascular Medicine, The Gill Heart and Vascular Institute, College of Medicine, University of Kentucky, and Lexington Veterans Affairs Medical Center, Lexington, KY, 40536, USA
| | - Andrew J Morris
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Division of Cardiovascular Medicine, The Gill Heart and Vascular Institute, College of Medicine, University of Kentucky, and Lexington Veterans Affairs Medical Center, Lexington, KY, 40536, USA
| | - Bernhard Hennig
- Superfund Research Center, University of Kentucky, Lexington, KY 40536, USA; Department of Animal and Food Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
161
|
Stress-induced RNA-chromatin interactions promote endothelial dysfunction. Nat Commun 2020; 11:5211. [PMID: 33060583 PMCID: PMC7566596 DOI: 10.1038/s41467-020-18957-w] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/11/2020] [Indexed: 01/06/2023] Open
Abstract
Chromatin-associated RNA (caRNA) has been proposed as a type of epigenomic modifier. Here, we test whether environmental stress can induce cellular dysfunction through modulating RNA-chromatin interactions. We induce endothelial cell (EC) dysfunction with high glucose and TNFα (H + T), that mimic the common stress in diabetes mellitus. We characterize the H + T-induced changes in gene expression by single cell (sc)RNA-seq, DNA interactions by Hi-C, and RNA-chromatin interactions by iMARGI. H + T induce inter-chromosomal RNA-chromatin interactions, particularly among the super enhancers. To test the causal relationship between H + T-induced RNA-chromatin interactions and the expression of EC dysfunction-related genes, we suppress the LINC00607 RNA. This suppression attenuates the expression of SERPINE1, a critical pro-inflammatory and pro-fibrotic gene. Furthermore, the changes of the co-expression gene network between diabetic and healthy donor-derived ECs corroborate the H + T-induced RNA-chromatin interactions. Taken together, caRNA-mediated dysregulation of gene expression modulates EC dysfunction, a crucial mechanism underlying numerous diseases. Global interaction of chromatin-associated RNAs and DNA can be identified in situ. Here the authors report the genome-wide increase of interchromosomal RNA-DNA interactions and demonstrate the importance of such RNA-DNA contacts exemplified by LINC00607 RNA and SERPINE1 gene’s super enhancer in dysfunctional endothelial cell models.
Collapse
|
162
|
Matsuyama T, Kubli SP, Yoshinaga SK, Pfeffer K, Mak TW. An aberrant STAT pathway is central to COVID-19. Cell Death Differ 2020. [PMID: 33037393 DOI: 10.1038/s41418‐020‐00633‐7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
COVID-19 is caused by SARS-CoV-2 infection and characterized by diverse clinical symptoms. Type I interferon (IFN-I) production is impaired and severe cases lead to ARDS and widespread coagulopathy. We propose that COVID-19 pathophysiology is initiated by SARS-CoV-2 gene products, the NSP1 and ORF6 proteins, leading to a catastrophic cascade of failures. These viral components induce signal transducer and activator of transcription 1 (STAT1) dysfunction and compensatory hyperactivation of STAT3. In SARS-CoV-2-infected cells, a positive feedback loop established between STAT3 and plasminogen activator inhibitor-1 (PAI-1) may lead to an escalating cycle of activation in common with the interdependent signaling networks affected in COVID-19. Specifically, PAI-1 upregulation leads to coagulopathy characterized by intravascular thrombi. Overproduced PAI-1 binds to TLR4 on macrophages, inducing the secretion of proinflammatory cytokines and chemokines. The recruitment and subsequent activation of innate immune cells within an infected lung drives the destruction of lung architecture, which leads to the infection of regional endothelial cells and produces a hypoxic environment that further stimulates PAI-1 production. Acute lung injury also activates EGFR and leads to the phosphorylation of STAT3. COVID-19 patients' autopsies frequently exhibit diffuse alveolar damage (DAD) and increased hyaluronan (HA) production which also leads to higher levels of PAI-1. COVID-19 risk factors are consistent with this scenario, as PAI-1 levels are increased in hypertension, obesity, diabetes, cardiovascular diseases, and old age. We discuss the possibility of using various approved drugs, or drugs currently in clinical development, to treat COVID-19. This perspective suggests to enhance STAT1 activity and/or inhibit STAT3 functions for COVID-19 treatment. This might derail the escalating STAT3/PAI-1 cycle central to COVID-19.
Collapse
Affiliation(s)
- Toshifumi Matsuyama
- Department of Pathology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Shawn P Kubli
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2M9, Canada
| | | | - Klaus Pfeffer
- Institute of Medical Microbiology and Hospital Hygiene, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Tak W Mak
- Princess Margaret Cancer Centre, University Health Network, 610 University Avenue, Toronto, ON, M5G 2M9, Canada. .,Department of Medical Biophysics and Department of Immunology, University of Toronto, 101 College Street, Toronto, ON, M5G 1L7, Canada. .,Department of Medicine, University of Hong Kong, Pok Fu Lam, 999077, Hong Kong.
| |
Collapse
|
163
|
An aberrant STAT pathway is central to COVID-19. Cell Death Differ 2020; 27:3209-3225. [PMID: 33037393 PMCID: PMC7545020 DOI: 10.1038/s41418-020-00633-7] [Citation(s) in RCA: 210] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/20/2020] [Accepted: 09/24/2020] [Indexed: 02/07/2023] Open
Abstract
COVID-19 is caused by SARS-CoV-2 infection and characterized by diverse clinical symptoms. Type I interferon (IFN-I) production is impaired and severe cases lead to ARDS and widespread coagulopathy. We propose that COVID-19 pathophysiology is initiated by SARS-CoV-2 gene products, the NSP1 and ORF6 proteins, leading to a catastrophic cascade of failures. These viral components induce signal transducer and activator of transcription 1 (STAT1) dysfunction and compensatory hyperactivation of STAT3. In SARS-CoV-2-infected cells, a positive feedback loop established between STAT3 and plasminogen activator inhibitor-1 (PAI-1) may lead to an escalating cycle of activation in common with the interdependent signaling networks affected in COVID-19. Specifically, PAI-1 upregulation leads to coagulopathy characterized by intravascular thrombi. Overproduced PAI-1 binds to TLR4 on macrophages, inducing the secretion of proinflammatory cytokines and chemokines. The recruitment and subsequent activation of innate immune cells within an infected lung drives the destruction of lung architecture, which leads to the infection of regional endothelial cells and produces a hypoxic environment that further stimulates PAI-1 production. Acute lung injury also activates EGFR and leads to the phosphorylation of STAT3. COVID-19 patients' autopsies frequently exhibit diffuse alveolar damage (DAD) and increased hyaluronan (HA) production which also leads to higher levels of PAI-1. COVID-19 risk factors are consistent with this scenario, as PAI-1 levels are increased in hypertension, obesity, diabetes, cardiovascular diseases, and old age. We discuss the possibility of using various approved drugs, or drugs currently in clinical development, to treat COVID-19. This perspective suggests to enhance STAT1 activity and/or inhibit STAT3 functions for COVID-19 treatment. This might derail the escalating STAT3/PAI-1 cycle central to COVID-19.
Collapse
|
164
|
Çevikelli-Yakut ZA, Özçelik R, Çevik Ö, Şener TE, Şener G. Exercise and caloric restriction improve cardiovascular and erectile function in rats with metabolic syndrome. Int J Impot Res 2020; 33:844-853. [PMID: 33009496 DOI: 10.1038/s41443-020-00356-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 09/03/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
The aim of this study is to examine the possible benefits of exercise and caloric restriction (CR) on cardiovascular hemodynamics, erectile function, and antioxidant system in metabolic syndrome (MS). Sixty male Spraque-Dawley rats were divided into five groups; control, MS, MS + CR, MS + exercise (EXC), and MS + CR + EXC. To induce MS, 10% fructose solution was applied for 3 months. Thereafter, in CR groups calorie was restricted 40% and in EXC groups swimming was performed for 6 weeks. Body weight, blood glucose, and blood pressure (BP) levels were measured before and after MS induction and at the end of the experiment. After decapitation, tumor necrosis factor (TNF)-α, adiponectin (ADP), and plasminogen activator inhibitor (PAI)-1 levels were investigated in blood, oxidative stress parameters were examined in heart, aorta, and corpus cavernosum (CC) tissues. Isometric contraction in isolated tissue bath was studied in aorta and CC tissues. Animals subjected to exercise and CR had decreased BP and blood glucose levels. Impaired contraction-relaxation responses in MS group were improved with exercise and CR. MS-induced increase in TNF-α, PAI-1, malondialdehyde (MDA), and decrease in ADP, glutathione (GSH), and superoxide dismutase (SOD) were normalized with exercise and CR. Exercise and CR may be beneficial against changes in cardiovascular hemodynamics caused by MS.
Collapse
Affiliation(s)
- Zatiye Ayça Çevikelli-Yakut
- Department of Pharmacology, School of Pharmacy, Marmara University, İstanbul, Turkey.,Department of Pharmacognosy, School of Pharmacy, Trakya University, Edirne, Turkey
| | - Reyhan Özçelik
- Department of Pharmacology, School of Pharmacy, Marmara University, İstanbul, Turkey
| | - Özge Çevik
- Department of Biochemistry, School of Medicine, Aydın Adnan Menderes University, Aydın, Turkey
| | - Tarık Emre Şener
- Department of Urology, School of Medicine, Marmara University, İstanbul, Turkey
| | - Göksel Şener
- Department of Pharmacology, School of Pharmacy, Marmara University, İstanbul, Turkey.
| |
Collapse
|
165
|
Robison LS, Gannon OJ, Thomas MA, Salinero AE, Abi-Ghanem C, Poitelon Y, Belin S, Zuloaga KL. Role of sex and high-fat diet in metabolic and hypothalamic disturbances in the 3xTg-AD mouse model of Alzheimer's disease. J Neuroinflammation 2020; 17:285. [PMID: 32993686 PMCID: PMC7526387 DOI: 10.1186/s12974-020-01956-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 09/18/2020] [Indexed: 12/14/2022] Open
Abstract
Background Hypothalamic dysfunction occurs early in the clinical course of Alzheimer’s disease (AD), likely contributing to disturbances in feeding behavior and metabolic function that are often observed years prior to the onset of cognitive symptoms. Late-life weight loss and low BMI are associated with increased risk of dementia and faster progression of disease. However, high-fat diet and metabolic disease (e.g., obesity, type 2 diabetes), particularly in mid-life, are associated with increased risk of AD, as well as exacerbated AD pathology and behavioral deficits in animal models. In the current study, we explored possible relationships between hypothalamic function, diet/metabolic status, and AD. Considering the sex bias in AD, with women representing two-thirds of AD patients, we sought to determine whether these relationships vary by sex. Methods WT and 3xTg-AD male and female mice were fed a control (10% fat) or high-fat (HF 60% fat) diet from ~ 3–7 months of age, then tested for metabolic and hypothalamic disturbances. Results On control diet, male 3xTg-AD mice displayed decreased body weight, reduced fat mass, hypoleptinemia, and mild systemic inflammation, as well as increased expression of gliosis- and inflammation-related genes in the hypothalamus (Iba1, GFAP, TNF-α, IL-1β). In contrast, female 3xTg-AD mice on control diet displayed metabolic disturbances opposite that of 3xTg-AD males (increased body and fat mass, impaired glucose tolerance). HF diet resulted in expected metabolic alterations across groups (increased body and fat mass; glucose intolerance; increased plasma insulin and leptin, decreased ghrelin; nonalcoholic fatty liver disease-related pathology). HF diet resulted in the greatest weight gain, adiposity, and glucose intolerance in 3xTg-AD females, which were associated with markedly increased hypothalamic expression of GFAP and IL-1β, as well as GFAP labeling in several hypothalamic nuclei that regulate energy balance. In contrast, HF diet increased diabetes markers and systemic inflammation preferentially in AD males but did not exacerbate hypothalamic inflammation in this group. Conclusions These findings provide further evidence for the roles of hypothalamic and metabolic dysfunction in AD, which in the 3xTg-AD mouse model appears to be dependent on both sex and diet.
Collapse
Affiliation(s)
- Lisa S Robison
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Olivia J Gannon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Melissa A Thomas
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Abigail E Salinero
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Charly Abi-Ghanem
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Yannick Poitelon
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Sophie Belin
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA
| | - Kristen L Zuloaga
- Department of Neuroscience & Experimental Therapeutics, Albany Medical College, 47 New Scotland Avenue, MC-136, Albany, NY, 12208, USA.
| |
Collapse
|
166
|
PAI-1, the Plasminogen System, and Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21197066. [PMID: 32993026 PMCID: PMC7582753 DOI: 10.3390/ijms21197066] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 09/21/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022] Open
Abstract
The plasminogen system is a critical proteolytic system responsible for the remodeling of the extracellular matrix (ECM). The master regulator of the plasminogen system, plasminogen activator inhibitor-1 (PAI-1), has been implicated for its role in exacerbating various disease states not only through the accumulation of ECM (i.e., fibrosis) but also its role in altering cell fate/behaviour. Examination of PAI-1 has extended through various tissues and cell-types with recent investigations showing its presence in skeletal muscle. In skeletal muscle, the role of this protein has been implicated throughout the regeneration process, and in skeletal muscle pathologies (muscular dystrophy, diabetes, and aging-driven pathology). Needless to say, the complete function of this protein in skeletal muscle has yet to be fully elucidated. Given the importance of skeletal muscle in maintaining overall health and quality of life, it is critical to understand the alterations—particularly in PAI-1—that occur to negatively impact this organ. Thus, we provide a comprehensive review of the importance of PAI-1 in skeletal muscle health and function. We aim to shed light on the relevance of this protein in skeletal muscle and propose potential therapeutic approaches to aid in the maintenance of skeletal muscle health.
