1
|
Mahmoud EO, Elsabagh YA, Abd El Ghaffar N, Fawzy MW, Hussein MA. Atherosclerosis Associated With COVID-19: Acute, Tends to Severely Involve Peripheral Arteries, and May be Reversible. Angiology 2025; 76:77-84. [PMID: 37611951 DOI: 10.1177/00033197231198253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/25/2023]
Abstract
Arterial stiffness was reported with corona virus disease 2019 (COVID-19). We studied atherosclerosis in COVID-19 directly through duplex ultrasound measurements and their relation to co-morbidities, clinical and laboratory severity markers, and serum interleukin (IL) 6 and 17. Serum IL 6 and 17, average carotid intima-media thickness (cIMT), diameter and peak systolic velocities (PSV) of tibial, ulnar, radial arteries, and ankle brachial index (ABI) were measured in 44 COVID-19 patients and 44 healthy controls. Serum IL6, IL17, PSV, and cIMT were higher while diameter was lower (P ≤ .01) in cases. Clinical severity index correlated positively with age, co-morbidities, ferritin, IL6, IL17, cIMT, and PSV (P ≤ .04) and negatively with diameter and ABI (P = .04). Patients with severe lymphopenia had higher PSV, IL6, and IL17 and lower diameter (P < .00001). Ferritin positively correlated with PSV and negatively with diameter and ABI (P ≤ .01). Those who received an IL6 inhibitor (tocilizumab) showed lower PSV and higher diameter (P ≤ .01). In multiple regression analysis, IL17 and (age, co-morbidities) were related to (PSV, diameter) and cIMT (P ≤ .001, ≤0.02), respectively. COVID-19 may be associated with subclinical acute and may be reversible atherosclerosis severely involving peripheral arteries.
Collapse
Affiliation(s)
- Eman O Mahmoud
- Rheumatology and Clinical Immunology Unit, Internal Medicine Department, Cairo University, Cairo, Egypt
| | - Yumn A Elsabagh
- Rheumatology and Clinical Immunology Unit, Internal Medicine Department, Cairo University, Cairo, Egypt
| | | | - Mary Wadie Fawzy
- Rheumatology and Clinical Immunology Unit, Internal Medicine Department, Cairo University, Cairo, Egypt
| | - Mohamed A Hussein
- Rheumatology and Clinical Immunology Unit, Internal Medicine Department, Cairo University, Cairo, Egypt
| |
Collapse
|
2
|
Smith EE, Biessels GJ, Gao V, Gottesman RF, Liesz A, Parikh NS, Iadecola C. Systemic determinants of brain health in ageing. Nat Rev Neurol 2024; 20:647-659. [PMID: 39375564 DOI: 10.1038/s41582-024-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/09/2024]
Abstract
Preservation of brain health is a worldwide priority. The traditional view is that the major threats to the ageing brain lie within the brain itself. Consequently, therapeutic approaches have focused on protecting the brain from these presumably intrinsic pathogenic processes. However, an increasing body of evidence has unveiled a previously under-recognized contribution of peripheral organs to brain dysfunction and damage. Thus, in addition to the well-known impact of diseases of the heart and endocrine glands on the brain, accumulating data suggest that dysfunction of other organs, such as gut, liver, kidney and lung, substantially affects the development and clinical manifestation of age-related brain pathologies. In this Review, a framework is provided to indicate how organ dysfunction can alter brain homeostasis and promote neurodegeneration, with a focus on dementia. We delineate the associations of subclinical dysfunction in specific organs with dementia risk and provide suggestions for public health promotion and clinical management.
Collapse
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Virginia Gao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Neal S Parikh
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
3
|
Chen J, Wang B, Meng T, Li C, Liu C, Liu Q, Wang J, Liu Z, Zhou Y. Oxidative Stress and Inflammation in Myocardial Ischemia-Reperfusion Injury: Protective Effects of Plant-Derived Natural Active Compounds. J Appl Toxicol 2024. [PMID: 39482870 DOI: 10.1002/jat.4719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/12/2024] [Accepted: 10/18/2024] [Indexed: 11/03/2024]
Abstract
Acute myocardial infarction (AMI) remains a leading cause of death among patients with cardiovascular diseases. Percutaneous coronary intervention (PCI) has been the preferred clinical treatment for AMI due to its safety and efficiency. However, research indicates that the rapid restoration of myocardial oxygen supply following PCI can lead to secondary myocardial injury, termed myocardial ischemia-reperfusion injury (MIRI), posing a grave threat to patient survival. Despite ongoing efforts, the mechanisms underlying MIRI are not yet fully elucidated. Among them, oxidative stress and inflammation stand out as critical pathophysiological mechanisms, playing significant roles in MIRI. Natural compounds have shown strong clinical therapeutic potential due to their high efficacy, availability, and low side effects. Many current studies indicate that natural compounds can mitigate MIRI by reducing oxidative stress and inflammatory responses. Therefore, this paper reviews the mechanisms of oxidative stress and inflammation during MIRI and the role of natural compounds in intervening in these processes, aiming to provide a basis and reference for future research and development of drugs for treating MIRI.
Collapse
Affiliation(s)
- Jia Chen
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Boyu Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Tianwei Meng
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chengjia Li
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Qingnan Liu
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiameng Wang
- Heilongjiang University of Chinese Medicine, Harbin, China
| | - Zhiping Liu
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| | - Yabin Zhou
- The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
4
|
Jin L, Chen J, Wu L, Zhang M, Tang X, Shen C, Sun J, Du L, Wang X, Li Z. Central artery pulse pressure, not central arterial stiffness impact on all-cause mortality in patients with viral pneumonia infection. BMC Infect Dis 2024; 24:1183. [PMID: 39434023 PMCID: PMC11492499 DOI: 10.1186/s12879-024-10091-y] [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: 07/06/2023] [Accepted: 10/16/2024] [Indexed: 10/23/2024] Open
Abstract
OBJECTIVES COVID-19 viral pneumonia can result in increased arterial stiffness, along with cardiac and systemic inflammatory responses. This study aimed to investigate the association between arterial stiffness, inflammation severity, and all-cause mortality in patients with COVID-19. METHODS In this study, anthropometric data, pneumonia infection severity, and blood tests were analyzed. Arterial stiffness was assessed using the non-invasive assessment indices, including arterial velocity pulse index (AVI) and central arterial pulse pressure (CAPP). Infection volumes and percentages for the whole lungs, most lobes, and most segments were extracted from CT images using artificial intelligence-based quantitative analysis software. The relationship between arterial stiffness, central hemodynamics, and all-cause mortality was investigated. RESULTS In multivariable Cox regression analysis, high CAPP was significantly associated with all-cause mortality (hazard ratio: 0.263, 95% CI, 0.073-0.945, p = 0.041). Whole lung infection percentages were independently associated with high CAPP, with an area under the curve (AUC) of 0.662 and a specificity of 89.09%. CONCLUSIONS High CAPP, but not high AVI, demonstrated independent prognostic value for all-cause mortality in patients due to COVID-19 pneumonia infection. Evaluating this parameter could help in risk assessment and improve diagnostic and therapeutic strategies in viral pneumonia infections.
Collapse
Affiliation(s)
- Lin Jin
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Jiading District, Shanghai, 201803, China
- Department of Ultrasound, Guanghua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
| | - Jianxiong Chen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Jiading District, Shanghai, 201803, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Lingheng Wu
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Jiading District, Shanghai, 201803, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Mengjiao Zhang
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Jiading District, Shanghai, 201803, China
| | - Xiaobo Tang
- Department of Radiology, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201803, China
| | - Cuiqin Shen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Jiading District, Shanghai, 201803, China
| | - Jiali Sun
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Jiading District, Shanghai, 201803, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xifu Wang
- Department of Radiology, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201803, China
| | - Zhaojun Li
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, 800 Huangjiahuayuan Road, Jiading District, Shanghai, 201803, China.
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
5
|
Salvado R, Santos-Minguez S, Lugones-Sánchez C, Gonzalez-Sánchez S, Tamayo-Morales O, Quesada-Rico JA, Benito R, Rodríguez-Sánchez E, Gómez-Marcos MA, Casado-Vicente V, Guimarães-Cunha P, Hernandez-Rivas JM, Mira A, García-Ortiz L. Gut microbiota and its relationship with early vascular ageing in a Spanish population (MIVAS study). Eur J Clin Invest 2024; 54:e14228. [PMID: 38655910 DOI: 10.1111/eci.14228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 04/01/2024] [Accepted: 04/06/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Gut microbiota and its by-products are increasingly recognized as having a decisive role in cardiovascular diseases. The aim is to study the relationship between gut microbiota and early vascular ageing (EVA). METHODS A cross-sectional study was developed in Salamanca (Spain) in which 180 subjects aged 45-74 years were recruited. EVA was defined by the presence of at least one of the following: carotid-femoral pulse wave velocity (cf-PWV), cardio-ankle vascular index (CAVI) or brachial-ankle pulse wave velocity (ba-PWV) above the 90th percentile of the reference population. All other cases were considered normal vascular ageing (NVA). MEASUREMENTS cf-PWV was measured by SphygmoCor® System; CAVI and ba-PWV were determined by Vasera 2000® device. Gut microbiome composition in faecal samples was determined by 16S rRNA Illumina sequencing. RESULTS Mean age was 64.4 ± 6.9 in EVA group and 60.4 ± 7.6 years in NVA (p < .01). Women in EVA group were 41% and 53% in NVA. There were no differences in the overall composition of gut microbiota between the two groups when evaluating Firmicutes/Bacteriodetes ratio, alfa diversity (Shannon Index) and beta diversity (Bray-Curtis). Bilophila, Faecalibacterium sp.UBA1819 and Phocea, are increased in EVA group. While Cedecea, Lactococcus, Pseudomonas, Succiniclasticum and Dielma exist in lower abundance. In logistic regression analysis, Bilophila (OR: 1.71, 95% CI: 1.12-2.6, p = .013) remained significant. CONCLUSIONS In the studied Spanish population, early vascular ageing is positively associated with gut microbiota abundance of the genus Bilophila. No relationship was found between phyla abundance and measures of diversity.