Collapse
|
167
|
Tahtamouni LH, Hamdan MN, Al-Mazaydeh ZA, Bawadi RM, Rammaha MS, Zghoul AM, Ahram MA, Yasin SR. Alu-repeat polymorphism in the tissue plasminogen activator ( t-PA) gene, seminal t-PA concentration, and male fertility impairment: A case-control study. Int J Reprod Biomed 2020; 18:571-578. [PMID: 32930675 PMCID: PMC7457157 DOI: 10.18502/ijrm.v13i8.7496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/09/2019] [Accepted: 01/14/2020] [Indexed: 11/24/2022] Open
Abstract
Background Tissue plasminogen activator (t-PA) is a protein involved in the fibrinolytic system that catalyzes the conversion of plasminogen into the active plasmin. The activity of t-PA is controlled by plasminogen activator inhibitor-1. t-PA has crucial functions during spermatogenesis. One polymorphism was reported for t-PA gene, either the presence of a 300-bp Alu-repeat (Alu + ) or its absence (Alu - ). Objective The current work aimed at studying the association between Alu polymorphism in the t-PA gene and male infertility. Materials and Methods Using polymerase chain reaction on genomic DNA isolated from the blood of 79 participants, a region polymorphic for Alu element insertion in t-PA gene was amplified. In addition, total t-PA concentration, plasminogen activator inhibitor-1 /t-PA complex concentration, and t-PA activity in seminal plasma were measured by enzyme-linked immunosorbent assay. Results The results indicate that the percentage of infertile participants (n = 50) who were homozygous for t-PA Alu insertion (Alu + / + ), heterozygous Alu + / - or homozygous for t-PA Alu deletion (Alu - / - ) did not change significantly (p = 0.43, 0.81, and 0.85, respectively) when compared with the control participants (n = 29). On the other hand, a significant decrease (p = 0.0001) of t-PA total concentration in seminal plasma was observed in the infertile group in comparison with the control group. However, the results indicate that there is no association between the t-PA Alu different genotypes and the total t-PA seminal concentration in the infertile group when compared to the control group (p = 0.63). Conclusion Data obtained from the current study does not support an association between t-PA Alu polymorphism and t-PA seminal concentration or male infertility.
Collapse
Affiliation(s)
- Lubna Hamid Tahtamouni
- Department of Biology and Biotechnology, Faculty of Science, the Hashemite University, Zarqa, Jordan.,Department of Biochemistry and Molecular Biology, College of Natural Sciences, Colorado State University, Fort Collins, Colorado, USA
| | - Mahmoud Nael Hamdan
- Department of Biology and Biotechnology, Faculty of Science, the Hashemite University, Zarqa, Jordan
| | - Zainab Ali Al-Mazaydeh
- Department of Biology and Biotechnology, Faculty of Science, the Hashemite University, Zarqa, Jordan
| | - Randa Mahmoud Bawadi
- Department of Physiology and Biochemistry, School of Medicine, the University of Jordan, Amman, Jordan
| | - Majdoleen Sobhi Rammaha
- Department of Biology and Biotechnology, Faculty of Science, the Hashemite University, Zarqa, Jordan
| | - Ahmad Mohammad Zghoul
- Department of Biology and Biotechnology, Faculty of Science, the Hashemite University, Zarqa, Jordan
| | - Mamoun Ahmad Ahram
- Department of Physiology and Biochemistry, School of Medicine, the University of Jordan, Amman, Jordan
| | - Salem Refat Yasin
- Department of Biology and Biotechnology, Faculty of Science, the Hashemite University, Zarqa, Jordan
| |
Collapse
|
168
|
Vives-Usano M, Hernandez-Ferrer C, Maitre L, Ruiz-Arenas C, Andrusaityte S, Borràs E, Carracedo Á, Casas M, Chatzi L, Coen M, Estivill X, González JR, Grazuleviciene R, Gutzkow KB, Keun HC, Lau CHE, Cadiou S, Lepeule J, Mason D, Quintela I, Robinson O, Sabidó E, Santorelli G, Schwarze PE, Siskos AP, Slama R, Vafeiadi M, Martí E, Vrijheid M, Bustamante M. In utero and childhood exposure to tobacco smoke and multi-layer molecular signatures in children. BMC Med 2020; 18:243. [PMID: 32811491 PMCID: PMC7437049 DOI: 10.1186/s12916-020-01686-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 06/29/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The adverse health effects of early life exposure to tobacco smoking have been widely reported. In spite of this, the underlying molecular mechanisms of in utero and postnatal exposure to tobacco smoke are only partially understood. Here, we aimed to identify multi-layer molecular signatures associated with exposure to tobacco smoke in these two exposure windows. METHODS We investigated the associations of maternal smoking during pregnancy and childhood secondhand smoke (SHS) exposure with molecular features measured in 1203 European children (mean age 8.1 years) from the Human Early Life Exposome (HELIX) project. Molecular features, covering 4 layers, included blood DNA methylation and gene and miRNA transcription, plasma proteins, and sera and urinary metabolites. RESULTS Maternal smoking during pregnancy was associated with DNA methylation changes at 18 loci in child blood. DNA methylation at 5 of these loci was related to expression of the nearby genes. However, the expression of these genes themselves was only weakly associated with maternal smoking. Conversely, childhood SHS was not associated with blood DNA methylation or transcription patterns, but with reduced levels of several serum metabolites and with increased plasma PAI1 (plasminogen activator inhibitor-1), a protein that inhibits fibrinolysis. Some of the in utero and childhood smoking-related molecular marks showed dose-response trends, with stronger effects with higher dose or longer duration of the exposure. CONCLUSION In this first study covering multi-layer molecular features, pregnancy and childhood exposure to tobacco smoke were associated with distinct molecular phenotypes in children. The persistent and dose-dependent changes in the methylome make CpGs good candidates to develop biomarkers of past exposure. Moreover, compared to methylation, the weak association of maternal smoking in pregnancy with gene expression suggests different reversal rates and a methylation-based memory to past exposures. Finally, certain metabolites and protein markers evidenced potential early biological effects of postnatal SHS, such as fibrinolysis.
Collapse
Affiliation(s)
- Marta Vives-Usano
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Carles Hernandez-Ferrer
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Léa Maitre
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Carlos Ruiz-Arenas
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Sandra Andrusaityte
- Department of Environmental Sciences, Vytautas Magnus University, K. Donelaicio Street 58, 44248, Kaunas, Lithuania
| | - Eva Borràs
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Ángel Carracedo
- Grupo de Medicina Xenómica, Fundación Pública Galega de Medicina Xenómica, Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), SERGAS, Rúa Choupana s/n, 15706, Santiago de Compostela, Spain
- Centro de Investigación en Red de Enfermedades Raras (CIBERER) y Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII), Universidade de Santiago de Compostela, Praza do Obradoiro s/n, 15782, Santiago de Compostela, Spain
| | - Maribel Casas
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Leda Chatzi
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, 1540 Alcazar Street, Los Angeles, 90033, USA
| | - Muireann Coen
- Oncology Safety, Clinical Pharmacology and Safety Sciences, R&D Biopharmaceuticals, AstraZeneca, 1 Francis Crick Avenue, Cambridge, CB2 0RE, UK
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Xavier Estivill
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Quantitative Genomics Medicine Laboratories (qGenomics), Esplugues del Llobregat, Barcelona, Catalonia, Spain
| | - Juan R González
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Regina Grazuleviciene
- Department of Environmental Sciences, Vytautas Magnus University, K. Donelaicio Street 58, 44248, Kaunas, Lithuania
| | - Kristine B Gutzkow
- Department af Environmental Health, Norwegian Institute of Public Health, Lovisenberggt 6, 0456, Oslo, Norway
| | - Hector C Keun
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
- Cancer Metabolism and Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Chung-Ho E Lau
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Solène Cadiou
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, 38000, Grenoble, France
| | - Johanna Lepeule
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, 38000, Grenoble, France
| | - Dan Mason
- Bradford Institute for Health Research, Bradford Royal Infirmary, Bradford, BD9 6RJ, UK
| | - Inés Quintela
- Grupo de Medicina Xenómica, Centro Nacional de Genotipado (CEGEN-PRB3-ISCIII), Universidade de Santiago de Compostela, Praza do Obradoiro s/n, 15782, Santiago de Compostela, Spain
| | - Oliver Robinson
- MRC Centre for Environment and Health, School of Public Health, Imperial College London, St. Mary's Hospital Campus, London, W21PG, UK
| | - Eduard Sabidó
- Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Gillian Santorelli
- Bradford Institute for Health Research, Bradford Royal Infirmary, Bradford, BD9 6RJ, UK
| | - Per E Schwarze
- Department af Environmental Health, Norwegian Institute of Public Health, Lovisenberggt 6, 0456, Oslo, Norway
| | - Alexandros P Siskos
- Division of Systems Medicine, Department of Metabolism, Digestion and Reproduction, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
- Cancer Metabolism and Systems Toxicology Group, Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Rémy Slama
- University Grenoble Alpes, Inserm, CNRS, Team of Environmental Epidemiology Applied to Reproduction and Respiratory Health, IAB, 38000, Grenoble, France
| | - Marina Vafeiadi
- Department of Social Medicine, School of Medicine, University of Crete, Heraklion, Crete, Greece
| | - Eulàlia Martí
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
- Departament de Biomedicina, Institut de Neurociències, Universitat de Barcelona, Barcelona, Spain
| | - Martine Vrijheid
- ISGlobal, Barcelona, Spain
- Universitat Pompeu Fabra (UPF), Barcelona, Spain
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain
| | - Mariona Bustamante
- ISGlobal, Barcelona, Spain.
- Universitat Pompeu Fabra (UPF), Barcelona, Spain.
- CIBER Epidemiología y Salud Pública (CIBERESP), Madrid, Spain.
| |
Collapse
|
169
|
Determination of genetic changes of Rev-erb beta and Rev-erb alpha genes in Type 2 diabetes mellitus by next-generation sequencing. Gene 2020; 763:145058. [PMID: 32798635 DOI: 10.1016/j.gene.2020.145058] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/07/2020] [Accepted: 08/12/2020] [Indexed: 12/30/2022]
Abstract
BACKGROUND The nuclear receptors Rev-erb alpha and Rev-erb beta are transcription factors that regulate the function of genes in glucose and lipid metabolism, and they also form a link between circadian rhythm and metabolism. We evaluated the variations in Rev-erb alpha and Rev-erb beta genes together with biochemical parameters as risk factors in type 2 diabetic (T2DM) patients. METHODS Molecular analyses of Rev-erb alpha and Rev-erb beta genes were performed on genomic DNA by using next-generation sequencing in 42 T2DM patients (21 obese and 21 non-obese) and 66 healthy controls. RESULTS We found 26 rare mutations in the study groups, including 13 missense mutations, 9 silent mutations, 3 5'UTR variations, and a 3'UTR variation, of which 9 were novel variations (5 missense and 3 silent and 1 5'UTR). Six common variations were also found in the Rev-erb genes; Rev-erb beta Chr3:24003765 A > G, Rev-erb beta rs924403442 (Chr3:24006717) G > T, Rev-erb alpha Chr17:38253751 T > C, Rev-erb alpha rs72836608 C > A, Rev-erb alpha rs2314339 C > T and Rev-erb alpha rs2102928 C > T. Of these, Rev-erb beta Chr3:24003765 A > G was a novel missense mutation (p.Q197R), while others were identified as intronic variants. T2DM patients with Rev-erb beta rs924403442 T allele had lower body surface area (BSA) than noncarriers (GG genotype) (p = 0.039). Rev-erb alpha rs72836608 A allele and Rev-erb alpha rs2314339 CC genotype were associated with decreased serum HDL-cholesterol levels in T2DM patients (p = 0.025 and p = 0.027, respectively). In our study, different effects of Rev-erbs polymorphisms were found according to gender and presence of obesity. Rev-erb alpha rs72836608 (C > A) and rs2314339 (C > T) and Rev-erb alpha rs2102928 (C > T) were associated with low HDL-C levels in male T2DM patients. In female patients, Rev-erb alpha rs2102928 (C > T) was associated with high microalbuminuria and Rev-erb beta rs9244403442 G > T was associated with low HDL and high BSA values. In addition, Rev-erb alpha Chr17: 38,253,751 (T > C), rs72836608 (C > A), and rs2314339 (C > T) and Rev-erb beta Chr3:24003765 (A > G) were associated with increased serum GGT levels in obese T2DM patients. In non-obese patients, Rev-erbs SNPs had no effect on serum GGT levels. CONCLUSION Our findings indicate that variations in the Rev-erb alpha and Rev-erb beta genes can affect metabolic changes in T2DM and these effects may vary depending on gender and obesity.
Collapse
|
170
|
Grynchuk FV, Dutka II, Panchuk II, Volkov RA, Sheremet MI, Maksymyuk VV, Tarabanchuk VV, Bilyk II, Myshkovskii YM. Justification of Genetic Factors for Predicting the Risk of Acute Bleeding in Peptic Ulcer Disease. J Med Life 2020; 13:255-259. [PMID: 32742523 PMCID: PMC7378332 DOI: 10.25122/jml-2020-0041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
PAI genotyping for the G43A and 4G/5G polymorphisms was performed in 60 patients with peptic ulcer disease: 12 with an uncomplicated ulcer, 5 with perforation, the rest with ongoing bleeding. Fourteen patients had recurrent bleeding. The 5G/5G and G43A genotypes were not detected in patients with uncomplicated ulcers. All patients with ulcer perforation had the G43G genotype, 60% of patients had the 4G/4G genotype, and the rest of them had the 4G/5G and 5G/5G genotypes. The number of carriers of the 5G allele (86.05%) was higher in patients with bleeding than in ones with ulcer perforation (p=0.036) and ulcer without bleeding (p=0.021, χ2=5.32). The number of carriers of the 5G allele was higher in patients with recurrent bleeding (92.86%) than those without any relapses (82.76%) but there were no statistically significant differences (p=0.27, χ2=0.802). The G43G homozygous genotype was found in 94.12% of patients with peptic ulcer without bleeding, which was statistically significantly higher (p=0.02) than the ones with bleeding. The A allele was observed in 27.91% of patients with bleeding and 8.33% patients without any bleeding (p=0.05). The number of carriers of the A allele in patients with recurrent bleeding was statistically significantly higher than in ones without any bleeding (p=0.046). The 5G and A alleles in patients with a peptic ulcer can be used to predict the course of peptic ulcer disease and can be regarded as a predictor of the risk of bleeding relapse.
Collapse
Affiliation(s)
- Fedir Vasilyevich Grynchuk
- First Department of Surgery, Higher State Educational Establishment "Bukovinian State Medical University", Chernivtsi, Ukraine
| | - Ivan Ivanovich Dutka
- First Department of Surgery, Higher State Educational Establishment "Bukovinian State Medical University", Chernivtsi, Ukraine
| | - Iryna Ihorivna Panchuk
- Department of Molecular Genetics and Biotechnology, Yuriy Fedkovych Chernivtsi National University, Institute of Biology, Chemistry and Bioresources, Chernivtsi, Ukraine
| | - Roman Anatolyevich Volkov
- Department of Molecular Genetics and Biotechnology, Yuriy Fedkovych Chernivtsi National University, Institute of Biology, Chemistry and Bioresources, Chernivtsi, Ukraine
| | - Michael Ivanovich Sheremet
- First Department of Surgery, Higher State Educational Establishment "Bukovinian State Medical University", Chernivtsi, Ukraine
| | - Vitaliy Vasilyevich Maksymyuk
- First Department of Surgery, Higher State Educational Establishment "Bukovinian State Medical University", Chernivtsi, Ukraine
| | | | - Ihor Ivanovich Bilyk
- Department of General Surgery, Higher State Educational Establishment "Bukovinian State Medical University", Chernivtsi, Ukraine
| | - Yuriy Mykolayovych Myshkovskii
- Department of General Surgery, Higher State Educational Establishment "Bukovinian State Medical University", Chernivtsi, Ukraine
| |
Collapse
|
171
|
Kris-Etherton PM, Stewart PW, Ginsberg HN, Tracy RP, Lefevre M, Elmer PJ, Berglund L, Ershow AG, Pearson TA, Ramakrishnan R, Holleran SF, Dennis BH, Champagne CM, Karmally W. The Type and Amount of Dietary Fat Affect Plasma Factor VIIc, Fibrinogen, and PAI-1 in Healthy Individuals and Individuals at High Cardiovascular Disease Risk: 2 Randomized Controlled Trials. J Nutr 2020; 150:2089-2100. [PMID: 32492148 PMCID: PMC7398773 DOI: 10.1093/jn/nxaa137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/12/2019] [Accepted: 04/21/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Factor VIIc, fibrinogen, and plasminogen activator inhibitor 1 (PAI-1) are cardiovascular disease (CVD) risk factors and are modulated, in part, by fat type and amount. OBJECTIVE We evaluated fat type and amount on the primary outcomes: factor VIIc, fibrinogen, and PAI-1. METHODS In the Dietary Effects on Lipoproteins and Thrombogenic Activity (DELTA) Trial, 2 controlled crossover feeding studies evaluated substituting carbohydrate or MUFAs for SFAs. Study 1: healthy participants (n = 103) were provided with (8 wk) an average American diet [AAD; designed to provide 37% of energy (%E) as fat, 16% SFA], a Step 1 diet (30%E fat, 9% SFA), and a diet low in SFA (Low-Sat; 26%E fat, 5% SFA). Study 2: participants (n = 85) at risk for CVD and metabolic syndrome (MetSyn) were provided with (7 wk) an AAD, a step 1 diet, and a high-MUFA diet (designed to provide 37%E fat, 8% SFA, 22% MUFA). RESULTS Study 1: compared with AAD, the Step 1 and Low-Sat diets decreased mean factor VIIc by 1.8% and 2.6% (overall P = 0.0001), increased mean fibrinogen by 1.2% and 2.8% (P = 0.0141), and increased mean square root PAI-1 by 0.0% and 6.0% (P = 0.0037), respectively. Study 2: compared with AAD, the Step 1 and high-MUFA diets decreased mean factor VIIc by 4.1% and 3.2% (overall P < 0.0001), increased mean fibrinogen by 3.9% and 1.5% (P = 0.0083), and increased mean square-root PAI-1 by 2.0% and 5.8% (P = 0.1319), respectively. CONCLUSIONS Replacing SFA with carbohydrate decreased factor VIIc and increased fibrinogen in healthy and metabolically unhealthy individuals and also increased PAI-1 in healthy subjects. Replacing SFA with MUFA decreased factor VIIc and increased fibrinogen but less than carbohydrate. Our results indicate an uncertain effect of replacing SFA with carbohydrate or MUFA on cardiometabolic risk because of small changes in hemostatic factors and directionally different responses to decreasing SFA. This trial was registered at https://clinicaltrials.gov/ct2/show/NCT00000538?term=NCT00000538&rank=1 as NCT00000538.
Collapse
Affiliation(s)
- Penny M Kris-Etherton
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | - Paul W Stewart
- Department of Biostatistics, Collaborative Studies Coordinating Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Henry N Ginsberg
- Department of Medicine, Irving Center for Clinical Research, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Russell P Tracy
- Colchester Research Facility, University of Vermont, Colchester, VT, USA
| | - Michael Lefevre
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Patricia J Elmer
- Division of Epidemiology, University of Minnesota School of Public Health, Minneapolis, MN, USA
| | - Lars Berglund
- Department of Medicine, Irving Center for Clinical Research, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Abby G Ershow
- Division of Heart and Vascular Diseases, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Thomas A Pearson
- The Mary Imogene Bassett Research Institute, Cooperstown, NY, USA
- School of Public Health, State University of New York at Albany, Albany, NY, USA
- Columbia University College of Physicians and Surgeons, New York, NY, USA
- Department of Community and Preventive Medicine, University of Rochester, Rochester, NY, USA
| | - Rajasekhar Ramakrishnan
- Department of Medicine, Irving Center for Clinical Research, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Stephen F Holleran
- Department of Medicine, Irving Center for Clinical Research, Columbia University College of Physicians and Surgeons, New York, NY, USA
| | - Barbara H Dennis
- Department of Biostatistics, Collaborative Studies Coordinating Center, The University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Catherine M Champagne
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA, USA
| | - Wahida Karmally
- Department of Medicine, Irving Center for Clinical Research, Columbia University College of Physicians and Surgeons, New York, NY, USA
| |
Collapse
|
172
|
Zhang X, Cai X, Pan J. Correlation Between PAI-1 Gene 4G/5G Polymorphism and the Risk of Thrombosis in Ph Chromosome-Negative Myeloproliferative Neoplasms. Clin Appl Thromb Hemost 2020; 26:1076029620935207. [PMID: 32683889 PMCID: PMC7372617 DOI: 10.1177/1076029620935207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Thrombosis has been recognized as one of the most significant risk factors of high mortality and disability in patients with Philadelphia (Ph) chromosome negative myeloproliferative neoplasms (MPNs). However, the risk factors of thrombotic events in these patients have not been completely understood. In this study, the clinical data of 58 patients with Ph-MPNs were obtained and analyzed, including 34 cases of essential thrombocytopenia (ET), 23 thrombotic events happened in 21 (36%) patients, among which 60% (14 of 23) with cerebral infarction, 17% (4 of 23) with coronary heart disease and 23% (5 of 23) with venous thrombosis. There were no significant differences in age, sex, and blood cell count between polycythemia vera (PV) and ET patients who have experienced thrombotic events and those who have not. In ET patients, the incidence of thrombotic events in plasminogen activator inhibitor-1 (PAI-1) genotype 4G4G was significantly higher than that in genotype 4G5G and genotype 5G5G (P < .05). The incidence of thrombotic events in PV and ET patients with infection was higher than those without infection (P < .05). Using logistic regression analysis, we found that PAI-1 genotype 4G4G and infection were associated with thrombotic events (odds ratio 6.744, 95% CI: 1.195-38.056 and 15.641 95% CI: 3.327-73.522). The 4G/4G polymorphism of PAI-1 gene and infection are independent risk factors of thrombotic events in patients with Ph-MPNs. PAI-1 gene 4G4G and infection in ET and PV patients with Janus kinase 2 (JAK2) V617F mutation were shown to be high risk of thrombotic events. Therefore, clinical doctors should put more attention on PAI-1 genotype 4G4G and infection in JAK2 V617F mutated patients with Ph-MPNs to prevent the thrombosis.
Collapse
Affiliation(s)
- Xueya Zhang
- Department of Hematology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Xuerong Cai
- Department of Respiratory Medicine, The Quanzhou First Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| | - Jingxin Pan
- Department of Hematology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, China
| |
Collapse
|
173
|
Rahman FA, Angus SA, Stokes K, Karpowicz P, Krause MP. Impaired ECM Remodeling and Macrophage Activity Define Necrosis and Regeneration Following Damage in Aged Skeletal Muscle. Int J Mol Sci 2020; 21:ijms21134575. [PMID: 32605082 PMCID: PMC7369722 DOI: 10.3390/ijms21134575] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 06/16/2020] [Accepted: 06/24/2020] [Indexed: 12/28/2022] Open
Abstract
Regenerative capacity of skeletal muscle declines with age, the cause of which remains largely unknown. We investigated extracellular matrix (ECM) proteins and their regulators during early regeneration timepoints to define a link between aberrant ECM remodeling, and impaired aged muscle regeneration. The regeneration process was compared in young (three month old) and aged (18 month old) C56BL/6J mice at 3, 5, and 7 days following cardiotoxin-induced damage to the tibialis anterior muscle. Immunohistochemical analyses were performed to assess regenerative capacity, ECM remodeling, and the macrophage response in relation to plasminogen activator inhibitor-1 (PAI-1), matrix metalloproteinase-9 (MMP-9), and ECM protein expression. The regeneration process was impaired in aged muscle. Greater intracellular and extramyocellular PAI-1 expression was found in aged muscle. Collagen I was found to accumulate in necrotic regions, while macrophage infiltration was delayed in regenerating regions of aged muscle. Young muscle expressed higher levels of MMP-9 early in the regeneration process that primarily colocalized with macrophages, but this expression was reduced in aged muscle. Our results indicate that ECM remodeling is impaired at early time points following muscle damage, likely a result of elevated expression of the major inhibitor of ECM breakdown, PAI-1, and consequent suppression of the macrophage, MMP-9, and myogenic responses.
Collapse
Affiliation(s)
- Fasih Ahmad Rahman
- Department of Kinesiology, University of Windsor. Windsor, ON N9B 3P4, Canada; (F.A.R.); (S.A.A.)
| | - Sarah Anne Angus
- Department of Kinesiology, University of Windsor. Windsor, ON N9B 3P4, Canada; (F.A.R.); (S.A.A.)
| | - Kyle Stokes
- Department of Biomedical Sciences, University of Windsor. Windsor, ON N9B 3P4, Canada; (K.S.); (P.K.)
| | - Phillip Karpowicz
- Department of Biomedical Sciences, University of Windsor. Windsor, ON N9B 3P4, Canada; (K.S.); (P.K.)
| | - Matthew Paul Krause
- Department of Kinesiology, University of Windsor. Windsor, ON N9B 3P4, Canada; (F.A.R.); (S.A.A.)