Collapse
Affiliation(s)
- Rita Salvado
- Unidad de Investigación de Atención Primaria de Salamanca (APISAL), Gerencia de Atención Primaria de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Sandra Santos-Minguez
- Instituto de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de Salamanca-CSIC, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Cristina Lugones-Sánchez
- Unidad de Investigación de Atención Primaria de Salamanca (APISAL), Gerencia de Atención Primaria de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Red de Investigación en Cronicidad Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Barcelona, Spain
| | - Susana Gonzalez-Sánchez
- Unidad de Investigación de Atención Primaria de Salamanca (APISAL), Gerencia de Atención Primaria de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Red de Investigación en Cronicidad Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Barcelona, Spain
| | - Olaya Tamayo-Morales
- Unidad de Investigación de Atención Primaria de Salamanca (APISAL), Gerencia de Atención Primaria de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Red de Investigación en Cronicidad Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Barcelona, Spain
| | - José A Quesada-Rico
- Red de Investigación en Cronicidad Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Barcelona, Spain
- Facultad de Medicina, Universidad Miguel Hernández de Elche, Sant Joan, D'Alacant, Spain
| | - Rocío Benito
- Instituto de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de Salamanca-CSIC, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Emiliano Rodríguez-Sánchez
- Unidad de Investigación de Atención Primaria de Salamanca (APISAL), Gerencia de Atención Primaria de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Red de Investigación en Cronicidad Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Barcelona, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Manuel A Gómez-Marcos
- Unidad de Investigación de Atención Primaria de Salamanca (APISAL), Gerencia de Atención Primaria de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Red de Investigación en Cronicidad Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Barcelona, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
| | - Verónica Casado-Vicente
- Centro de Salud Parquesol. Gerencia de Salud Valladolid Oeste, Gerencia Regional de Salud de Castilla y Leon (SACyL), Valladolid, Spain
- Departamento de Medicina, Dermatología and Toxicología, Universidad de Valladolid, Valladolid, Spain
| | - Pedro Guimarães-Cunha
- Life and Health Sciences Research Institute (IICVS) and School of Medicine, Universidade do Minho, Braga, Portugal
- Center for the Research and Treatment of arterial Hypertension and cardiovascular Risk, Hospital Senhora da Oliveira, Guimarães, Portugal
| | - Jesús M Hernandez-Rivas
- Instituto de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Universidad de Salamanca-CSIC, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Medicina, Universidad de Salamanca, Salamanca, Spain
- Departamento de Hematología, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Alex Mira
- Departamento de Salud y Genómica, Fundación FISABIO, Valencia, Spain
- CIBER Centro de Epidemiología y Salud Pública, Madrid, Spain
| | - Luis García-Ortiz
- Unidad de Investigación de Atención Primaria de Salamanca (APISAL), Gerencia de Atención Primaria de Salamanca, Gerencia Regional de Salud de Castilla y León (SACYL), Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Red de Investigación en Cronicidad Atención Primaria y Prevención y Promoción de la Salud (RICAPPS), Barcelona, Spain
- Departamento de Ciencias Biomédicas y del Diagnóstico, Universidad de Salamanca, Salamanca, Spain
| |
Collapse
|
6
|
Gardikioti V, Georgakopoulos C, Solomou E, Lazarou E, Fasoulakis K, Terentes-Printzios D, Tsioufis K, Iliopoulos D, Vlachopoulos C. Effect of FluoRoquinolones on Aortic Growth, aortic stIffness and wave refLEctionS (FRAGILES study). Life (Basel) 2024; 14:992. [PMID: 39202735 PMCID: PMC11355559 DOI: 10.3390/life14080992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Background: The widespread use of fluoroquinolones has been associated with the formation, dissection, and rupture of aortic aneurysms. Arterial biomarkers are established predictors of cardiovascular events. The present study was designed to investigate the effect of quinolones on arterial stiffness and aortic size for the first time. Methods: We studied 28 subjects receiving short-term (<15 days) antibiotic therapy involving quinolones and 27 age- and sex-matched subjects receiving an alternative to quinolone antibiotics. The follow-up period was approximately 2 months. The study's primary endpoint was the carotid-femoral pulse wave velocity (cfPWV) difference between the two groups 2 months after therapy initiation. Secondary endpoints were the augmentation index corrected for heart rate (AIx@75) and sonographically assessed aortic diameters 2 months after the initial treatment. Results: Subjects had similar values of arterial biomarkers, blood pressure measurements, and aortic diameters at baseline. At follow-up, no significant change was observed between the two groups regarding the hemodynamic parameters and arterial biomarkers (p > 0.05 for all), i.e., cfPWV (7.9 ± 2.6 m/s for the control group vs. 8.1 ± 2.4 m/s for the fluoroquinolones group; p = 0.79), AIx@75 (22.6 ± 9.0% for the control group vs. 26.6 ± 8.1% for the fluoroquinolones group; p = 0.09), and aortic diameters. Conclusions: To our knowledge, FRAGILES is the first study to provide insights into the possible effects of fluoroquinolones on arterial biomarkers, showing that, at least in the short term, treatment with fluoroquinolones does not affect aortic function and diameter.
Collapse
Affiliation(s)
- Vasiliki Gardikioti
- First Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Christos Georgakopoulos
- First Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eirini Solomou
- First Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Emilia Lazarou
- First Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | | | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Dimitrios Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Charalambos Vlachopoulos
- First Department of Cardiology, Hippokration General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| |
Collapse
|
7
|
Mustonen T, Kanerva M, Luukkonen R, Lantto H, Uusitalo A, Piirilä P. Cardiopulmonary exercise testing in long covid shows the presence of dysautonomia or chronotropic incompetence independent of subjective exercise intolerance and fatigue. BMC Cardiovasc Disord 2024; 24:413. [PMID: 39117999 PMCID: PMC11308233 DOI: 10.1186/s12872-024-04081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND After COVID-19 infection, 10-20% of patients suffer from varying symptoms lasting more than 12 weeks (Long COVID, LC). Exercise intolerance and fatigue are common in LC. The aim was to measure the maximal exercise capacity of the LC patients with these symptoms and to analyze whether this capacity was related to heart rate (HR) responses at rest and during exercise and recovery, to find out possible sympathetic overactivity, dysautonomia or chronotropic incompetence. METHODS Cardiopulmonary exercise test was conducted on 101 LC patients, who were admitted to exercise testing. The majority of them (86%) had been treated at home during their acute COVID-19 infection. Peak oxygen uptake (VO2peak), maximal power during the last 4 min of exercise (Wlast4), HRs, and other exercise test variables were compared between those with or without subjective exercise intolerance, fatigue, or both. RESULTS The measurements were performed in mean 12.7 months (SD 5.75) after COVID-19 infection in patients with exercise intolerance (group EI, 19 patients), fatigue (group F, 31 patients), their combination (group EI + F, 37 patients), or neither (group N, 14 patients). Exercise capacity was, in the mean, normal in all symptom groups and did not significantly differ among them. HRs were higher in group EI + F than in group N at maximum exercise (169/min vs. 158/min, p = 0.034) and 10 min after exercise (104/min vs. 87/min, p = 0.028). Independent of symptoms, 12 patients filled the criteria of dysautonomia associated with slightly decreased Wlast4 (73% vs. 91% of sex, age, height, and weight-based reference values p = 0.017) and 13 filled the criteria of chronotropic incompetence with the lowest Wlast4 (63% vs. 93%, p < 0.001), VO2peak (70% vs. 94%, p < 0.001), the lowest increase of systolic blood pressure (50 mmHg vs. 67 mmHg, p = 0.001), and the greatest prevalence of slight ECG-findings (p = 0.017) compared to patients without these features. The highest prevalence of chronotropic incompetence was seen in the group N (p = 0.022). CONCLUSIONS This study on LC patients with different symptoms showed that cardiopulmonary exercise capacity was in mean normal, with increased sympathetic activity in most patients. However, we identified subgroups with dysautonomia or chronotropic incompetence with a lowered exercise capacity as measured by Wlast4 or VO2peak. Subjective exercise intolerance and fatigue poorly foresaw the level of exercise capacity. The results could be used to plan the rehabilitation from LC and for selection of the patients suitable for it.
Collapse
Affiliation(s)
- Timo Mustonen
- Department of Clinical Physiology, Peijas Hospital, HUS Medical Diagnostic Center, Helsinki University Hospital and Helsinki University, Stenbäckinkatu 11 C, PL 281, Helsinki, 00029, Finland.
| | - Mari Kanerva
- Department of Internal Medicine and Rehabilitation, Helsinki University and Helsinki University Hospital, Helsinki, Finland
- Department of Infection Control, Turku University Hospital, The wellbeing services county of Southwest Finland, Turku, Finland
| | | | - Hanna Lantto
- Department of Clinical Physiology, Park Hospital, HUS Medical Diagnostic Center, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Arja Uusitalo
- Division of Clinical Physiology and Nuclear Medicine, HUS Medical Diagnostic Center, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| | - Päivi Piirilä
- Department of Clinical Physiology, Park Hospital, HUS Medical Diagnostic Center, Helsinki University Hospital and Helsinki University, Helsinki, Finland
| |
Collapse
|
8
|
Cavinato C, Spronck B, Caulk AW, Murtada SI, Humphrey JD. AT1b receptors contribute to regional disparities in angiotensin II mediated aortic remodelling in mice. J R Soc Interface 2024; 21:20240110. [PMID: 39192727 PMCID: PMC11350382 DOI: 10.1098/rsif.2024.0110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/21/2024] [Accepted: 07/26/2024] [Indexed: 08/29/2024] Open
Abstract
The renin-angiotensin system plays a key role in regulating blood pressure, which has motivated many investigations of associated mouse models of hypertensive arterial remodelling. Such studies typically focus on histological and cell biological changes, not wall mechanics. This study explores tissue-level ramifications of chronic angiotensin II infusion in wild-type (WT) and type 1b angiotensin II (AngII) receptor null (Agtr1b -/-) mice. Biaxial biomechanical and immunohistological changes were quantified and compared in the thoracic and abdominal aorta in these mice following 14 and 28 days of angiotensin II infusion. Preliminary results showed that changes were largely independent of sex. Associated thickening and stiffening of the aortic wall in male mice differed significantly between thoracic and abdominal regions and between genotypes. Notwithstanding multiple biomechanical changes in both WT and Agtr1b -/- mice, AngII infusion caused distinctive wall thickening and inflammation in the descending thoracic aorta of WT, but not Agtr1b -/-, mice. Our study underscores the importance of exploring differential roles of receptor-dependent angiotensin II signalling along the aorta and its influence on distinct cell types involved in regional histomechanical remodelling. Disrupting the AT1b receptor primarily affected inflammatory cell responses and smooth muscle contractility, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- Cristina Cavinato
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- LMGC, Univ. Montpellier, CNRS, Montpellier, France
| | - Bart Spronck
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Department of Biomedical Engineering, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University, Maastricht, The Netherlands
| | - Alexander W. Caulk
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Sae-Il Murtada
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| | - Jay D. Humphrey
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Vascular Biology and Therapeutics Program, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
9
|
Agbo LD, Girerd N, Lamiral Z, Duarte K, Bozec E, Merckle L, Hoge A, Guillaume M, Laville M, Nazare JA, Rossignol P, Boivin JM, Wagner S. Dietary inflammatory potential and arterial stiffness in a French cohort: Insights from the STANISLAS study. Nutr Metab Cardiovasc Dis 2024; 34:1959-1967. [PMID: 38677885 DOI: 10.1016/j.numecd.2024.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 04/29/2024]
Abstract
BACKGROUND AND AIMS Chronic inflammation plays a key role in arterial stiffness pathogenesis. Dietary components can display anti- or pro-inflammatory properties. Nonetheless, the association between the diet's overall inflammatory potential and arterial stiffness is unclear. This study aimed to assess the association between the diet's overall inflammatory potential and arterial stiffness assessed by carotid-femoral pulse wave velocity (cfPWV). METHODS AND RESULTS This cross-sectional study included 1307 participants from the STANISLAS family cohort study. Dietary data were collected using a validated food frequency questionnaire. The adapted dietary inflammatory index (ADII) score was calculated to assess the inflammatory potential of the participants' diet. The association of ADII score quartile with cfPWV was assessed using IPW-weighted linear mixed models with random family effect. The median (Q1-Q3) ADII score was 0.45 (-1.57, 2.04). Participants exhibiting higher ADII scores demonstrated elevated energy intake, dietary saturated fat, and ultra-processed foods. Conversely, individuals with lower ADII scores exhibited higher vitamins and omega intakes, and a higher diet quality, as assessed by the DASH score. Despite these observations from the descriptive analyses, ADII score quartiles were not significantly associated with cfPWV (β(95% CI) were 0.01 (-0.02,0.04) for Q2, 0.02 (-0.01,0.05) for Q3, and 0.02 (-0.01,0.05) for Q4 compared to Q1). CONCLUSION In this cross-sectional study, participants had a relatively modest consumption of pro-inflammatory foods, no substantial associations were observed between the diet inflammatory potential and arterial stiffness. Further longitudinal studies in larger cohorts are needed to better understand the link between inflammatory diet and arterial stiffness.
Collapse
Affiliation(s)
- Louis-Désiré Agbo
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France
| | - Nicolas Girerd
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France.
| | - Zohra Lamiral
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France
| | - Kevin Duarte
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France
| | - Erwan Bozec
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France
| | - Ludovic Merckle
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France
| | - Axelle Hoge
- Département des Sciences de la Santé Publique, Université de Liège, Liège, Belgium
| | - Michèle Guillaume
- Département des Sciences de la Santé Publique, Université de Liège, Liège, Belgium
| | - Martine Laville
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Hospices Civils de Lyon, F-CRIN/FORCE Network, Pierre-Bénite, France
| | - Julie-Anne Nazare
- Centre de Recherche en Nutrition Humaine Rhône-Alpes, Univ-Lyon, CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Hospices Civils de Lyon, F-CRIN/FORCE Network, Pierre-Bénite, France
| | - Patrick Rossignol
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France; Medicine and Nephrology-Dialysis Departments, Princess Grace Hospital, and Monaco Private Hemodialysis Centre, Monaco, Monaco
| | - Jean-Marc Boivin
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France; Department of General Medicine, University of Lorraine, Vandoeuvre-lès-Nancy, France
| | - Sandra Wagner
- INSERM CIC 1433, Nancy CHRU, Inserm U1116, FCRIN, INI-CRCT, University of Lorraine, Nancy, France
| |
Collapse
|
10
|
Durieux JC, Zisis SN, Mouchati C, Labbato D, Abboud M, McComsey GA. Sex Modifies the Effect of COVID-19 on Arterial Elasticity. Viruses 2024; 16:1089. [PMID: 39066250 PMCID: PMC11281515 DOI: 10.3390/v16071089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/28/2024] Open
Abstract
There is limited long-term evidence on the effects of COVID-19 on vascular injury between male and female sex. An adult cohort of COVID-19 survivors (COVID+) and confirmed SARS-CoV-2 antibody-negative participants (COVID-) were prospectively enrolled. COVID+ participants who have documented the presence of persistent symptoms four weeks following infection were considered to have post-acute sequelae of COVID-19 (PASC). Non-invasive, FDA-approved EndoPAT (Endo-PAT2000) was used for endothelial assessment. COVID-(n = 94) were 1:1 propensity score matched to COVID+ (n = 151) on baseline covariates including sex. Among COVID+, 66.2% (n = 100) had PASC. Higher levels of coagulation marker, D-dimer (p = 0.001), and gut permeability marker, zonulin (p = 0.001), were associated with female sex. Estimated differences in augmentation index (AI) between COVID- (0.9 ± 17.2) and COVID+ (8.4 ± 15.7; p = 0.001) and between female and male sex (12.9 ± 1.9; p < .0001) were observed. Among COVID+ with PASC, the average AI (10.5 ± 1.6) was 9.7 units higher than COVID- (p < .0001) and 6.2 units higher compared to COVID+ with no PASC (p = 0.03). COVID+ PASC+ female sex had the highest AI (14.3 ± 1.9). The effects of SARS-CoV-2 infection on vascular function varies across strata of sex and female sex in the post-acute phase of COVID-19 have the worse arterial elasticity (highest AI).
Collapse
Affiliation(s)
- Jared C. Durieux
- University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.)
| | - Sokratis N. Zisis
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (S.N.Z.); (C.M.)
| | - Christian Mouchati
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (S.N.Z.); (C.M.)
| | - Danielle Labbato
- University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.)
| | - Marc Abboud
- Faculty of Medicine, Saint Joseph University of Beirut, Beirut 1104 2020, Lebanon;
| | - Grace A. McComsey
- University Hospitals Cleveland Medical Center, Cleveland, OH 44106, USA; (J.C.D.); (D.L.)
- School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; (S.N.Z.); (C.M.)
| |
Collapse
|
11
|
Mouhanni S, Hassani AA, Lekehal M, Bounssir A, Bakkali T, Lekehal B. Ruptured abdominal aortic aneurysm in psoriasis: A case report and review of literature. Int J Surg Case Rep 2024; 120:109829. [PMID: 38870660 PMCID: PMC11225200 DOI: 10.1016/j.ijscr.2024.109829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/22/2024] [Accepted: 05/25/2024] [Indexed: 06/15/2024] Open
Abstract
INTRODUCTION Psoriasis, a chronic inflammatory skin condition, affects 4 % of the population and is associated with various comorbidities, making it a public health concern. CASE REPORT We discuss the case of a 50-year-old man with severe erythrodermic psoriasis who presented with a ruptured saccular abdominal aortic aneurysm (AAA), requiring emergency surgery with good postoperative follow-up. shedding light on the link between psoriasis and cardiovascular complications. DISCUSSION Psoriasis severity correlates with cardiovascular risk and shares common development pathways with aortic aneurysms such as systemic and aortic inflammation, and arterial stiffness, emphasizing the importance of managing both skin symptoms and systemic inflammation to reduce vascular comorbidities. Psoriasis patients have a higher risk of AAA, warranting consideration for AAA screening. Controversies exist regarding corticosteroid therapy and choice of surgical intervention for AAA in psoriatic patients. CONCLUSION Psoriasis patients face an increased risk of AAA, highlighting the need for vigilant screening and comprehensive management. Further research is essential to understanding the pathophysiological connections between psoriasis and arterial diseases, guiding preventive strategies and optimal medical treatments for these high-risk patients.