- Correspondence: ; Tel.: +1-519-253-3000
| |
Collapse
|
174
|
Oh J, An HJ, Kim JO, Jun HH, Kim WR, Kim EJ, Oh D, Kim JW, Kim NK. Association between Five Common Plasminogen Activator Inhibitor-1 ( PAI-1) Gene Polymorphisms and Colorectal Cancer Susceptibility. Int J Mol Sci 2020; 21:ijms21124334. [PMID: 32570732 PMCID: PMC7352892 DOI: 10.3390/ijms21124334] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Revised: 06/16/2020] [Accepted: 06/16/2020] [Indexed: 12/31/2022] Open
Abstract
The plasminogen activator inhibitor-1 (PAI-1) is expressed in many cancer cell types and modulates cancer growth, invasion, and angiogenesis. The present study investigated the association between five PAI-1 gene polymorphisms and colorectal cancer (CRC) risk. Five PAI-1 polymorphisms (−844G > A [rs2227631], −675 4G > 5G [rs1799889], +43G > A [rs6092], +9785G > A [rs2227694], and +11053T > G [rs7242]) were genotyped using a polymerase chain reaction-restriction fragment length polymorphism assay in 459 CRC cases and 416 controls. Increased CRC risk was more frequently associated with PAI-1 −675 5G5G polymorphism than with 4G4G (adjusted odds ratio (AOR) = 1.556; 95% confidence interval (CI): 1.012–2.391; p = 0.04). In contrast, for the PAI-1 +11053 polymorphism, we found a lower risk of CRC with the GG genotype (AOR = 0.620; 95% CI: 0.413–0.932; p = 0.02) than with the TT genotype, as well as for recessive carriers (TT + TG vs. GG, AOR = 0.662; 95% CI: 0.469–0.933; p = 0.02). The +43AA genotype was associated with lower overall survival (OS) than the +43GG genotype. Our results suggest that the PAI-1 genotype plays a role in CRC risk. This is the first study to identify an association between five PAI-1 polymorphisms and CRC incidence worldwide.
Collapse
Affiliation(s)
- Jisu Oh
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (J.O.); (D.O.)
| | - Hui Jeong An
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea; (H.J.A.); (J.O.K.)
| | - Jung Oh Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea; (H.J.A.); (J.O.K.)
| | - Hak Hoon Jun
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (H.H.J.); (W.R.K.)
| | - Woo Ram Kim
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (H.H.J.); (W.R.K.)
| | - Eo Jin Kim
- Department on Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Korea;
| | - Doyeun Oh
- Department of Internal Medicine, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (J.O.); (D.O.)
| | - Jong Woo Kim
- Department of Surgery, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea; (H.H.J.); (W.R.K.)
- Correspondence: (J.W.K.); (N.K.K.); Tel.: +82-31-881-7137 (N.K.K.); Fax: +82-31-881-7249 (N.K.K.)
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea; (H.J.A.); (J.O.K.)
- Correspondence: (J.W.K.); (N.K.K.); Tel.: +82-31-881-7137 (N.K.K.); Fax: +82-31-881-7249 (N.K.K.)
| |
Collapse
|
175
|
Olabode OP, Akinlade OM, Babatunde AS, Abdulazeez MI, Biliaminu SA, Oyabambi AO, Olatunji VA, Soladoye AO, Olatunji LA. Triglyceride/HDL-cholesterol ratio and plasminogen activator inhibitor-1 independently predict high pulse pressure in sickle cell trait and disease. Arch Physiol Biochem 2020; 126:166-171. [PMID: 30145922 DOI: 10.1080/13813455.2018.1499118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
We hypothesised that TG/HDL-C ratio and PAI-1 would be associated with high pulse pressure (PP) in young adults with sickle cell trait (SCT) and sickle cell disease (SCD). We compared the clinical, biochemical, and cardiometabolic parameters among individuals with normal genotype (HbAA; n = 60), SCT (HbAS; n = 60), and SCD (HbSS; n = 60), all in steady state. Using multivariate linear regression analysis, high PP was positively related to TG/HDL-C ratio in SCT (β = 0.307; p = .014) and PAI-1 (β = 0.499; p = .001) in SCD. The curve of receiver operating characteristic also showed that TG/HDL-C ratio and PAI-1 are efficient predictors of high PP in SCT carriers and SCD patients, respectively. This study suggests that increased levels of TG/HDL-C ratio and PAI-1 may be salient risk factors that would promote the development of arterial stiffness and other CVD in SCT carriers and SCD patients.
Collapse
Affiliation(s)
- Olatunde P Olabode
- HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Olawale M Akinlade
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Medicine, Ladoke Akintola University of Technology Teaching Hospital, Ogbomoso, Nigeria
| | - Abiola S Babatunde
- Department of Haematology and Blood Transfusion, College of Health Sciences, University of Ilorin, Nigeria
| | - Musbau I Abdulazeez
- Department of Chemical Pathology and Immunology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Sikiru A Biliaminu
- Department of Chemical Pathology and Immunology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Adewumi O Oyabambi
- HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Victoria A Olatunji
- HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Ophthalmology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Ayodele O Soladoye
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| | - Lawrence A Olatunji
- HOPE Cardiometabolic Research Team, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
- Department of Physiology, College of Health Sciences, University of Ilorin, Ilorin, Nigeria
| |
Collapse
|
176
|
Garcia V, Park EJ, Siragusa M, Frohlich F, Mahfuzul Haque M, Pascale JV, Heberlein KR, Isakson BE, Stuehr DJ, Sessa WC. Unbiased proteomics identifies plasminogen activator inhibitor-1 as a negative regulator of endothelial nitric oxide synthase. Proc Natl Acad Sci U S A 2020; 117:9497-9507. [PMID: 32300005 PMCID: PMC7196906 DOI: 10.1073/pnas.1918761117] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Nitric oxide (NO) produced by endothelial nitric oxide synthase (eNOS) is a critical mediator of vascular function. eNOS is tightly regulated at various levels, including transcription, co- and posttranslational modifications, and by various protein-protein interactions. Using stable isotope labeling with amino acids in cell culture (SILAC) and mass spectrometry (MS), we identified several eNOS interactors, including the protein plasminogen activator inhibitor-1 (PAI-1). In cultured human umbilical vein endothelial cells (HUVECs), PAI-1 and eNOS colocalize and proximity ligation assays demonstrate a protein-protein interaction between PAI-1 and eNOS. Knockdown of PAI-1 or eNOS eliminates the proximity ligation assay (PLA) signal in endothelial cells. Overexpression of eNOS and HA-tagged PAI-1 in COS7 cells confirmed the colocalization observations in HUVECs. Furthermore, the source of intracellular PAI-1 interacting with eNOS was shown to be endocytosis derived. The interaction between PAI-1 and eNOS is a direct interaction as supported in experiments with purified proteins. Moreover, PAI-1 directly inhibits eNOS activity, reducing NO synthesis, and the knockdown or antagonism of PAI-1 increases NO bioavailability. Taken together, these findings place PAI-1 as a negative regulator of eNOS and disruptions in eNOS-PAI-1 binding promote increases in NO production and enhance vasodilation in vivo.
Collapse
Affiliation(s)
- Victor Garcia
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Eon Joo Park
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
| | - Mauro Siragusa
- Institute for Vascular Signaling, Centre for Molecular Medicine, Goethe University, 60596 Frankfurt am Main, Germany
| | - Florian Frohlich
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520
- Department of Biology/Chemistry, Molecular Membrane Biology Section, University of Osnabrück, 49076 Osnabrück, Germany
| | - Mohammad Mahfuzul Haque
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - Jonathan V Pascale
- Department of Pharmacology, New York Medical College, Valhalla, NY 10595
| | - Katherine R Heberlein
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Brant E Isakson
- Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908
| | - Dennis J Stuehr
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195
| | - William C Sessa
- Vascular Biology and Therapeutics Program, Department of Pharmacology, Yale University School of Medicine, New Haven, CT 06520;
| |
Collapse
|
177
|
Muñoz-Galván S, Rivero M, Peinado-Serrano J, Martinez-Pérez J, Fernández-Fernández MC, José Ortiz M, García-Heredia JM, Carnero A. PAI1 is a Marker of Bad Prognosis in Rectal Cancer but Predicts a Better Response to Treatment with PIM Inhibitor AZD1208. Cells 2020; 9:cells9051071. [PMID: 32344898 PMCID: PMC7291071 DOI: 10.3390/cells9051071] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/24/2022] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer worldwide. The standard treatment in locally advanced rectal cancer is preoperative radiation alone or in combination with chemotherapy, followed by adjuvant chemotherapy. Rectal cancer is highly lethal, with only 20% of patients showing a complete remission (by RECIST) after standard treatment, although they commonly show local or systemic relapse likely due to its late detection and high chemotherapy resistance, among other reasons. Here, we explored the role of PAI1 (Serpin E1) in rectal cancer through the analyses of public patient databases, our own cohort of locally advanced rectal cancer patients and a panel of CRC cell lines. We showed that PAI1 expression is upregulated in rectal tumors, which is associated with decreased overall survival and increased metastasis and invasion in advanced rectal tumors. Accordingly, PAI1 expression is correlated with the expression of (Epithelial-to-Mesenchymal Transition) EMT-associated genes and genes encoding drug targets, including the tyrosine kinases PDGFRb, PDGFRa and FYN, the serine/threonine kinase PIM1 and BRAF. In addition, we demonstrate that cells expressing PAI1 protein are more sensitive to the PIM inhibitor AZD1208, suggesting that PAI1 could be used to predict response to treatment with PIM inhibitors and to complement radiotherapy in rectal tumors.
Collapse
Affiliation(s)
- Sandra Muñoz-Galván
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain; (S.M.-G.); (M.R.); (J.P.-S.); (J.M.-P.); (J.M.G.-H.)
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Maria Rivero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain; (S.M.-G.); (M.R.); (J.P.-S.); (J.M.-P.); (J.M.G.-H.)
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Javier Peinado-Serrano
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain; (S.M.-G.); (M.R.); (J.P.-S.); (J.M.-P.); (J.M.G.-H.)
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Radiation Oncology, HUVR, 41013 Seville, Spain; (M.C.F.-F.); (M.J.O.)
| | - Julia Martinez-Pérez
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain; (S.M.-G.); (M.R.); (J.P.-S.); (J.M.-P.); (J.M.G.-H.)
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Department of Medical Oncology, HUVR, 41013 Seville, Spain
| | | | - María José Ortiz
- Department of Radiation Oncology, HUVR, 41013 Seville, Spain; (M.C.F.-F.); (M.J.O.)
| | - José M. García-Heredia
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain; (S.M.-G.); (M.R.); (J.P.-S.); (J.M.-P.); (J.M.G.-H.)
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Amancio Carnero
- Instituto de Biomedicina de Sevilla, IBIS, Hospital Universitario Virgen del Rocío, Universidad de Sevilla, Consejo Superior de Investigaciones Científicas, Avda. Manuel Siurot s/n, 41013 Seville, Spain; (S.M.-G.); (M.R.); (J.P.-S.); (J.M.-P.); (J.M.G.-H.)
- CIBERONC, Instituto de Salud Carlos III, 28029 Madrid, Spain
- Correspondence: ; Tel.: +34955923110
| |
Collapse
|
178
|
Gruevska A, Moragrega AB, Galindo MJ, Esplugues JV, Blas-Garcia A, Apostolova N. p53 and p53-related mediators PAI-1 and IGFBP-3 are downregulated in peripheral blood mononuclear cells of HIV-patients exposed to non-nucleoside reverse transcriptase inhibitors. Antiviral Res 2020; 178:104784. [PMID: 32272174 DOI: 10.1016/j.antiviral.2020.104784] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 03/22/2020] [Accepted: 03/23/2020] [Indexed: 01/11/2023]
Abstract
The improved effectiveness and safety of the combined antiretroviral therapy (cART) has largely diminished mortality and AIDS-defining morbidity of HIV-patients. Nevertheless, chronic age-related diseases in these individuals are more common and their underlying pathogenic mechanisms of these actions seem to involve accelerated aging and enhanced inflammation. The present study explores markers of these processes in a heterogenous Spanish HIV cohort using peripheral blood samples of HIV-patients and matched uninfected controls. We isolated periheral blood mononuclear cells (PBMCs) and i) compared the expression of a panel of 14 genes related to inflammation and senescence in PBMCs of HIV-patients vs matched uninfected controls, ii) analyzed the expression in HIV-patients in association with a number of demographic, biochemical and immunological parameters and iii) in relation with the current cART they received. PBMCs of HIV-patients displayed significantly increased expression of general inflammatory genes (IL6, IL18 and CXCL10) and this occurs irrespectively of the antiviral therapy they have been receiving. Conversely, levels of senescence-associated genes TP53, SERPINE1andIGFBP3 were slightly but significantly reduced in patients compared to uninfected matched individuals and this effect is related to NNRTI-containing treatments. The expression of the inflammatory markers IL6, IL18, IL1B, TNFA, RELA, CCL2, CCL20 and CXCL10 displayed correlation with certain demographic, morbidity- and HIV infection-related parameters. The levels of TP53 mRNA were positively associated only with plasma LDL. Correlation analysis between the expressions of pairs of genes revealed a different pattern between HIV-patients and controls. The diminished expression of TP53 and SERPINE1 in HIV-patients was also observed at a protein level, and the correlation between the two proteins (p53 and PAI1) in patients and controls showed the opposite trend. In conclusion, HIV-patients show dysregulation of p53 and p53-related mediators, a phenomenon which may be of pathophysiological relevance and could be related to the shorter health- and/or life-span observed in these individuals.