Collapse
Affiliation(s)
- Safaa Mouhanni
- Mohammed V University in Rabat, Rabat, Morocco; Vascular Surgery Department, Ibn Sina University Hospital Centre, Rabat 10104, Morocco.
| | - Amine Azami Hassani
- Mohammed V University in Rabat, Rabat, Morocco; Vascular Surgery Department, Ibn Sina University Hospital Centre, Rabat 10104, Morocco
| | - Mehdi Lekehal
- Mohammed V University in Rabat, Rabat, Morocco; Vascular Surgery Department, Ibn Sina University Hospital Centre, Rabat 10104, Morocco
| | - Ayoub Bounssir
- Mohammed V University in Rabat, Rabat, Morocco; Vascular Surgery Department, Ibn Sina University Hospital Centre, Rabat 10104, Morocco
| | - Tarik Bakkali
- Mohammed V University in Rabat, Rabat, Morocco; Vascular Surgery Department, Ibn Sina University Hospital Centre, Rabat 10104, Morocco
| | - Brahim Lekehal
- Mohammed V University in Rabat, Rabat, Morocco; Vascular Surgery Department, Ibn Sina University Hospital Centre, Rabat 10104, Morocco
| |
Collapse
|
12
|
Lu G, Guo H, Zhang Y, Zhang M, Zhang T, Hu G, Zhang Q. Graphene Far-Infrared Irradiation Can Effectively Relieve the Blood Pressure Level of Rat Untr-HT in Primary Hypertension. Int J Mol Sci 2024; 25:6675. [PMID: 38928382 PMCID: PMC11204347 DOI: 10.3390/ijms25126675] [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: 05/27/2024] [Revised: 06/14/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
Graphene, when electrified, generates far-infrared radiation within the wavelength range of 4 μm to 14 μm. This range closely aligns with the far-infrared band (3 μm to 15 μm), which produces unique physiological effects. Contraction and relaxation of vascular smooth muscle play a significant role in primary hypertension, involving the nitric oxide-soluble guanylate cyclase-cyclic guanosine monophosphate pathway and the renin-angiotensin-aldosterone system. This study utilized spontaneously hypertensive rats (SHRs) as an untr-HT to investigate the impact of far-infrared radiation at specific wavelengths generated by electrified graphene on vascular smooth muscle and blood pressure. After 7 weeks, the blood pressure of the untr-HT group rats decreased significantly with a notable reduction in the number of vascular wall cells and the thickness of the vascular wall, as well as a decreased ratio of vessel wall thickness to lumen diameter. Additionally, blood flow perfusion significantly increased, and the expression of F-actin in vascular smooth muscle myosin decreased significantly. Serum levels of angiotensin II (Ang-II) and endothelin 1 (ET-1) were significantly reduced, while nitric oxide synthase (eNOS) expression increased significantly. At the protein level, eNOS expression decreased significantly, while α-SMA expression increased significantly in aortic tissue. At the gene level, expressions of eNOS and α-SMA in aortic tissue significantly increased. Furthermore, the content of nitric oxide (NO) in the SHR's aortic tissue increased significantly. These findings confirm that graphene far-infrared radiation enhances microcirculation, regulates cytokines affecting vascular smooth muscle contraction, and modifies vascular morphology and smooth muscle phenotype, offering relief for primary hypertension.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Qian Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, Beijing 102206, China; (G.L.); (H.G.); (Y.Z.); (M.Z.); (T.Z.); (G.H.)
| |
Collapse
|
13
|
Taibi M, Elbouzidi A, Haddou M, Baraich A, Loukili EH, Moubchir T, Allali A, Amine khoulati, Bellaouchi R, Asehraou A, Addi M, Salamatullah AM, Bourhia M, Siddique F, El Guerrouj B, Chaabane K. Phytochemical characterization and multifaceted bioactivity assessment of essential oil from Ptychotis verticillata Duby: Anti-diabetic, anti-tyrosinase, and anti-inflammatory activity. Heliyon 2024; 10:e29459. [PMID: 38699706 PMCID: PMC11063393 DOI: 10.1016/j.heliyon.2024.e29459] [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: 01/11/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 05/05/2024] Open
Abstract
The aim of this study is to explore the pharmacological properties of the essential oil derived from Ptychotis verticillata Duby (PVEO), a medicinal plant native to Morocco, focusing on its antidiabetic, anti-tyrosinase, and anti-inflammatory effects. Additionally, the study aims to characterize the phytochemical composition of PVEO and evaluate its potential as a natural therapeutic alternative for various health conditions. To achieve this, phytochemical analysis was conducted using gas chromatography-mass spectrometry (GC-MS). Furthermore, in vitro assessments were conducted to investigate PVEO's antidiabetic activity by inhibiting α-amylase, xanthine oxidase, and α-glucosidase. Tests were also undertaken to evaluate the anti-inflammatory effect of PVEO on RAW 264.7 cells stimulated by lipopolysaccharide (LPS), as well as its efficacy as an anti-tyrosinase agent and its lipoxygenase inhibition activity. The results of the phytochemical analysis revealed that PVEO is rich in terpene compounds, with percentages of 40.35 % γ-terpinene, 22.40 % carvacrol, and 19.77 % β-cymene. Moreover, in vitro evaluations demonstrated that PVEO exhibits significant inhibitory activity against α-amylase, xanthine oxidase, and α-glucosidase, indicating promising antidiabetic, and anti-gout potential. Furthermore, PVEO showed significant anti-tyrosinase activity, with an IC50 of 27.39 ± 0.44 μg/mL, and remarkable lipoxygenase inhibition (87.33 ± 2.6 %), suggesting its candidacy for dermatoprotection. Additionally, PVEO displayed a dose-dependent capacity to attenuate the production of NO and PGE2, two inflammatory mediators implicated in various pathologies, without compromising cellular viability. The findings of this study provide a solid foundation for future research on natural therapies and the development of new drugs, highlighting the therapeutic potential of PVEO in the treatment of gout, diabetes, pigmentation disorders, and inflammation.
Collapse
Affiliation(s)
- Mohamed Taibi
- Laboratoire d’Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Faculté des Sciences, Université Mohammed Premier, Oujda, 60000, Morocco
- Centre de L’Oriental des Sciences et Technologies de L’Eau et de L’Environnement (COSTEE), Université Mohammed Premier, Oujda, 60000, Morocco
| | - Amine Elbouzidi
- Laboratoire d’Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Faculté des Sciences, Université Mohammed Premier, Oujda, 60000, Morocco
- Euro-Mediterranean University of Fes (UEMF), Fes, Morocco
| | - Mounir Haddou
- Laboratoire d’Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Faculté des Sciences, Université Mohammed Premier, Oujda, 60000, Morocco
- Centre de L’Oriental des Sciences et Technologies de L’Eau et de L’Environnement (COSTEE), Université Mohammed Premier, Oujda, 60000, Morocco
| | - Abdellah Baraich
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Boulevard Mohamed VI, B.P. 717, Oujda, 60000, Morocco
| | | | - Tarik Moubchir
- Polyvalent Team in Research and Development, Polydisciplinary Faculty of Beni Mellal (FPBM), University Sultan Moulay Slimane (USMS), Beni Mellal, 23000, Morocco
| | - Aimad Allali
- High Institute of Nursing Professions and Health Techniques Annex Taza, Fez, Morocco
| | - Amine khoulati
- Faculté de Médecine et de Pharmacie, Université Mohammed Premier, Oujda, 60000, Morocco
| | - Reda Bellaouchi
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Boulevard Mohamed VI, B.P. 717, Oujda, 60000, Morocco
| | - Abdeslam Asehraou
- Laboratory of Bioresources, Biotechnology, Ethnopharmacology and Health, Faculty of Sciences, Mohammed First University, Boulevard Mohamed VI, B.P. 717, Oujda, 60000, Morocco
| | - Mohamed Addi
- Laboratoire d’Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Faculté des Sciences, Université Mohammed Premier, Oujda, 60000, Morocco
| | - Ahmad Mohammad Salamatullah
- Department of Food Science & Nutrition, College of Food and Agricultural Sciences, King Saud University, 11 P.O. Box 2460, Riyadh, 11451, Saudi Arabia
| | - Mohammed Bourhia
- Laboratory of Biotechnology and Natural Resources Valorization, Faculty of Sciences, Ibn Zohr University, 80060, Agadir, Morocco
| | - Farhan Siddique
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, SE-60174, Norrköping, Sweden
| | - Bouchra El Guerrouj
- Laboratoire d’Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Faculté des Sciences, Université Mohammed Premier, Oujda, 60000, Morocco
- Centre de L’Oriental des Sciences et Technologies de L’Eau et de L’Environnement (COSTEE), Université Mohammed Premier, Oujda, 60000, Morocco
| | - Khalid Chaabane
- Laboratoire d’Amélioration des Productions Agricoles, Biotechnologie et Environnement (LAPABE), Faculté des Sciences, Université Mohammed Premier, Oujda, 60000, Morocco
| |
Collapse
|
14
|
Ferreira J, Roque S, Lima Carneiro A, Longatto‐Filho A, Vila I, Cunha C, Silva C, Mesquita A, Cotter J, Correia‐Neves M, Mansilha A, Cunha PG. Reversion of the Inflammatory Markers in Patients With Chronic Limb-Threatening Ischemia. J Am Heart Assoc 2024; 13:e031922. [PMID: 38606780 PMCID: PMC11262488 DOI: 10.1161/jaha.123.031922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 02/23/2024] [Indexed: 04/13/2024]
Abstract
BACKGROUND Peripheral artery disease is characterized by an intense inflammatory process that can be associated with a higher mortality rate, particularly in chronic limb-threatening ischemia (CLTI). This study aims to compare the evolution of inflammatory markers between patients with claudication with those with CLTI at 3, 6, and 12 months. METHODS AND RESULTS An observational, single-center, and prospective study was conducted. A total of 119 patients with peripheral artery disease (65 with claudication and 54 with CLTI) were observed and inflammatory markers collected at admission and 3, 6, and 12 months. At admission, patients with CLTI, when compared with patients with claudication, had significantly higher serum levels of C-reactive protein and fibrinogen (positive acute-phase proteins) and lower serum level of albumin, total cholesterol, and high-density lipoprotein (negative acute-phase proteins): C-reactive protein (g/dL), 2.90 (25th-75th percentile, 2.90-4.90) versus 6.80 (25th-75th percentile, 2.90-53.26) (P=0.000); fibrinogen (mg/dL), 293.00 (25th-75th percentile, 269.25-349.00) versus 415.50 (25th-75th percentile, 312.00-615.75) (P=0.000); total cholesterol (mg/dL), 161.79±95% [152.74-170.85] versus 146.42%±95% [135.30-157.53] (P=0.034); high-density lipoprotein (mg/dL), 50.00 (25th-75th percentile, 41.00-60.00) versus 37.00 (25th-75th percentile, 30.00-45.50) (P=0.000); albumin (g/dL): 4.00 (25th-75th percentile, 3.70-4.20) versus 3.60 (25th-75th percentile, 3.10-4.00) (P=0.003). The association between CLTI and total cholesterol was lost after adjusting for confounders. Three months after the resolution of the CLTI, there was an increase in the levels of negative acute-phase proteins and a decrease in positive acute-phase proteins. These inflammatory proteins did not register an evolution in patients with claudication. The differences in the inflammatory proteins between groups disappeared at 6 months. CONCLUSIONS CLTI has an inflammatory environment that can be partially reverted after resolution of the ischemic process, emphasizing the importance of timely intervention.