Collapse
Affiliation(s)
- Aleksandra Gruevska
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
| | - Angela B Moragrega
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain
| | - Maria J Galindo
- Unidad de Enfermedades Infecciosas - Medicina Interna, Hospital Clínico Universitario de Valencia, Spain
| | - Juan V Esplugues
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain; CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Spain
| | - Ana Blas-Garcia
- FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain; CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Spain; Departamento de Fisiología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Nadezda Apostolova
- Departamento de Farmacología, Facultad de Medicina, Universidad de Valencia, Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Valencia, Spain; CIBERehd (Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas), Spain.
| |
Collapse
|
179
|
Lu AT, Quach A, Wilson JG, Reiner AP, Aviv A, Raj K, Hou L, Baccarelli AA, Li Y, Stewart JD, Whitsel EA, Assimes TL, Ferrucci L, Horvath S. DNA methylation GrimAge strongly predicts lifespan and healthspan. Aging (Albany NY) 2020; 11:303-327. [PMID: 30669119 PMCID: PMC6366976 DOI: 10.18632/aging.101684] [Citation(s) in RCA: 1275] [Impact Index Per Article: 255.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/22/1969] [Indexed: 12/16/2022]
Abstract
It was unknown whether plasma protein levels can be estimated based on DNA methylation (DNAm) levels, and if so, how the resulting surrogates can be consolidated into a powerful predictor of lifespan. We present here, seven DNAm-based estimators of plasma proteins including those of plasminogen activator inhibitor 1 (PAI-1) and growth differentiation factor 15. The resulting predictor of lifespan, DNAm GrimAge (in units of years), is a composite biomarker based on the seven DNAm surrogates and a DNAm-based estimator of smoking pack-years. Adjusting DNAm GrimAge for chronological age generated novel measure of epigenetic age acceleration, AgeAccelGrim.Using large scale validation data from thousands of individuals, we demonstrate that DNAm GrimAge stands out among existing epigenetic clocks in terms of its predictive ability for time-to-death (Cox regression P=2.0E-75), time-to-coronary heart disease (Cox P=6.2E-24), time-to-cancer (P= 1.3E-12), its strong relationship with computed tomography data for fatty liver/excess visceral fat, and age-at-menopause (P=1.6E-12). AgeAccelGrim is strongly associated with a host of age-related conditions including comorbidity count (P=3.45E-17). Similarly, age-adjusted DNAm PAI-1 levels are associated with lifespan (P=5.4E-28), comorbidity count (P= 7.3E-56) and type 2 diabetes (P=2.0E-26). These DNAm-based biomarkers show the expected relationship with lifestyle factors including healthy diet and educational attainment.Overall, these epigenetic biomarkers are expected to find many applications including human anti-aging studies.
Collapse
Affiliation(s)
- Ake T Lu
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Austin Quach
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - James G Wilson
- Department of Physiology and Biophysics, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Alex P Reiner
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Abraham Aviv
- Center of Development and Aging, New Jersey Medical School, Rutgers State University of New Jersey, Newark, NJ 07103, USA
| | - Kenneth Raj
- Radiation Effects Department, Centre for Radiation, Chemical and Environmental Hazards, Public Health England, Chilton, Didcot, Oxfordshire OX11 0RQ, United Kingdom
| | - Lifang Hou
- Center for Population Epigenetics, Robert H. Lurie Comprehensive Cancer Center and Department of Preventive Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Andrea A Baccarelli
- Laboratory of Environmental Epigenetics, Departments of Environmental Health Sciences Epidemiology, Columbia University Mailman School of Public Health, New York, NY 10032, USA
| | - Yun Li
- Departments of Genetics, Biostatistics, Computer Science, University of North Carolina, Chapel Hill, NC 27599, USA
| | - James D Stewart
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eric A Whitsel
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599, USA.,Department of Medicine, School of Medicine, University of North Carolina, Chapel Hill, NC 27516, USA
| | - Themistocles L Assimes
- Department of Medicine (Division of Cardiovascular Medicine), Stanford University School of Medicine, Stanford, CA 94305, USA.,VA Palo Alto Health Care System, Palo Alto, CA 94304, USA
| | - Luigi Ferrucci
- Longitudinal Studies Section, Translational Gerontology Branch, National Institute on Aging, National Institutes of Health, USA, Baltimore, MD 21224, USA
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA.,Department of Biostatistics, Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
180
|
Vasculoprotective Effects of Vildagliptin. Focus on Atherogenesis. Int J Mol Sci 2020; 21:ijms21072275. [PMID: 32218354 PMCID: PMC7177465 DOI: 10.3390/ijms21072275] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 03/19/2020] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
Vildagliptin is a representative of Dipeptidyl Peptidase-4 (DPP-4) inhibitors, antihyperglycemic drugs, approved for use as monotherapy and combination therapy in type 2 diabetes mellitus. By inhibiting enzymatic decomposition, DPP-4 inhibitors increase the half-life of incretins such as GLP-1 (Glucagon-like peptide-1) and GIP (Gastric inhibitors polypeptide) and prolong their action. Some studies present results suggesting the anti-sclerotic and vasculoprotective effects of vildagliptin reaching beyond glycemic control. Vildagliptin is able to limit inflammation by suppression of the NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) signaling pathway and proinflammatory agents such as TNF-α (tumor necrosis factor α), IL-1β (Interleukin-1β), and IL-8 (Interleukin 8). Moreover, vildagliptin regulates lipid metabolism; attenuates postprandial hypertriglyceridemia; and lowers serum triglycerides, apolipoprotein B, and blood total cholesterol levels. This DPP-4 inhibitor also reduces macrophage foam cell formation, which plays a key role in atheromatous plaque formation and stability. Vildagliptin reduces vascular stiffness via elevation of nitric oxide synthesis, improves vascular relaxation, and results in reduction in both systolic and diastolic blood pressure. Treatment with vildagliptin lowers the level of PAI-1 presenting possible antithrombotic effect. By affecting the endothelium, inflammation, and lipid metabolism, vildagliptin may affect the development of atherosclerosis at its various stages. The article presents a summary of the studies assessing vasculoprotective effects of vildagliptin with special emphasis on atherogenesis.
Collapse
|
181
|
Assem S, Abdelbaki TN, Mohy-El Dine SH, Ketat AF, Abdelmonsif DA. SERPINE-1 Gene Methylation and Protein as Molecular Predictors of Laparoscopic Sleeve Gastrectomy Outcome. Obes Surg 2020; 30:2620-2630. [PMID: 32170551 DOI: 10.1007/s11695-020-04533-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
BACKGROUND Body weight is subjected to genetic and epigenetic modifiers that might affect the success of weight loss interventions. Because of its possible complications and disparity in patients' response, identification of predictors to the outcome of bariatric surgery is indispensable. OBJECTIVES This prospective study aims to investigate serpin peptidase inhibitor type 1 (SERPINE-1) protein and gene methylation as molecular predictors to the outcome of bariatric surgery. PATIENTS AND METHODS One hundred participants were enrolled and divided to control group (n = 50) and obese patients who underwent laparoscopic sleeve gastrectomy (LSG) (n = 50). Anthropometric measurements were assessed and blood samples were collected preoperatively and 6 months postoperatively for assessment of SERPINE-1 protein and gene methylation, C-reactive protein (CRP), and homeostatic model assessment of insulin resistance (HOMA-IR). Moreover, subjects were followed for 2 years for weight loss parameters. RESULTS Patients with obesity showed high baseline SERPINE-1 protein and gene hypermethylation where LSG was followed by a drop in SERPINE-1 protein level but not gene hypermethylation. Baseline SERPINE-1 gene methylation was negatively related to postoperative weight loss and was the independent predictor to weight loss after LSG. Likewise, postoperative SERPINE-1 protein was negatively related to weight loss with independent expression from its gene methylation state. Furthermore, postoperative SERPINE-1 gene methylation correlated to CRP and HOMA-IR. CONCLUSION Baseline SERPINE-1 gene methylation might be a predictor of weight loss after LSG. Meanwhile, postoperative SERPINE-1 protein could be a predictor to weight loss maintenance after LSG. Lastly, postoperative SERPINE-1 gene methylation might serve as an index to postoperative changes in obesity-related comorbidities.
Collapse
Affiliation(s)
- Sara Assem
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Tamer N Abdelbaki
- Department of Surgery, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Safaa H Mohy-El Dine
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Amel F Ketat
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | - Doaa A Abdelmonsif
- Department of Medical Biochemistry, Faculty of Medicine, University of Alexandria, Alexandria, Egypt. .,Molecular Biology Lab. and Nanomedicine Lab., Center of Excellence for Research in Regenerative Medicine and Applications, University of Alexandria, Alexandria, Egypt.
| |
Collapse
|
182
|
Antunes BM, Rossi FE, Oyama LM, Rosa-Neto JC, Lira FS. Exercise intensity and physical fitness modulate lipoproteins profile during acute aerobic exercise session. Sci Rep 2020; 10:4160. [PMID: 32139762 PMCID: PMC7058045 DOI: 10.1038/s41598-020-61039-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Accepted: 02/20/2020] [Indexed: 12/11/2022] Open
Abstract
Physical inactivity has emerged as an important cardiometabolic risk factor; however, the beneficial impacts of physical exercise according physical fitness status are still unclear. To analyze the lipoproteins and immune-endocrine response to acute aerobic exercise sessions performed at different intensities according physical fitness status and evaluated the gene expression in monocyte cells. Twelve individuals, divided into Low and High VO2max, performed three randomized acute exercise sessions at low (<60% VO2max), moderate (60-75% VO2max), and high (>90% VO2max) intensities. Blood samples were collected pre, immediately post, and 60 minutes post-exercise to analyze NEFA, triacylglycerol, non-HDL-c, HDL-c, PAI-1, leptin and adiponectin concentrations. Blood samples were collected from another set of twelve individuals for use in monocyte cell cultures to analyze L-CAT, CETP, and AMPK gene expressions. Low VO2max group pre-exercise exhibited higher postprandial leptin and total cholesterol concentrations than High VO2max group (p < 0.05). Exercise performed in high-intensity promoted a decreased leptin and NEFA levels (p < 0.05, for both), but for PAI-1 levels was decreased (p < 0.05) only for the Low VO2max group. Triacylglycerol levels decreased after all exercise sessions (p < 0.05) for both groups, and HDL-c exhibited decrease during moderate-intensity (p < 0.05), but this scenario was attenuated in Low VO2max group. Low VO2max individuals exhibit some metabolic-endocrine disruption, and acute aerobic exercise sessions performed at low, moderate, and high intensities are capable of modulating metabolic-endocrine parameters, mainly at high-intensity, in a physical fitness-dependent way, given that Low VO2max group was more responsive and seem to be able to appropriate more exercise-related benefits.
Collapse
Affiliation(s)
- B M Antunes
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil.
| | - F E Rossi
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| | - L M Oyama
- Universidade Federal de São Paulo, Escola Paulista de Medicina, Departamento de Fisiologia, São Paulo, Brazil
| | - J C Rosa-Neto
- Immunometabolism Research Group, Department of Cell Biology and Development, Institute of Biomeical Science of University of São Paulo (USP), São Paulo, Brazil
| | - F S Lira
- Exercise and Immunometabolism Research Group, Post-Graduation Program in Movement Sciences, Department of Physical Education, São Paulo State University (UNESP), Presidente Prudente, SP, Brazil
| |
Collapse
|
183
|
Rana T, Jiang C, Liu G, Miyata T, Antony V, Thannickal VJ, Liu RM. PAI-1 Regulation of TGF-β1-induced Alveolar Type II Cell Senescence, SASP Secretion, and SASP-mediated Activation of Alveolar Macrophages. Am J Respir Cell Mol Biol 2020; 62:319-330. [PMID: 31513752 PMCID: PMC7055702 DOI: 10.1165/rcmb.2019-0071oc] [Citation(s) in RCA: 88] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 09/11/2019] [Indexed: 12/16/2022] Open
Abstract
Senescence of alveolar type II (ATII) cells, progenitors of the alveolar epithelium, is a pathological feature and contributes importantly to the pathogenesis of idiopathic pulmonary fibrosis. Despite recognition of the importance of ATII cell senescence in idiopathic pulmonary fibrosis pathogenesis, how ATII cell senescence is regulated and how senescent ATII cells contribute to lung fibrogenesis remain unclear. In this study, we show that TGF-β1 (transforming growth factor-β1), a most ubiquitous and potent profibrotic cytokine, induces plasminogen activator inhibitor-1 (PAI-1), a cell senescence and fibrosis mediator, and p16 as well as senescence, but not apoptosis, in primary mouse ATII cells. We also found that senescent ATII cells secrete various cytokines and chemokines, including IL-4 and IL-13, which stimulate the expression of genes associated with a profibrotic phenotype in alveolar macrophages. Similar responses were also observed in TGF-β1-treated rat ATII (L2) and rat macrophage NR8383 cells. Deletion of PAI-1 or inhibition of PAI-1 activity with a small molecule PAI-1 inhibitor, however, blocks TGF-β1-induced senescence as well as a senescence-associated secretory phenotype in ATII and L2 cells and, consequently, the stimulatory effects of the conditioned medium from senescent ATII/L2 cells on macrophages. Moreover, we show that silencing p16 ameliorates PAI-1 protein-induced ATII cell senescence and secretion of profibrotic mediators. Our data suggest that PAI-1 mediates TGF-β1-induced ATII cell senescence and secretion of profibrotic mediators through inducing p16, and they also suggest that senescent ATII cells contribute to lung fibrogenesis in part by activating alveolar macrophages through secreting profibrotic and proinflammatory mediators.