Collapse
Affiliation(s)
- Joana Ferreira
- Vascular Surgery Department—Physiology and SurgeryCentro Hospitalar Universitário de São JoãoPortoPortugal
- Life and Health Science Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B’s—PT Government Associated LaboratoryBragaPortugal
- Academic Center Hospital da Senhora da OliveiraGuimarãesPortugal
- Clinical Academic Center Hospital de Trás‐os‐Montes e Alto Douro—Professor Doutor Nuno Grande—CACTMADVila RealPortugal
| | - Susana Roque
- Life and Health Science Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B’s—PT Government Associated LaboratoryBragaPortugal
| | | | - Adhemar Longatto‐Filho
- Life and Health Science Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B’s—PT Government Associated LaboratoryBragaPortugal
- Department of Pathology (LIM‐14)University of São Paulo School of MedicineSão PauloBrazil
- Molecular Oncology Research CenterBarretos Cancer HospitalBarretosSão PauloBrazil
| | - Isabel Vila
- Academic Center Hospital da Senhora da OliveiraGuimarãesPortugal
- Center for the Research and Treatment of Arterial Hypertension and Cardiovascular Risk,Hospital da Senhora da OliveiraGuimarãesPortugal
- Internal Medicine DepartmentHospital da Senhora da Oliveira, GuimarãesBragaPortugal
| | - Cristina Cunha
- Academic Center Hospital da Senhora da OliveiraGuimarãesPortugal
- Center for the Research and Treatment of Arterial Hypertension and Cardiovascular Risk,Hospital da Senhora da OliveiraGuimarãesPortugal
- Internal Medicine DepartmentHospital da Senhora da Oliveira, GuimarãesBragaPortugal
| | - Cristina Silva
- Academic Center Hospital da Senhora da OliveiraGuimarãesPortugal
- Center for the Research and Treatment of Arterial Hypertension and Cardiovascular Risk,Hospital da Senhora da OliveiraGuimarãesPortugal
- Internal Medicine DepartmentHospital da Senhora da Oliveira, GuimarãesBragaPortugal
| | - Amílcar Mesquita
- Vascular Surgery Department—Hospital da Senhora da OliveiraGuimarãesPortugal
| | - Jorge Cotter
- Life and Health Science Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B’s—PT Government Associated LaboratoryBragaPortugal
- Academic Center Hospital da Senhora da OliveiraGuimarãesPortugal
- Center for the Research and Treatment of Arterial Hypertension and Cardiovascular Risk,Hospital da Senhora da OliveiraGuimarãesPortugal
- Internal Medicine DepartmentHospital da Senhora da Oliveira, GuimarãesBragaPortugal
| | - Margarida Correia‐Neves
- Life and Health Science Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B’s—PT Government Associated LaboratoryBragaPortugal
| | - Armando Mansilha
- Vascular Surgery Department—Physiology and SurgeryCentro Hospitalar Universitário de São JoãoPortoPortugal
- Department of Angiology and Vascular SurgeryCentro Hospitalar Universitário de São JoãoPortoPortugal
- Faculty of MedicineUniversity of PortoPortoPortugal
| | - Pedro Guimarães Cunha
- Life and Health Science Research Institute (ICVS), School of MedicineUniversity of MinhoBragaPortugal
- ICVS/3B’s—PT Government Associated LaboratoryBragaPortugal
- Academic Center Hospital da Senhora da OliveiraGuimarãesPortugal
- Center for the Research and Treatment of Arterial Hypertension and Cardiovascular Risk,Hospital da Senhora da OliveiraGuimarãesPortugal
- Internal Medicine DepartmentHospital da Senhora da Oliveira, GuimarãesBragaPortugal
| |
Collapse
|
15
|
Malik MI, Nagpal D. Estimated pulse-wave velocity predicts survival in patients requiring extracorporeal membrane oxygenation. Perfusion 2024; 39:344-352. [PMID: 36419384 DOI: 10.1177/02676591221141963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2023]
Abstract
INTRODUCTION Arterial stiffness, measured by estimated pulse-wave velocity is a known predictor of major adverse cardiovascular events, however its predictive value in patients requiring extracorporeal membrane oxygenation (ECMO) is unknown. METHODS A retrospective cohort study was performed at the London Health Science Centre in London, Canada between 1996-2021, totaling 255 patients requiring ECMO. Estimated pulse-wave velocity (ePWV) was calculated using an algorithm from the Reference Values for Arterial Stiffness Collaboration. Recorded outcomes included in-hospital death, ischemic stroke, hemorrhagic stroke, renal failure and need for renal replacement therapy (RRT). For adjusted analysis, survival-to-discharge was used. Multivariate logistic regression and propensity-score matching were utilized to control for confounding. RESULTS On univariate analysis, higher ePWV was significantly predictive of ischemic stroke (OR 1.676, p = 0.0002) and in-hospital death (OR 1.20, p = 0.006), but insignificant for predicting hemorrhagic stroke (OR 1.07, p = 0.710), and appeared protective for renal failure (OR 0.88 [0.78-0.99], p = 0.034) and RRT (OR 0.87, p = 0.027). On multivariate analysis and propensity-score matching, five of six models demonstrated ePWV as an independent predictor of survival-to-discharge. (OR 0.70, p = 0.00,021; OR 0.72, p = 0.0002; OR 0.87, p = 0.045; OR 0.85, p = 0.013; OR 0.57, p = 0.012). CONCLUSIONS ePWV is a promising marker for risk-stratification in ECMO patients. Further investigation is required to better delineate the role of arterial health assessment in disease trajectory and strengthen the validity of AS as a marker of interest in medical and surgical management.
Collapse
Affiliation(s)
- Mohsyn I Malik
- Department of Cardiac Surgery, Schulich School of Medicine and Dentistry, London, ON, Canada
| | - Dave Nagpal
- Department of Cardiac Surgery, Schulich School of Medicine and Dentistry, London, ON, Canada
| |
Collapse
|
16
|
Alemany M. The Metabolic Syndrome, a Human Disease. Int J Mol Sci 2024; 25:2251. [PMID: 38396928 PMCID: PMC10888680 DOI: 10.3390/ijms25042251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
This review focuses on the question of metabolic syndrome (MS) being a complex, but essentially monophyletic, galaxy of associated diseases/disorders, or just a syndrome of related but rather independent pathologies. The human nature of MS (its exceptionality in Nature and its close interdependence with human action and evolution) is presented and discussed. The text also describes the close interdependence of its components, with special emphasis on the description of their interrelations (including their syndromic development and recruitment), as well as their consequences upon energy handling and partition. The main theories on MS's origin and development are presented in relation to hepatic steatosis, type 2 diabetes, and obesity, but encompass most of the MS components described so far. The differential effects of sex and its biological consequences are considered under the light of human social needs and evolution, which are also directly related to MS epidemiology, severity, and relations with senescence. The triggering and maintenance factors of MS are discussed, with especial emphasis on inflammation, a complex process affecting different levels of organization and which is a critical element for MS development. Inflammation is also related to the operation of connective tissue (including the adipose organ) and the widely studied and acknowledged influence of diet. The role of diet composition, including the transcendence of the anaplerotic maintenance of the Krebs cycle from dietary amino acid supply (and its timing), is developed in the context of testosterone and β-estradiol control of the insulin-glycaemia hepatic core system of carbohydrate-triacylglycerol energy handling. The high probability of MS acting as a unique complex biological control system (essentially monophyletic) is presented, together with additional perspectives/considerations on the treatment of this 'very' human disease.
Collapse
Affiliation(s)
- Marià Alemany
- Faculty of Biology, Universitat de Barcelona, 08028 Barcelona, Catalonia, Spain
| |
Collapse
|
17
|
Kawamoto R, Kikuchi A, Niomiya D, Kumagi T. High-sensitivity C-reactive protein is a predictor of all-cause mortality in a rural Japanese population. J Clin Lab Anal 2024; 38:e25015. [PMID: 38419270 PMCID: PMC10943256 DOI: 10.1002/jcla.25015] [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: 10/24/2023] [Revised: 01/01/2024] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND High-sensitivity C-reactive protein (hsCRP) is a sensitive marker of inflammation. This study aimed to determine whether increased hsCRP levels are associated with all-cause mortality rate. METHODS We examined data for participants from the 2002 Nomura Cohort Study who attended follow-ups for 20 years (follow-up rate: 93.3%). Of these, 793 were male (aged 61 ± 14 years) and 1040 were female (aged 63 ± 11 years). The Japanese Basic Resident Registry provided data on adjusted relative hazards for all-cause mortality. The data were subjected to a Cox regression analysis using a time variable of age and confounding risk factors. RESULTS The median (interquartile range) follow-up period was 6548 days (6094-7452 days). The follow-up confirmed that there were 632 (34.8%) deaths, of which 319 were male (40.2% of all males) and 313 were female (30.6% of all females). Multivariable-adjusted hazard ratio (1.27; 95% confidence interval, 1.01-1.59) in the highest hsCRP category was also significantly higher compared with reference. A higher hsCRP was associated with a greater risk of all-cause mortality in male participants aged ≥65 years, a BMI < 25 kg/m2 , and no history of CVD or diabetes, and this association was particularly significant among participants with both of the latter two risk factors (p = 0.004 and 0.022 for interaction, respectively). CONCLUSIONS Our results indicate a significant association between hsCRP levels and all-cause mortality in a rural Japanese population. Specifically, hsCRP appears to be a crucial biomarker for predicting long-term survival, particularly among older persons.