Collapse
Affiliation(s)
- Tapasi Rana
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Chunsun Jiang
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Gang Liu
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Toshio Miyata
- United Centers for Advanced Research and Translational Medicine, Tohoku University, Tohoku, Japan
| | - Veena Antony
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Victor J. Thannickal
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Rui-Ming Liu
- Division of Pulmonary Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama; and
| |
Collapse
|
184
|
Synergy between plasminogen activator inhibitor-1, α-synuclein, and neuroinflammation in Parkinson's disease. Med Hypotheses 2020; 138:109602. [PMID: 32035284 DOI: 10.1016/j.mehy.2020.109602] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/24/2020] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a progressive degenerative nervous system disorder and is the second most common neurodegenerative disorder in the elderly population. The disease originates from the loss of dopamine-producing neurons in the substantia nigra in the brain, resulting in unregulated activity of the basal ganglia. Αlpha-synuclein (α-syn) is a protein found to aggregate in the substantia nigra region of patients with PD, forming Lewy Body inclusions; its aggregation may contribute to neuronal cell death in PD. This work hypothesizes about the synergistic relationship between α-syn aggregation and neuroinflammation to up-regulate expression of the serine protease inhibitor (serpin) plasminogen activator inhibitor-1 (PAI-1). The protease, plasmin, has been shown to cleave extracellular α-syn (including its monomeric, oligomeric, and fibrillary forms), resulting in less aggregation and Lewy Body formation. The zymogen plasminogen is converted to its active serine protease form, plasmin, either by tissue plasminogen activator (tPA) or by urokinase plasminogen activator (uPA) bound to urokinase receptor (uPAR). Both tPA and uPA/uPAR are inhibited by PAI-1. Thus, when PAI-1 levels increase, less plasmin is generated, which would lead to reduced proteolysis of α-syn. Expression of PAI-1 is increased both in inflammatory environments and in the presence of extracellular α-syn aggregates. This scenario suggests a pathological amplification loop: increased extracellular α-syn aggregation activates an inflammatory response from microglia and astrocytes, increasing PAI-1 levels, and decreasing the generation of plasmin. With reduced plasmin, less α-syn can be cleaved, and aggregation continues, sustaining the pathological process. Understanding this putative pathogenic loop could provide insight into the means by which neurodegeneration progresses in PD, and it may offer possible novel therapeutic strategies.
Collapse
|
185
|
de Miguel-Gómez L, Ferrero H, López-Martínez S, Campo H, López-Pérez N, Faus A, Hervás D, Santamaría X, Pellicer A, Cervelló I. Stem cell paracrine actions in tissue regeneration and potential therapeutic effect in human endometrium: a retrospective study. BJOG 2020; 127:551-560. [PMID: 31876085 DOI: 10.1111/1471-0528.16078] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/17/2019] [Indexed: 01/07/2023]
Abstract
OBJECTIVE Determining genetic and paracrine mechanisms behind endometrial regeneration in Asherman's syndrome and endometrial atrophy (AS/EA) patients after autologous CD133+ bone marrow-derived stem cell (CD133+ BMDSC) transplantation. DESIGN Retrospective study using human endometrial biopsies and mouse models. SETTING Fundación-IVI, IIS-La Fe, Valencia, Spain. SAMPLES Endometrial biopsies collected before and after CD133+ BMDSC therapy, from eight women with AS/EA (NCT02144987) from the uterus of five mice with only left horns receiving CD133+ BMDSC therapy. METHODS In human samples, haematoxylin and eosin (H&E) staining, RNA arrays, PCR validation, and neutrophil elastase (NE) immunohistochemistry (IHQ). In mouse samples, PCR validation and protein immunoarrays. MAIN OUTCOME MEASURES H&E microscopic evaluation, RNA expression levels, PCR, and growth/angiogenic factors quantification, NE IHQ signal. RESULTS Treatment improved endometrial morphology and thickness for all patients. In human samples, Jun, Serpine1, and Il4 were up-regulated whereas Ccnd1 and Cxcl8 were down-regulated after treatment. The significant decrease of NE signal corroborated Cxcl8 expression. Animal model analysis confirmed human results and revealed a higher expression of pro-angiogenic cytokines (IL18, HGF, MCP-1, MIP2) in treated uterine horns. CONCLUSIONS CD133+ BMDSC seems to activate several factors through a paracrine mechanism to help tissue regeneration, modifying endometrial behaviour through an immunomodulatory milieu that precedes proliferation and angiogenic processes. Insight into these processes could bring us one step closer to a non-invasive treatment for AS/EA patients. TWEETABLE ABSTRACT CD133+ BMDSC therapy regenerates endometrium, modifying the immunological milieu that precedes proliferation and angiogenesis.
Collapse
Affiliation(s)
- L de Miguel-Gómez
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - H Ferrero
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - S López-Martínez
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - H Campo
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - N López-Pérez
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - A Faus
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - D Hervás
- Data Science, Biostatistics and Bioinformatics, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - X Santamaría
- Igenomix Academy, Valencia, Spain.,IVIRMA, Barcelona, Barcelona, Spain
| | - A Pellicer
- IVIRMA Valencia, Valencia, Spain.,Reproductive Medicine Research Group, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - I Cervelló
- Fundación Instituto Valenciano de Infertilidad (FIVI), Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| |
Collapse
|
186
|
Geng B, Li S, Zhou J, Feng G. Correlation between PAI-1 rs1799889 polymorphism and venous thromboembolism: A meta-analysis of 48 case-control studies. Phlebology 2020; 35:472-479. [PMID: 31948344 DOI: 10.1177/0268355519897552] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
BACKGROUND We performed this meta-analysis to better assess the relationship between plasminogen activator inhibitor-1 (PAI-1) rs1799889 polymorphism and the risk of venous thromboembolism. METHOD Eligible studies were searched in PubMed, Medline, Embase, Web of Science and CNKI. Odds ratios with 95% confidence intervals were calculated. RESULT A total of 48 studies with 14,806 participants were analyzed. No positive results were detected in overall analyses. Further subgroup analyses revealed that the PAI-1 rs1799889 polymorphism was significantly associated with the risk of venous thromboembolism in Caucasians and East Asians. When we stratified available data according to type of disease, we found that the PAI-1 rs1799889 polymorphism was significantly correlated with the risk of DVT. No any other positive results were observed in overall and subgroup analyses. CONCLUSION Our findings indicate that the PAI-1 rs1799889 polymorphism may serve as a potential biological marker for venous thromboembolism in Caucasians and East Asians.
Collapse
Affiliation(s)
- Baoyu Geng
- Department of Cardiology, Taixing People's Hospital of Jiangsu Province, Taixing, China
| | - Shenghui Li
- Department of Cardiology, Taixing People's Hospital of Jiangsu Province, Taixing, China
| | - Jungao Zhou
- Department of Cardiology, Taixing People's Hospital of Jiangsu Province, Taixing, China
| | - Guangzhi Feng
- Department of Cardiology, Taixing People's Hospital of Jiangsu Province, Taixing, China
| |
Collapse
|
187
|
Ibrahim RR, Amer RA, Abozeid AA, Elsharaby RM, Shafik NM. Micro RNA 146a gene variant / TNF-α / IL-6 / IL-1 β; A cross-link axis inbetween oxidative stress, endothelial dysfunction and neuro-inflammation in acute ischemic stroke and chronic schizophrenic patients. Arch Biochem Biophys 2020; 679:108193. [DOI: 10.1016/j.abb.2019.108193] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/09/2019] [Accepted: 11/12/2019] [Indexed: 12/18/2022]
|
188
|
Mankowski RT, You L, Buford TW, Leeuwenburgh C, Manini TM, Schneider S, Qiu P, Anton SD. Higher dose of resveratrol elevated cardiovascular disease risk biomarker levels in overweight older adults - A pilot study. Exp Gerontol 2019; 131:110821. [PMID: 31891746 DOI: 10.1016/j.exger.2019.110821] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Revised: 12/06/2019] [Accepted: 12/26/2019] [Indexed: 02/05/2023]
Abstract
Older adults are at high risk of developing cardiovascular disease (CVD). Pre-clinical studies indicate that resveratrol (RSV), a polyphenol commonly found in grapes and red wine, may help prevent development of CVD. Based on our previous reports where the 300 mg and 1000 mg doses appeared safe and improved psychomotor function in a dose-dependent manner, our hypothesis was that RSV would reduce biomarkers of CVD risk in overweight, but otherwise healthy older adults and that 1000 mg would lower CVD biomarkers >300 mg. This analysis was performed on samples from older participants (65 years and older) who were randomized to a 90 day RSV treatment with 300 mg (n = 10), 1000 mg (n = 9) or placebo (n = 10). We measured levels of CVD risk biomarkers i.e. oxidized low-density lipoprotein (oxLDL), soluble E-selectin-1 (sE-selectin), soluble Intercellular Adhesion Molecule-1 (sICAM-1), Soluble Vascular Cell Adhesion Molecule-1 (sVCAM-1), total plasminogen activator inhibitor (tPAI-1). Statistical significance was set at p < 0.05. Both sVCAM-1 and tPAI increased significantly more in the 1000 mg vs. 300 mg and placebo groups. Other biomarkers (300 mg vs. 1000 mg vs. placebo: oxLDL, sEselectin-1 and sICAM-1) followed the same trend toward higher levels in the 1000 mg group compared to the 300 mg and placebo groups, without reaching statistical significance. This pilot project suggests that a higher dose of RSV may increase the levels of CVD risk biomarkers in overweight older adults. Given no change in the CVD risk biomarkers in response to a lower dose, future studies should test the effects of different doses of RSV to evaluate potential detrimental effects of higher doses on CVD biomarkers and measures of cardiovascular function in older adults at risk for CVD.
Collapse
Affiliation(s)
- R T Mankowski
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA.
| | - L You
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - T W Buford
- Department of Medicine, UAB School of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - C Leeuwenburgh
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA
| | - T M Manini
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA
| | - S Schneider
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA
| | - P Qiu
- Department of Biostatistics, University of Florida, Gainesville, FL, USA
| | - S D Anton
- Department of Aging and Geriatric Research, University of Florida, Gainesville, FL, USA
| |
Collapse
|
189
|
Yoon J, Um HN, Jang J, Bae YA, Park WJ, Kim HJ, Yoon MS, Chung IY, Jung Y. Eosinophil Activation by Toll-Like Receptor 4 Ligands Regulates Macrophage Polarization. Front Cell Dev Biol 2019; 7:329. [PMID: 31921842 PMCID: PMC6933835 DOI: 10.3389/fcell.2019.00329] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 11/26/2019] [Indexed: 01/21/2023] Open
Abstract
Eosinophils are terminally differentiated granulocytes that have long been considered as destructive cells associated with Th2 type immune responses such as allergic inflammation and helminth infections. Recently, eosinophils have been actively studied as multifunctional leukocytes regulating an array of physiological responses through interaction with other immune cells. In this study, we examined the expression and function of Toll-like receptors (TLRs) in eosinophilic EoL-1 cells and demonstrated the expression of a number of immune mediators in activated EoL-1 cells and their interaction with the macrophage cell line THP-1 upon TLR4 ligand stimulation. EoL-1 cells differentiated with butyrate increased expression of TLR3, TLR4, and TLR7 at mRNA and protein level with flow cytometry analysis. Mature eosinophils derived from human cord blood CD34+ cells were subjected to RNA-sequencing, and showed the expression of a panel of TLR transcripts and TLR4 was the most highly expressed TLR. Among the cognate ligands of TLR3, TLR4, and TLR7, lipopolysaccharide (LPS) or palmitic acid significantly increased mRNA expression of immune mediators in differentiated EoL-1 cells. Notably, Western blot analysis of palmitic acid-treated differentiated EoL-1 cells showed significantly up-regulated expression of Th2 type cytokines and transcription factors driving eosinophil differentiation. To evaluate functional significance of TLR4 ligand-stimulated eosinophils, we added conditioned media (CM) from EoL-1 cells to differentiated THP-1 cells and assessed the expression of M1 macrophage or M2 macrophage-related markers. M1 and M2 macrophage markers were significantly upregulated by CM from LPS and palmitic acid stimulated EoL-1 cells, respectively. In addition, the adipose tissue of obese mice, where eosinophils are decreased due to obesity-induced inflammation, showed significantly decreased frequency of M2 macrophages, despite an increase in the total macrophage numbers. Based on these collective data, we proposed that eosinophils regulate both inflammatory and anti-inflammatory polarization of macrophages through functional changes induced by different TLR4 ligands.