Collapse
Affiliation(s)
- Ryuichi Kawamoto
- Department of Community MedicineEhime University Graduate School of MedicineToon‐CityEhimeJapan
- Department of Internal MedicineSeiyo Municipal Nomura HospitalSeiyo‐CityEhimeJapan
| | - Asuka Kikuchi
- Department of Community MedicineEhime University Graduate School of MedicineToon‐CityEhimeJapan
- Department of Internal MedicineSeiyo Municipal Nomura HospitalSeiyo‐CityEhimeJapan
| | - Daisuke Niomiya
- Department of Community MedicineEhime University Graduate School of MedicineToon‐CityEhimeJapan
- Department of Internal MedicineSeiyo Municipal Nomura HospitalSeiyo‐CityEhimeJapan
| | - Teru Kumagi
- Department of Community MedicineEhime University Graduate School of MedicineToon‐CityEhimeJapan
| |
Collapse
|
18
|
Lataro RM, Brognara F, Iturriaga R, Paton JFR. Inflammation of some visceral sensory systems and autonomic dysfunction in cardiovascular disease. Auton Neurosci 2024; 251:103137. [PMID: 38104365 DOI: 10.1016/j.autneu.2023.103137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 12/19/2023]
Abstract
The sensitization and hypertonicity of visceral afferents are highly relevant to the development and progression of cardiovascular and respiratory disease states. In this review, we described the evidence that the inflammatory process regulates visceral afferent sensitivity and tonicity, affecting the control of the cardiovascular and respiratory system. Some inflammatory mediators like nitric oxide, angiotensin II, endothelin-1, and arginine vasopressin may inhibit baroreceptor afferents and contribute to the baroreflex impairment observed in cardiovascular diseases. Cytokines may act directly on peripheral afferent terminals that transmit information to the central nervous system (CNS). TLR-4 receptors, which recognize lipopolysaccharide, were identified in the nodose and petrosal ganglion and have been implicated in disrupting the blood-brain barrier, which can potentiate the inflammatory process. For example, cytokines may cross the blood-brain barrier to access the CNS. Additionally, pro-inflammatory cytokines such as IL-1β, IL-6, TNF-α and some of their receptors have been identified in the nodose ganglion and carotid body. These pro-inflammatory cytokines also sensitize the dorsal root ganglion or are released in the nucleus of the solitary tract. In cardiovascular disease, pro-inflammatory mediators increase in the brain, heart, vessels, and plasma and may act locally or systemically to activate/sensitize afferent nervous terminals. Recent evidence demonstrated that the carotid body chemoreceptor cells might sense systemic pro-inflammatory molecules, supporting the novel proposal that the carotid body is part of the afferent pathway in the central anti-inflammatory reflexes. The exact mechanisms of how pro-inflammatory mediators affects visceral afferent signals and contribute to the pathophysiology of cardiovascular diseases awaits future research.
Collapse
Affiliation(s)
- R M Lataro
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| | - F Brognara
- Department of Nursing, General and Specialized, Nursing School of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - R Iturriaga
- Facultad de Ciencias Biológicas, Pontificia Universidad Catolica de Chile, Santiago, Chile; Centro de Investigación en Fisiología y Medicina en Altura - FIMEDALT, Universidad de Antofagasta, Antofagasta, Chile
| | - J F R Paton
- Manaaki Manawa - The Centre for Heart Research, Department of Physiology, Faculty of Medical & Health Sciences, University of Auckland, Grafton, Auckland, New Zealand
| |
Collapse
|
19
|
Zeng X, Yang Y. Molecular Mechanisms Underlying Vascular Remodeling in Hypertension. Rev Cardiovasc Med 2024; 25:72. [PMID: 39077331 PMCID: PMC11263180 DOI: 10.31083/j.rcm2502072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 07/31/2024] Open
Abstract
Hypertension, a common cardiovascular disease, is primarily characterized by vascular remodeling. Recent extensive research has led to significant progress in understanding its mechanisms. Traditionally, vascular remodeling has been described as a unidirectional process in which blood vessels undergo adaptive remodeling or maladaptive remodeling. Adaptive remodeling involves an increase in vessel diameter in response to increased blood flow, while maladaptive remodeling refers to the narrowing or thickening of blood vessels in response to pathological conditions. However, recent research has revealed that vascular remodeling is much more complex. It is now understood that vascular remodeling is a dynamic interplay between various cellular and molecular events. This interplay process involves different cell types, including endothelial cells, smooth muscle cells, and immune cells, as well as their interactions with the extracellular matrix. Through these interactions, blood vessels undergo intricate and dynamic changes in structure and function in response to various stimuli. Moreover, vascular remodeling involves various factors and mechanisms such as the renin-angiotensin-aldosterone system (RAS), oxidative stress, inflammation, the extracellular matrix (ECM), sympathetic nervous system (SNS) and mechanical stress that impact the arterial wall. These factors may lead to vascular and circulatory system diseases and are primary causes of long-term increases in systemic vascular resistance in hypertensive patients. Additionally, the presence of stem cells in adventitia, media, and intima of blood vessels plays a crucial role in vascular remodeling and disease development. In the future, research will focus on examining the underlying mechanisms contributing to hypertensive vascular remodeling to develop potential solutions for hypertension treatment. This review provides us with a fresh perspective on hypertension and vascular remodeling, undoubtedly sparking further research efforts aimed at uncovering more potent treatments and enhanced preventive and control measures for this disease.
Collapse
Affiliation(s)
- Xinyi Zeng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
20
|
Zanoli L, Gaudio A, Lo Cicero L. Reversibility of Aortic Stiffening During the First Waves of COVID-19. Angiology 2024; 75:105-106. [PMID: 37358839 PMCID: PMC10293861 DOI: 10.1177/00033197231186092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2023]
Affiliation(s)
- Luca Zanoli
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Agostino Gaudio
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Lorenzo Lo Cicero
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
21
|
D’Elia L, Strazzullo P. Dietary Salt Restriction and Adherence to the Mediterranean Diet: A Single Way to Reduce Cardiovascular Risk? J Clin Med 2024; 13:486. [PMID: 38256620 PMCID: PMC10816989 DOI: 10.3390/jcm13020486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 01/08/2024] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
The dietary restriction of salt intake and the adhesion to Mediterranean dietary patterns are among the most recommended lifestyle modifications for the prevention of cardiovascular diseases. A large amount of evidence supports these recommendations; indeed, several studies show that a higher adherence to Mediterranean dietary patterns is associated with a reduced risk of cardiovascular disease. Likewise, findings from observational and clinical studies suggest a causal role of excess salt intake in blood pressure increase, cardiovascular organ damage, and the incidence of cardiovascular diseases. In this context, it is also conceivable that the beneficial effects of these two dietary patterns overlap because Mediterranean dietary patterns are typically characterized by a large consumption of plant-based foods with low sodium content. However, there is little data on this issue, and heterogeneous results are available on the relationship between adherence to salt restriction and to Mediterranean dietary patterns. Thus, this short review focuses on the epidemiological and clinical evidence of the relationship between the adherence to Mediterranean dietary patterns and dietary salt restriction in the context of cardiovascular risk.
Collapse
Affiliation(s)
| | - Pasquale Strazzullo
- Department of Clinical Medicine and Surgery, “Federico II” University of Naples Medical School, 80131 Naples, Italy;
| |
Collapse
|
22
|
Turgunova L, Mekhantseva I, Laryushina Y, Alina A, Bacheva I, Zhumadilova Z, Turmukhambetova A. The Association of Endothelin-1 with Early and Long-Term Mortality in COVID-19. J Pers Med 2023; 13:1558. [PMID: 38003873 PMCID: PMC10672100 DOI: 10.3390/jpm13111558] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 10/22/2023] [Accepted: 10/26/2023] [Indexed: 11/26/2023] Open
Abstract
(1) Background: Endothelial dysfunction is a key mechanism in the pathogenesis of COVID-19. High endothelin-1 during COVID-19 is associated with severe complications and increased mortality rates during hospitalization. This study is aimed to investigate the association of endothelin-1 levels with the risk of 30-day and 12-month all-cause mortality in patients with prior COVID-19. (2) Methods: A prospective study was conducted involving patients with COVID-19 in Karaganda, Kazakhstan. The level of endothelin-1 in the blood serum was evaluated by ELISA. Univariate and multivariate Cox regression was used to determine factors and significance of endothelin-1 associated with the risk of mortality within 30 and 365 days from hospitalization. (3) Results: The median endothelin-1 was higher in the group of patients who passed away within 30 days. The group showed statistically significant differences when compared to healthy volunteers from the control group (p = 0.0001), surviving patients (p = 0.001), and those who passed away within a year (p = 0.002). (4) Conclusions: Endothelin-1 levels are associated with increased mortality risk during the acute period of COVID-19, while plasma endothelin-1 level association with COVID-19 survivor mortality risk does not persist after 12 months.
Collapse
Affiliation(s)
| | - Irina Mekhantseva
- Department of Internal Medicine, Karaganda Medical University, Karaganda 100000, Kazakhstan (I.B.); (Z.Z.)
| | | | | | | | | | | |
Collapse
|
23
|
Mclaughlin M, Sanal-Hayes NEM, Hayes LD, Berry EC, Sculthorpe NF. People with Long COVID and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome Exhibit Similarly Impaired Vascular Function. Am J Med 2023:S0002-9343(23)00609-5. [PMID: 37832757 DOI: 10.1016/j.amjmed.2023.09.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/15/2023]
Abstract
BACKGROUND This study aimed to compare flow-mediated dilation values between individuals with long COVID, individuals with myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS), and healthy age-matched controls to assess the potential implications for clinical management and long-term health outcomes. METHODS A case-case-control approach was employed, and flow-mediated dilation measurements were obtained from 51 participants (17 long COVID patients, 17 ME/CFS patients, and 17 healthy age-matched controls). Flow-mediated dilation values were analyzed using 1-way analysis of variance for between-group comparisons. RESULTS Results revealed significantly impaired endothelial function in both long COVID and ME/CFS groups compared with healthy age-matched controls as determined by maximum % brachial artery diameter post-occlusion compared with pre-occlusion resting diameter (6.99 ± 4.33% and 6.60 ± 3.48% vs 11.30 ± 4.44%, respectively, both P < .05). Notably, there was no difference in flow-mediated dilation between long COVID and ME/CFS groups (P = .949), despite significantly longer illness duration in the ME/CFS group (ME/CFS: 16 ± 11.15 years vs long COVID: 1.36 ± 0.51 years, P < .0001). CONCLUSION The study demonstrates that both long COVID and ME/CFS patients exhibit similarly impaired endothelial function, indicating potential vascular involvement in the pathogenesis of these post-viral illnesses. The significant reduction in flow-mediated dilation values suggests an increased cardiovascular risk in these populations, warranting careful monitoring and the development of targeted interventions to improve endothelial function and mitigate long-term health implications.
Collapse
Affiliation(s)
- Marie Mclaughlin
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom; School of Sport, Exercise & Rehabilitation Sciences, Faculty of Health Sciences, University of Hull, United Kingdom.
| | - Nilihan E M Sanal-Hayes
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom; School of Health and Society, University of Salford, United Kingdom
| | - Lawrence D Hayes
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom
| | - Ethan C Berry
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom
| | - Nicholas F Sculthorpe
- Sport and Physical Activity Research Institute, School of Health and Life Sciences, University of the West of Scotland, Glasgow, United Kingdom
| |
Collapse
|
24
|
Jin L, Wu L, Chen J, Zhang M, Sun J, Shen C, Du L, She X, Li Z. Uncoupling of the center-to-periphery arterial stiffness gradient and pulse pressure amplification in viral pneumonia infection. BMC Infect Dis 2023; 23:657. [PMID: 37798630 PMCID: PMC10552441 DOI: 10.1186/s12879-023-08650-w] [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: 05/15/2023] [Accepted: 09/26/2023] [Indexed: 10/07/2023] Open
Abstract
OBJECTIVES Arterial stiffness is a common manifestation of viral pneumonia infections, including COVID-19. Nevertheless, the relationship between the center-to-periphery arterial stiffness gradient and pulse pressure amplification (PPA) in infectious diseases remains unclear. This study aimed to investigate this relationship utilizing arterial pressure volume index (API) and arterial velocity pulse index (AVI) ratio. METHODS API/AVI and PPA were measured in 219 participants with COVID-19 and 374 normal participants. Multiple linear regression was used to assess the association of API/AVI and PPA, and restricted cubic spline was used to investigate the non-linear relationship between API/AVI and PPA. Receiver operating characteristic curve (ROC) analysis was used to evaluate the effects of API/AVI in identifying COVID-19 infection and severe stage. RESULTS There was a significant J-shaped relationship between API/AVI and PPA in COVID-19 group, while a M-shaped relationship was observed in normal group. API/AVI decreased rapidly as PPA decreased until API/AVI decreased slowly at PPA of 1.07, and then API/AVI decreased slowly again at PPA of 0.78. ROC results showed that API/AVI demonstrated excellent accuracy in identifying COVID-19 infection (AUC = 0.781) and a high specificity (84.88%) in identifying severe stage. CONCLUSIONS There was a J-shaped association between the API/AVI and PPA in viral infected patients, while a M-shaped relationship in the normal participants. API/AVI is better for identifying infected and uninfected patients, with a high specificity in identifying those in severe stages of the disease. The attenuation or reversal of API/AVI may be associated with the loss of PPA coupling.
Collapse
Affiliation(s)
- Lin Jin
- Department of Ultrasound, Guanghua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, 200052, China
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Lingheng Wu
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Jianxiong Chen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
- Department of Ultrasound, Shanghai General Hospital of Nanjing Medical University, Shanghai, 200080, China
| | - Mengjiao Zhang
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Jiali Sun
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Cuiqin Shen
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Lianfang Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China
| | - Xiaoyin She
- Department of Emergency and Critical Care, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China
| | - Zhaojun Li
- Department of Ultrasound, Jiading Branch of Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 201812, China.
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
25
|
Mengozzi A, de Ciuceis C, Dell'oro R, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Anyfanti P, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Mavraganis G, Montezano AC, Rios FJ, Winklewski PJ, Wolf J, Costantino S, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Triantafyllou A, Virdis A. The importance of microvascular inflammation in ageing and age-related diseases: a position paper from the ESH working group on small arteries, section of microvascular inflammation. J Hypertens 2023; 41:1521-1543. [PMID: 37382158 DOI: 10.1097/hjh.0000000000003503] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/30/2023]
Abstract
Microcirculation is pervasive and orchestrates a profound regulatory cross-talk with the surrounding tissue and organs. Similarly, it is one of the earliest biological systems targeted by environmental stressors and consequently involved in the development and progression of ageing and age-related disease. Microvascular dysfunction, if not targeted, leads to a steady derangement of the phenotype, which cumulates comorbidities and eventually results in a nonrescuable, very high-cardiovascular risk. Along the broad spectrum of pathologies, both shared and distinct molecular pathways and pathophysiological alteration are involved in the disruption of microvascular homeostasis, all pointing to microvascular inflammation as the putative primary culprit. This position paper explores the presence and the detrimental contribution of microvascular inflammation across the whole spectrum of chronic age-related diseases, which characterise the 21st-century healthcare landscape. The manuscript aims to strongly affirm the centrality of microvascular inflammation by recapitulating the current evidence and providing a clear synoptic view of the whole cardiometabolic derangement. Indeed, there is an urgent need for further mechanistic exploration to identify clear, very early or disease-specific molecular targets to provide an effective therapeutic strategy against the otherwise unstoppable rising prevalence of age-related diseases.
Collapse
Affiliation(s)
- Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
| | - Raffaella Dell'oro
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - George Pavlidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
| | | | - Panagiota Anyfanti
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Livia L Camargo
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine
- Center of Translational Medicine
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Marcin Hellmann
- Department of Cardiac Diagnostics, Medical University, Gdansk, Poland
| | - Stefano Masi
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Institute of Cardiovascular Science, University College London, London, UK
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Francesco J Rios
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | | | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine
| | - Sarah Costantino
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute; University of Edinburgh, University of Edinburgh, Edinburgh, UK
- Department of Internal Medicine
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Ignatios Ikonomidis
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2 Cardiology Department, Attikon Hospital, Athens
- Medical School, National and Kapodistrian University of Athens, Greece
| | | | - Francesco Paneni
- Center for Translational and Experimental Cardiology (CTEC), Department of Cardiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
- University Heart Center, Cardiology, University Hospital Zurich
- Department of Research and Education, University Hospital Zurich, Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia
- Division of Medicine, Spedali Civili di Brescia, Montichiari, Brescia, Italy
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School, National and Kapodistrian University of Athens, Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK
- Department of Cardiovascular Research, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University
- German Centre for Cardiovascular Research (Deutsches Zentrum für Herz-Kreislauf-Forschung, DZHK), Heidelberg/Mannheim Partner Site
- Department of Cardiology, University Hospital Mannheim, Heidelberg University, Manheim, Germany
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
- Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, Canada
| | - Areti Triantafyllou
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Thessaloniki, Greece
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| |
Collapse
|
26
|
Gómez-Sánchez L, Tamayo-Morales O, Suárez-Moreno N, Bermejo-Martín JF, Domínguez-Martín A, Martín-Oterino JA, Martín-González JI, González-Calle D, García-García Á, Lugones-Sánchez C, González-Sánchez S, Jiménez-Gómez R, García-Ortiz L, Gómez-Marcos MA, Navarro-Matías E. Relationship between the structure, function and endothelial damage, and vascular ageing and the biopsychological situation in adults diagnosed with persistent COVID (BioICOPER study). A research protocol of a cross-sectional study. Front Physiol 2023; 14:1236430. [PMID: 37772064 PMCID: PMC10523018 DOI: 10.3389/fphys.2023.1236430] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/17/2023] [Indexed: 09/30/2023] Open
Abstract
Background: SARS-CoV-2 infection affects the vascular endothelium, which mediates the inflammatory and thrombotic cascade. Moreover, alterations in the endothelium are related to arterial stiffness, which has been established as a marker of cardiovascular disease. The objective of this study is to analyse how the structure, vascular function, vascular ageing and endothelial damage are related to the biopsychological situation in adults diagnosed with persistent COVID and the differences by gender. Methods: This cross-sectional, descriptive, observational study will be carried out in the Primary Care Research Unit of Salamanca (APISAL) and in the BioSepsis laboratory of the University of Salamanca. The sample will be selected from the persistent COVID monographic office at the Internal Medicine Service of the University Hospital of Salamanca, and from the population of subjects diagnosed with persistent COVID in the clinical history of Primary Care. Through consecutive sampling, the study will include 300 individuals diagnosed with persistent COVID who meet the diagnosis criteria established by the WHO, after they sign the informed consent. Endothelial damage biomarkers will be measured using ELLA-SimplePlexTM technology (Biotechne). Their vascular structure and function will be analysed by measuring the carotid intima-media thickness (Sonosite Micromax); the pulse wave and carotid-femoral pulse wave velocity (cfPWV) will be recorded with Sphygmocor System®. Cardio Ankle Vascular Index (CAVI), brachial-ankle pulse wave velocity (baPWV) and ankle-brachial index will be analysed with Vasera VS-2000®. The integral assessment of the subjects with persistent COVID will be conducted with different scales that evaluate fatigue, sleep, dyspnea, quality of life, attention, nutrition state, and fragility. We will also evaluate their lifestyles (diet, physical activity, smoking habits and alcohol consumption), psychological factors, and cognitive deterioration, which will be gathered through validated questionnaires; moreover, physical activity will be objectively measured using a pedometer for 7 days. Body composition will be measured through impedance using an Inbody 230. Vascular ageing will be calculated with 10 and 90 percentiles of cfPWV and baPWV. Furthermore, we will analyse the presence of vascular injury in the retina, heart, kidneys and brain, as well as cardiovascular risk. Demographic and analytical variables will also be gathered. Discussion: Arterial stiffness reflects the mechanic and functional properties of the arterial wall, showing the changes in arterial pressure, blood flow, and vascular diameter that occur with each heartbeat. SARS-CoV-2 affects the endothelial cells that are infected with this virus, increasing the production of pro-inflammatory cytokines and pro-thrombotic factors, which can cause early vascular ageing and an increase of arterial stiffness. Persistent COVID is a complex heterogeneous disorder that affects the lives of millions of people worldwide. The identifications of potential risk factors to better understand who is at risk of developing persistent COVID is important, since this would enable early and appropriate clinical support. It is unknown whether vascular alterations caused by COVID-19 resolve after acute infection or remain over time, favouring the increase of arterial stiffness and early vascular ageing. Therefore, it is necessary to propose studies that analyse the evolution of persistent COVID in this group of patients, as well as the possible variables that influence it. Clinical Trial registration: ClinicalTrials.gov, identifier NCT05819840.