Collapse
Affiliation(s)
- Jiyoung Yoon
- Department of Microbiology, College of Medicine, Gachon University, Incheon, South Korea
| | - Han-Na Um
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| | - Jinsun Jang
- Department of Microbiology, College of Medicine, Gachon University, Incheon, South Korea.,Department of Dermatology, Gachon Gil Medical Center, College of Medicine, Gachon University, Incheon, South Korea
| | - Young-An Bae
- Department of Microbiology, College of Medicine, Gachon University, Incheon, South Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| | - Woo-Jae Park
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea.,Department of Biochemistry, College of Medicine, Gachon University, Incheon, South Korea
| | - Hee Joo Kim
- Department of Dermatology, Gachon Gil Medical Center, College of Medicine, Gachon University, Incheon, South Korea
| | - Mee-Sup Yoon
- Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea.,Department of Molecular Medicine, College of Medicine, Gachon University, Incheon, South Korea
| | - Il Yup Chung
- Department of Bionano Technology, Hanyang University, Ansan, South Korea
| | - YunJae Jung
- Department of Microbiology, College of Medicine, Gachon University, Incheon, South Korea.,Department of Health Sciences and Technology, GAIHST, Gachon University, Incheon, South Korea
| |
Collapse
|
190
|
Chu Y, Bucci JC, Peterson CB. Identification of a PAI-1-binding site within an intrinsically disordered region of vitronectin. Protein Sci 2019; 29:494-508. [PMID: 31682300 DOI: 10.1002/pro.3770] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Accepted: 10/28/2019] [Indexed: 12/14/2022]
Abstract
The serine protease inhibitor, plasminogen activator inhibitor Type-1 (PAI-1) is a metastable protein that undergoes an unusual transition to an inactive conformation with a short half-life of only 1-2 hr. Circulating PAI-1 is bound to a cofactor vitronectin, which stabilizes PAI-1 by slowing this latency conversion. A well-characterized PAI-1-binding site on vitronectin is located within the somatomedin B (SMB) domain, corresponding to the first 44 residues of the protein. Another PAI-1 recognition site has been identified with an engineered form of vitronectin lacking the SMB domain, yet retaining PAI-1 binding capacity (Schar, Blouse, Minor, Peterson. J Biol Chem. 2008;283:28487-28496). This additional binding site is hypothesized to lie within an intrinsically disordered domain (IDD) of vitronectin. To localize the putative binding site, we constructed a truncated form of vitronectin containing 71 amino acids from the N-terminus, including the SMB domain and an additional 24 amino acids from the IDD region. This portion of the IDD is rich in acidic amino acids, which are hypothesized to be complementary to several basic residues identified within an extensive vitronectin-binding site mapped on PAI-1 (Schar, Jensen, Christensen, Blouse, Andreasen, Peterson. J Biol Chem. 2008;283:10297-10309). Steady-state and stopped-flow fluorescence measurements demonstrate that the truncated form of vitronectin exhibits the same rapid biphasic association as full-length vitronectin and that the IDD hosts the elusive second PAI-1 binding site that lies external to the SMB domain of vitronectin.
Collapse
Affiliation(s)
- Yuzhuo Chu
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States
| | - Joel C Bucci
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States
| | - Cynthia B Peterson
- Department of Biological Sciences, Louisiana State University, Baton Rouge, Louisiana, United States
| |
Collapse
|
191
|
Huang G, Wang P, Li T, Deng X. Genetic association between plasminogen activator inhibitor-1 rs1799889 polymorphism and venous thromboembolism: Evidence from a comprehensive meta-analysis. Clin Cardiol 2019; 42:1232-1238. [PMID: 31701558 PMCID: PMC6906978 DOI: 10.1002/clc.23282] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2019] [Revised: 09/28/2019] [Accepted: 10/10/2019] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND Association between plasminogen activator inhibitor-1 (PAI-1) rs1799889 polymorphism and venous thromboembolism (VTE) were explored by many previous studies, yet the findings of these studies were conflicting. HYPOTHESIS PAI-1 rs1799889 polymorphism may serve as a genetic marker of VTE. We aimed to better clarify the relationship between PAI-1 rs1799889 polymorphism and VTE in a larger combined population by performing a meta-analysis. METHODS Literatures were searched in Pubmed, Embase, Web of Science, and China National Knowledge Infrastructure (CNKI). We used Review Manager to combine the results of individual studies. RESULTS Forty-eight studies involving 14 806 participants were eligible for inclusion. Combined results revealed that PAI-1 rs1799889 polymorphism was significantly associated with VTE in Caucasians (dominant comparison: odds ratio [OR] 1.20, 95% confidence interval [CI] 1.09-1.32; recessive comparison: OR 0.84, 95% CI 0.76-0.94; allele comparison: OR 1.08, 95% CI 1.02-1.15) and East Asians (dominant comparison: OR 1.60, 95% CI 1.17-2.19; allele comparison: OR 1.53, 95% CI 1.21-1.93). Further analyses obtained similar significant associations in these with deep vein thrombosis (DVT) and these with Factor V Leiden mutation. CONCLUSIONS Our findings supported that PAI-1 rs1799889 polymorphism may serve as one of the predisposing factors of VTE in both Caucasians and East Asians, especially in these with DVT and these with Factor V Leiden mutation.
Collapse
Affiliation(s)
- Guangbin Huang
- Department of Trauma Surgery, Emergency Medical Center of Chongqing, The Affiliated Central Hospital of Chongqing University, Chongqing, China
| | - Pan Wang
- Department of Trauma Surgery, Emergency Medical Center of Chongqing, The Affiliated Central Hospital of Chongqing University, Chongqing, China
| | - Tao Li
- Department of Trauma Surgery, Emergency Medical Center of Chongqing, The Affiliated Central Hospital of Chongqing University, Chongqing, China
| | - Xuejun Deng
- Department of Cardiology, Suining Central Hospital, Suining, China
| |
Collapse
|
192
|
Warren KJ, Poole JA, Sweeter JM, DeVasure JM, Wyatt TA. An association between MMP-9 and impaired T cell migration in ethanol-fed BALB/c mice infected with respiratory syncytial virus-2A. Alcohol 2019; 80:25-32. [PMID: 30291948 DOI: 10.1016/j.alcohol.2018.09.009] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 09/14/2018] [Accepted: 09/26/2018] [Indexed: 12/20/2022]
Abstract
Matrix metalloproteinases are important for proper airway matrix structure and wound healing. These enzymes are also implicated in many airway diseases. Previously, chronic ethanol consumption was shown to prolong inflammation and delay viral clearance in respiratory syncytial virus (RSV)-infected mice. We hypothesize that alcohol alters anti-viral immunity by disrupting immune cell chemotaxis in the lung. BALB/c mice were randomly selected to consume 18% alcohol ad libitum for 8 weeks prior to infection with RSV-2A. Bronchoalveolar lavage (BAL) cell populations were measured by flow cytometry, and chemokines were detected by Western blot or ELISA. MMP-9 levels were determined by polymerase chain reaction (PCR) in mouse lungs and in BAL fluid by ELISA. T cells were acquired from the spleens of water-fed, non-infected control mice (CTRL); alcohol-fed, non-infected (ETOH); water-fed, RSV-infected (RSV); or ethanol-fed, RSV-infected (ETOH-RSV) 4 days after RSV infection. T cells were placed in a transmigration system where chemokines had been treated with and without activated MMP-9. Lymphocyte recruitment was significantly reduced in the BAL 4 days after RSV infection in ETOH-RSV mice, whereas chemokine levels were the highest in this group at all experimental time points examined in comparison to RSV (p < 0.05). MMP-9 mRNA and protein were detected at high levels in ETOH-RSV mice compared to RSV. Using ex vivo transmigration to CCL2 and CXCL10, T cell migration was not impaired between any of the treatment groups, yet when CCL2 and CXCL10 were treated with activated MMP-9, significantly fewer T cells migrated across collagen-coated 5-μm membranes (p < 0.05). Immune cell recruitment is necessary for viral clearance. We show that immune cells are decreased in the lungs of ETOH-RSV mice. In contrast to decreased cell recruitment, key inflammatory chemokines were elevated in the lungs of ETOH-RSV mice. These proteins may be prematurely degraded by MMP-9 in the lung, leading to defective immunity and reduced viral clearance.
Collapse
Affiliation(s)
- Kristi J Warren
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Jill A Poole
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States
| | - Jenea M Sweeter
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Jane M DeVasure
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States.
| | - Todd A Wyatt
- University of Nebraska Medical Center, Pulmonary, Critical Care, Sleep & Allergy, 985910 Nebraska Medical Center, Omaha, NE, 68198-5910, United States; Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, 68105, United States; University of Nebraska Medical Center, Department of Environmental, Agricultural, & Occupational Health, Omaha, NE, 68198, United States.
| |
Collapse
|
193
|
Yildiz M, Bozkurtlar E, Azizy A, Agirbasli M. Immunohistochemical expression of plasminogen activator inhibitor-1 in subcutaneous versus omental adipose tissue in patients after elective abdominal surgery. AUTOPSY AND CASE REPORTS 2019; 9:e2019121. [PMID: 31641662 PMCID: PMC6771447 DOI: 10.4322/acr.2019.121] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 08/20/2019] [Indexed: 11/25/2022] Open
Abstract
Plasminogen activator inhibitor-1 (PAI-1) is a biomarker of thrombosis. Adipose and vascular tissues are among the major sources of PAI-1 production. Previous studies indicated that fat deposits mediate increased cardiovascular risk among obese individuals. We investigated the immunohistochemical (IHC) expression of PAI-1 in adipose and vascular tissues from the omentum and the subcutaneous tissue. The pathology samples were selected from 37 random patients who underwent elective abdominal surgery between 2008-2009. PAI-1 expression was semi-quantitatively scored and compared between the groups. Significant differences were noted in the IHC expression of PAI-1 between the omental and the subcutaneous adipose tissues (1.1 ± 0.8 versus 0.8 ± 0.6, respectively (p=0.05)). Adipose tissue displayed higher IHC expression of PAI-1 compared to vascular wall tissue in both omentum and subcutaneous sections (1.1 ± 0.8 versus 0.5 ± 0.9 (p=0.004), and 0.8 ± 0.6 versus 0.4 ± 0.6 (p=0.003), respectively). In conclusion, our study compared PAI-1 expression in the omentum versus the subcutaneous tissue and adipose versus vascular tissues. IHC expression of PAI-1 level was significantly higher in the omental adipose tissue compared to the subcutaneous adipose tissue. Adipose tissue displayed significantly higher PAI-1 expression than vascular tissue. The study elucidates the biological differences of adipose and vascular tissue from subcutaneous versus omental sections.
Collapse
Affiliation(s)
- Mehmet Yildiz
- Cleveland Clinic Fairview Hospital, Department of Internal Medicine. Cleveland, OH, USA
| | - Emine Bozkurtlar
- Marmara University Medical School, Department of Pathology. Istanbul, Turkey
| | - Abdulmunir Azizy
- Marmara University, Medical School, Department of Medicine. Istanbul, Turkey
| | - Mehmet Agirbasli
- Medeniyet University Medical School, Department of Cardiology. Istanbul, Turkey
| |
Collapse
|
194
|
Szegedi I, Nagy A, Székely EG, Czuriga-Kovács KR, Sarkady F, Lánczi LI, Berényi E, Csiba L, Bagoly Z. PAI-1 5G/5G genotype is an independent risk of intracranial hemorrhage in post-lysis stroke patients. Ann Clin Transl Neurol 2019; 6:2240-2250. [PMID: 31637872 PMCID: PMC6856768 DOI: 10.1002/acn3.50923] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/13/2019] [Accepted: 09/22/2019] [Indexed: 11/12/2022] Open
Abstract
Objective Thrombolysis by recombinant tissue plasminogen activator (rt‐PA) is the main pharmacological therapy in acute ischemic stroke (IS); however, it is only effective in a subset of patients. Here we aimed to investigate the role of plasminogen activator inhibitor‐1 (PAI‐1), an effective inhibitor of t‐PA, and its major polymorphism (PAI‐1 4G/5G) in therapy outcome. Methods Study population included 131 consecutive IS patients who all underwent thrombolysis. Blood samples were taken on admission, 1 and 24 h after rt‐PA infusion. PAI‐1 activity and antigen levels were measured from all blood samples and the PAI‐1 4G/5G polymorphism was determined. Clinical data including NIHSS were registered on admission and day 1. ASPECTS was assessed using CT images taken before and 24 h after thrombolysis. Intracranial hemorrhage (ICH) was classified according to ECASS II. Long‐term outcome was defined 90 days post‐event by the modified Rankin Scale (mRS). Results PAI‐1 activity levels dropped transiently after thrombolysis, while PAI‐1 antigen levels remained unchanged. PAI‐1 4G/5G polymorphism had no effect on PAI‐1 levels and did not influence stroke severity. PAI‐1 activity/antigen levels as measured on admission were significantly elevated in patients with worse 24 h ASPECTS (<7). Logistic regression analysis including age, sex, NIHSS on admission, BMI, history of arterial hypertension, and hyperlipidemia conferred a significant, independent risk for developing ICH in the presence of 5G/5G genotype (OR:4.75, 95%CI:1.18–19.06). PAI‐1 levels and PAI‐1 4G/5G polymorphism had no influence on long‐term outcomes. Interpretation PAI‐1 5G/5G genotype is associated with a significant risk for developing ICH in post‐lysis stroke patients.
Collapse
Affiliation(s)
- István Szegedi
- Faculty of Medicine, Department of Neurology, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt., Debrecen, 4032, Hungary
| | - Attila Nagy
- Faculty of Public Health, Department of Preventive Medicine, University of Debrecen, 26 Kassai út, Debrecen, 4032, Hungary
| | - Edina G Székely
- Faculty of Medicine, Department of Laboratory Medicine, Division of Clinical Laboratory Sciences, University of Debrecen, 98 Nagyerdei krt., Debrecen, 4032, Hungary
| | - Katalin R Czuriga-Kovács
- Faculty of Medicine, Department of Neurology, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt., Debrecen, 4032, Hungary
| | - Ferenc Sarkady
- Faculty of Medicine, Department of Laboratory Medicine, Division of Clinical Laboratory Sciences, University of Debrecen, 98 Nagyerdei krt., Debrecen, 4032, Hungary
| | - Levente I Lánczi
- Faculty of Medicine, Department of Radiology, University of Debrecen, 98 Nagyerdei krt., Debrecen, 4032, Hungary
| | - Ervin Berényi
- Faculty of Medicine, Department of Radiology, University of Debrecen, 98 Nagyerdei krt., Debrecen, 4032, Hungary
| | - László Csiba
- Faculty of Medicine, Department of Neurology, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt., Debrecen, 4032, Hungary.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, 22 Móricz Zsigmond krt., Debrecen, 4032, Hungary
| | - Zsuzsa Bagoly
- Faculty of Medicine, Department of Laboratory Medicine, Division of Clinical Laboratory Sciences, University of Debrecen, 98 Nagyerdei krt., Debrecen, 4032, Hungary.,MTA-DE Cerebrovascular and Neurodegenerative Research Group, 22 Móricz Zsigmond krt., Debrecen, 4032, Hungary
| |
Collapse
|
195
|
Patsouras MD, Vlachoyiannopoulos PG. Evidence of epigenetic alterations in thrombosis and coagulation: A systematic review. J Autoimmun 2019; 104:102347. [PMID: 31607428 DOI: 10.1016/j.jaut.2019.102347] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 10/03/2019] [Indexed: 02/07/2023]
Abstract
Thrombosis in the context of Cardiovascular disease (CVD) affects mainly the blood vessels supplying the heart, brain and peripheries and it is the leading cause of death worldwide. The pathophysiological thrombotic mechanisms are largely unknown. Heritability contributes to a 30% of the incidence of CVD. The remaining variation can be explained by life style factors such as smoking, dietary and exercise habits, environmental exposure to toxins, and drug usage and other comorbidities. Epigenetic variation can be acquired or inherited and constitutes an interaction between genes and the environment. Epigenetics have been implicated in atherosclerosis, ischemia/reperfusion damage and the cardiovascular response to hypoxia. Epigenetic regulators of gene expression are mainly the methylation of CpG islands, histone post translational modifications (PTMs) and microRNAs (miRNAs). These epigenetic regulators control gene expression either through activation or silencing. Epigenetic control is mostly dynamic and can potentially be manipulated to prevent or reverse the uncontrolled expression of genes, a trait that renders them putative therapeutic targets. In the current review, we systematically studied and present available data on epigenetic alterations implicated in thrombosis derived from human studies. Evidence of epigenetic alterations is observed in several thrombotic diseases such as Coronary Artery Disease and Cerebrovascular Disease, Preeclampsia and Antiphospholipid Syndrome. Differential CpG methylation and specific histone PTMs that control transcription of prothrombotic and proinflammatory genes have also been associated with predisposing factors of thrombosis and CVD, such us smoking, air pollution, hypertriglyceridemia, occupational exposure to particulate matter and comorbidities including cancer, Chronic Obstructive Pulmonary Disease and Chronic Kidney Disease. These clinical observations are further supported by in vitro experiments and indicate that epigenetic regulation affects the pathophysiology of thrombotic disorders with potential diagnostic or therapeutic utility.
Collapse
Affiliation(s)
- M D Patsouras
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece
| | - P G Vlachoyiannopoulos
- Department of Pathophysiology, School of Medicine, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
196
|
Yousuf SD, Ganie MA, Jeelani S, Mudassar S, Shah ZA, Zargar MA, Amin S, Wani IA, Rashid F. Effect of six-month use of oral contraceptive pills on plasminogen activator inhibitor-1 & factor VIII among women with polycystic ovary syndrome: An observational pilot study. Indian J Med Res 2019; 148:S151-S155. [PMID: 30964093 PMCID: PMC6469381 DOI: 10.4103/ijmr.ijmr_1899_17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Background & objectives: Polycystic ovary syndrome (PCOS) is an endocrinopathy warranting lifelong individualized management by lifestyle and pharmacological agents mainly oral contraceptive pills (OCPs). This study was aimed to report the impact of six-month OCP use on plasminogen activator inhibitor-1 (PAI-1) and factor VIII (FVIII) in women with PCOS. Methods: PCOS women diagnosed on the basis of Rotterdam 2003 criteria, either treated with OCPs (ethinyl estradiol-0.03 mg, levonorgestrel-0.15 mg) for a period of six months (n=40) or drug-naïve (n=42), were enrolled in this study. Blood was drawn to estimate glucose, insulin levels and lipid profile. Chemiluminescence immunoassays were used to measure hormones (LH, FSH, PRL, T4). Plasma levels of PAI-I and FVIII were measured by commercially available kits. Results: Menstrual regularity, Ferriman-Gallwey score and serum total testosterone significantly improved in the OCP group compared to drug-naïve group (P<0.01). No significant difference was observed in PAI-1 levels of the two groups; however, significant decrease in FVIII levels was observed in OCP group as compared to drug-naïve group. PAI-1 levels of OCP group correlated positively with blood glucose two hours, triglycerides and insulin two hours, while FVIII levels of OCP group correlated negatively with fasting insulin and homoeostatic model assessment-insulin resistance. Interpretation & conclusions: OCPs use has differential effect on pro-coagulant markers among women with PCOS. Well-designed, long-term, prospective, large-scale studies are prerequisite to elucidate the efficacy and safety of OCP in the treatment of PCOS.
Collapse
Affiliation(s)
- Syed Douhath Yousuf
- Department of Clinical Biochemistry/Biochemistry, University of Kashmir, Srinagar, India
| | - Mohammad Ashraf Ganie
- Department of Endocrinology & Metabolism, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Samoon Jeelani
- Department of Haematology, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Syed Mudassar
- Department of Clinical Biochemistry, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Zaffar Amin Shah
- Department of Immunology & Molecular Medicine, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Mohammad Afzal Zargar
- Department of Clinical Biochemistry/Biochemistry, University of Kashmir, Srinagar, India
| | - Shajrul Amin
- Department of Clinical Biochemistry/Biochemistry, University of Kashmir, Srinagar, India
| | - Imtiyaz Ahmad Wani
- Department of Endocrinology & Metabolism, Sher-i-Kashmir Institute of Medical Sciences, Srinagar, India
| | - Fouzia Rashid
- Department of Clinical Biochemistry/Biochemistry, University of Kashmir, Srinagar, India
| |
Collapse
|
197
|
Bu HQ, Shen F, Cui J. The inhibitory effect of oridonin on colon cancer was mediated by deactivation of TGF-β1/Smads-PAI-1 signaling pathway in vitro and vivo. Onco Targets Ther 2019; 12:7467-7476. [PMID: 31686852 PMCID: PMC6752205 DOI: 10.2147/ott.s220401] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Accepted: 08/27/2019] [Indexed: 12/30/2022] Open
Abstract
Background Oridonin, the main active component of Rabdosia rubescens, has been demonstrated to have anti-tumor effect on all kinds of cancer cells through various mechanisms and it has shown antitumor activity in some tumors partially via the suppression of TGF-β/Smads signaling pathway. The aim of this study was to explore the anticancer effect of oridonin on human colon carcinoma and underlying mechanism in vitro and vivo. Methods CCK-8 assay was employed to assess cell viability. The key target genes and proteins involved in TGF-β/Smads pathway was detected by RT-PCR, Western blotting and immunohistochemistry. The orthotopic transplantation tumor model of colon cance LOVO cell was introduced to detect anti-cancer effects in vivo. Results Oridonin inhibited the proliferation of colon cancer LOVO cells in a concentration and time dependent manner. In addition, oridonin reduced the levels of Smad2, Smad3, Smad4, PAI-1 and the phosphorylation of Smad2 and Smad3 induced by TGF-β1 in vitro. Subsequently, we established an orthotopically implanted tumor model in nude mice and found that oridonin treatment significantly suppressed tumor growth, and which was accompanied by the down-regulation of Smad2, Smad3, Smad4, PAI-1 and p-Smad2, p-Smad3 expression levels. Conclusion Our present study demonstrated that the growth inhibition of colon cancer by oridonin could be partially mediated through discontinuing TGF-β1/Smads-PAI-1 signaling pathway, suggesting it as a promising agent in treating colorectal cancer.
Collapse
Affiliation(s)
- He-Qi Bu
- Department of Coloproctological Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, People's Republic of China
| | - Feng Shen
- Department of Coloproctological Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, People's Republic of China
| | - Junhui Cui
- Department of Coloproctological Surgery, Tongde Hospital of Zhejiang Province, Hangzhou 310012, People's Republic of China
| |
Collapse
|
198
|
Diabetes drugs in the fight against Alzheimer's disease. Ageing Res Rev 2019; 54:100936. [PMID: 31330313 DOI: 10.1016/j.arr.2019.100936] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/20/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia, particularly in old age subjects. Hyperinsulinemia and insulin resistance, which are known as pathophysiological features of Type 2 Diabetes Mellitus (T2DM), have also been demonstrated to have a significant impact on cognitive impairment. Studies have shown that an altered insulin pathway may interact with amyloid-β protein deposition and tau protein phosphorylation, both leading factors for AD development. Drugs used for T2DM treatment from insulin and metformin through dipeptidyl peptidase-4 inhibitors and glucagon-like peptide-1 receptor agonists may represent a promising approach to fight AD. With this review from animal to human studies, we aim at responding to the reasons why drugs for diabetes may represent potential treatments for AD.
Collapse
|
199
|
Abolhasani S, Shahbazloo SV, Saadati HM, Mahmoodi N, Khanbabaei N. Evaluation of Serum Levels of Inflammation, Fibrinolysis and Oxidative Stress Markers in Coronary Artery Disease Prediction: A Cross-Sectional Study. Arq Bras Cardiol 2019; 113:667-674. [PMID: 31482948 PMCID: PMC7020864 DOI: 10.5935/abc.20190159] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/30/2019] [Indexed: 12/13/2022] Open
Abstract
Background Coronary Artery Disease (CAD) has long been recognized as a global health issue. Inflammation, Fibrinolysis and Oxidative Stress play an important role in the disruption of plaques leading to CAD. Markers that reflect this pathophysiologic mechanism may have prognostic value. Objective To estimate the serum concentrations of high-sensitivity C-reactive protein (hs-CRP), sialic acid (SA), vitronectin (VN), plasminogen activator inhibitor-1 (PAI-1), oxidized low density lipoprotein (OX-LDL) and malondialdehyde (MDA) with significant prognostic value in patients with CAD. Methods The markers included, hs-CRP, SA, VN, PAI-1, OX-LDL and MDA, were compared between 160 angiographically diagnosed CAD patients and 20 age- and sex-matched healthy individuals. The subjects were divided into 4 groups according to angiography results, and association between all risk factors of CAD was studied. Serum levels of SA, VN, PAI-1, and OX-LDL were measured by enzyme-linked immunosorbent assay (ELISA); MDA was measured based on reaction with thiobarbituric acid (TBA); and hs-CRP level was estimated by immunoturbidimetry using a commercial kit. The diagnostic value of these variables was further assessed by ROC curve analysis. Multiple logistic regression was used to evaluate the diagnostic power of the combination. Furthermore, p < 0.05 was considered as significant. Results Serum levels of hs-CRP, SA, VN, PAI-1, and OX-LDL were significantly higher in patient groups compared to control group (p < 0.001). Using both normal and CAD patients as subjects, ROC analysis was performed. The cutoff for OX-LDL, MDA, PAI-1, VN, hs-CRP and SA was 2.67 (ug/mL), 5.49 (mmol/mL), 67 (ng/mL), 254 (ng/mL), 3.4 (mg/dL), 7/89 (mg/dL), respectively. Eventually, the complete diagnostic efficacy was classified as: SA, hs-CRP, PAI-1, OX-LDL, MDA and VN. Conclusion Serum levels SA, hs-CRP, VN, PAI-1, OX-LDL and MDA may be predictive of adverse cardiovascular outcomes. Interestingly, these analyses can help as diagnostic and monitoring markers in CAD patients.
Collapse
Affiliation(s)
- Sakhavat Abolhasani
- Department of Clinical Biochemistry - Student Research Committee - School of Medicine - Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Department of Clinical Biochemistry - Faculty of Medicine - Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Hossein Mozafar Saadati
- Department of Epidemiology - School of Public Health and safety - Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Mahmoodi
- Department of Clinical Biochemistry - Faculty of Medicine - Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Khanbabaei
- Department of Clinical Biochemistry - Student Research Committee - School of Medicine - Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
200
|
Opara EI. Culinary herbs and spices: what can human studies tell us about their role in the prevention of chronic non-communicable diseases? JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2019; 99:4511-4517. [PMID: 30815875 DOI: 10.1002/jsfa.9658] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 02/15/2019] [Accepted: 02/24/2019] [Indexed: 05/16/2023]
Abstract
Culinary herbs and spices (CHS) are known primarily as flavour enhancers, and it is now well established that they possess bioactive properties that indicate that these foods may have a role to play in the prevention of non-communicable chronic diseases (CNCDs). Human studies are now beginning to provide insights into the significance of the potential health benefits of CHS in a dietary context, particularly concerning their antioxidant and anti-inflammatory properties and their impact on glucose homeostasis, appetite and the consumption of low/reduced fat, salt and sugar foods. However, these studies have also identified a number of factors that are very pertinent to furthering understanding of how CHS can be used for the maintenance of health and the prevention of CNCDs. The challenge for the next phase of studies will be how to incorporate, successfully, these factors into study methodology for investigating the preventative benefits of these foods. © 2019 Society of Chemical Industry.
Collapse
Affiliation(s)
- Elizabeth I Opara
- School of Life Sciences, Pharmacy and Chemistry, Kingston University, Kingston upon Thames, UK
| |
Collapse
|