Collapse
Affiliation(s)
- Leticia Gómez-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Hospital de la Paz de Madrid, Servicio de Urgencias, Madrid, Spain
| | - Olaya Tamayo-Morales
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
| | - Nuria Suárez-Moreno
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | - Jesus F. Bermejo-Martín
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Madrid, Spain
| | - Andrea Domínguez-Martín
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | - José A. Martín-Oterino
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Hospital Universitario de Salamanca, Internal Medicine Department, Salamanca, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - José I. Martín-González
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Hospital Universitario de Salamanca, Internal Medicine Department, Salamanca, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - David González-Calle
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
- Hospital Universitario de Salamanca, Cardiology Department, Salamanca, Spain
| | - Ángel García-García
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
- Hospital Universitario de Salamanca, Emergency Department, Salamanca, Spain
| | - Cristina Lugones-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
| | - Susana González-Sánchez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
| | - Raquel Jiménez-Gómez
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | - Luis García-Ortiz
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Biomedical and Diagnostic Sciences, University of Salamanca, Salamanca, Spain
| | - Manuel A. Gómez-Marcos
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Elena Navarro-Matías
- Primary Care Research Unit of Salamanca (APISAL), Salamanca Primary Care Management, Salamanca, Spain
- Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain
- Castilla and León Health Service–SACYL, Gerencia Regional de Salud, Valladolid, Spain
| | | |
Collapse
|
27
|
Madison AA, Andridge R, Kantaras AH, Renna ME, Bennett JM, Alfano CM, Povoski SP, Agnese DM, Lustberg M, Wesolowski R, Carson WE, Williams NO, Reinbolt RE, Sardesai SD, Noonan AM, Stover DG, Cherian MA, Malarkey WB, Kiecolt-Glaser JK. Depression, Inflammation, and Intestinal Permeability: Associations with Subjective and Objective Cognitive Functioning throughout Breast Cancer Survivorship. Cancers (Basel) 2023; 15:4414. [PMID: 37686689 PMCID: PMC10487080 DOI: 10.3390/cancers15174414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/10/2023] Open
Abstract
About one-in-three breast cancer survivors have lingering cognitive complaints and objective cognitive impairment. Chronic inflammation and intestinal permeability (i.e., leaky gut), two risk factors for cognitive decline, can also fuel depression-another vulnerability for cognitive decline. The current study tested whether depression accompanied by high levels of inflammation or intestinal permeability predicted lower subjective and objective cognitive function in breast cancer survivors. We combined data from four breast cancer survivor studies (n = 613); some had repeated measurements for a total of 1015 study visits. All participants had a blood draw to obtain baseline measures of lipopolysaccharide binding protein-a measure of intestinal permeability, as well as three inflammatory markers that were incorporated into an inflammatory index: C-reactive protein, interleukin-6, and tumor necrosis factor-α. They reported depressive symptoms on the Center for Epidemiological Studies depression scale (CES-D), and a binary variable indicated clinically significant depressive symptoms (CES-D ≥ 16). The Kohli (749 observations) and the Breast Cancer Prevention Trial (591 observations) scales assessed subjective cognitive function. Objective cognitive function tests included the trail-making test, Hopkins verbal learning test, Conners continuous performance test, n-back test, FAS test, and animal-naming test (239-246 observations). Adjusting for education, age, BMI, cancer treatment type, time since treatment, study visit, and fatigue, women who had clinically elevated depressive symptoms accompanied by heightened inflammation or intestinal permeability reported poorer focus and marginally poorer memory. However, poorer performance across objective cognitive measures was not specific to inflammation-associated depression. Rather, there was some evidence of lower verbal fluency; poorer attention, verbal learning and memory, and working memory; and difficulties with visuospatial search among depressed survivors, regardless of inflammation. By themselves, inflammation and intestinal permeability less consistently predicted subjective or objective cognitive function. Breast cancer survivors with clinically significant depressive symptoms accompanied by either elevated inflammation or intestinal permeability may perceive greater cognitive difficulty, even though depression-related objective cognitive deficits may not be specific to inflammation- or leaky-gut-associated depression.
Collapse
Affiliation(s)
- Annelise A Madison
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Psychology, The Ohio State University, Columbus, OH 43210, USA
| | - Rebecca Andridge
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Biostatistics, The Ohio State University, Columbus, OH 43210, USA
| | - Anthony H Kantaras
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210, USA
| | - Megan E Renna
- School of Psychology, University of Southern Mississippi, Hattiesburg, MS 39406, USA
| | - Jeanette M Bennett
- Department of Psychological Science, University of North Carolina at Charlotte, Charlotte, NC 28213, USA
| | | | - Stephen P Povoski
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Doreen M Agnese
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Maryam Lustberg
- Center for Breast Cancer, Yale Cancer Center, Yale University, New Haven, CT 06519, USA
| | - Robert Wesolowski
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - William E Carson
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Division of Surgical Oncology, Department of Surgery, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Nicole O Williams
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Raquel E Reinbolt
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Sagar D Sardesai
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Anne M Noonan
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Daniel G Stover
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Mathew A Cherian
- The Ohio State University Comprehensive Cancer Center, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - William B Malarkey
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Janice K Kiecolt-Glaser
- Institute for Behavioral Medicine Research, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
- Department of Psychiatry and Behavioral Health, The Ohio State University College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
28
|
Mrowietz U, Sümbül M, Gerdes S. Depression, a major comorbidity of psoriatic disease, is caused by metabolic inflammation. J Eur Acad Dermatol Venereol 2023; 37:1731-1738. [PMID: 37184282 DOI: 10.1111/jdv.19192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/26/2023] [Indexed: 05/16/2023]
Abstract
Psoriatic disease is a chronic, systemic immune-mediated inflammatory disorder comprising three major domains, skin, vascular and bone/joint inflammation. It is known for a long time that psoriatic disease is associated with a number of conditions such as hypertension, dyslipidemia, diabetes (metabolic syndrome) and depression. Up to one out of five people with psoriasis show concomitant depression. In the past, this was attributed to psychological stress of suffering from a chronic condition that is often visible and itchy, leading to stigmatization and adding to a significant burden of disease. Recent data provide evidence that depression associated with psoriatic disease is linked to the specific inflammatory pattern with IL-23, IL-17 family cytokines, TNF, IL-6 and IL-8 causing neuroinflammation and subsequently depression or depressive behaviour and/or anxiety. Psoriatic disease shows a distinct pattern of immune cells (e.g. dendritic cells, Th17 cells, neutrophils), mediators (e.g. IL-17A/F, IL-23, TNF) and tissue-related factors in all major domains that is different from other inflammatory dermatoses. There is a striking similarity between the inflammatory pattern in psoriatic disease and neuroinflammation that leads to depression. A number of risk factors have been identified in psoriatic disease, the most important of which are obesity and tobacco smoking. Obesity is known as a major risk factor for depression and anxiety due to its inflammatory signature. Apart from psychotherapy and anti-depressive medication, targeted treatments for psoriasis, including TNF, IL-17 and IL-23 inhibitors, can improve depression/depressive symptoms. The review summarizes the current knowledge about depression as a comorbidity in psoriatic disease.
Collapse
Affiliation(s)
- U Mrowietz
- Psoriasis Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - M Sümbül
- Psoriasis Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Kiel, Germany
| | - S Gerdes
- Psoriasis Center at the Department of Dermatology, University Medical Center Schleswig-Holstein, Kiel, Germany
| |
Collapse
|
29
|
Roldan L, Isaza C, Ospina J, Montoya C, Domínguez J, Orrego S, Correa S. A Comparative Study of HA/DBM Compounds Derived from Bovine and Porcine for Bone Regeneration. J Funct Biomater 2023; 14:439. [PMID: 37754853 PMCID: PMC10532284 DOI: 10.3390/jfb14090439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 07/25/2023] [Accepted: 08/17/2023] [Indexed: 09/28/2023] Open
Abstract
This comparative study investigated the tissue regeneration and inflammatory response induced by xenografts comprised of hydroxyapatite (HA) and demineralized bone matrix (DBM) extracted from porcine (P) and bovine (B) sources. First, extraction of HA and DBM was independently conducted, followed by chemical and morphological characterization. Second, mixtures of HA/DBM were prepared in 50/50 and 60/40 concentrations, and the chemical, morphological, and mechanical properties were evaluated. A rat calvarial defect model was used to evaluate the tissue regeneration and inflammatory responses at 3 and 6 months. The commercial allograft DBM Puros® was used as a clinical reference. Different variables related to tissue regeneration were evaluated, including tissue thickness regeneration (%), amount of regenerated bone area (%), and amount of regenerated collagen area (%). The inflammatory response was evaluated by quantifying the blood vessel area. Overall, tissue regeneration from porcine grafts was superior to bovine. After 3 months of implantation, the tissue thickness regeneration in the 50/50P compound and the commercial DBM was significantly higher (~99%) than in the bovine materials (~23%). The 50/50P and DBM produced higher tissue regeneration than the naturally healed controls. Similar trends were observed for the regenerated bone and collagen areas. The blood vessel area was correlated with tissue regeneration in the first 3 months of evaluation. After 6 months of implantation, HA/DBM compounds showed less regenerated collagen than the DBM-only xenografts. In addition, all animal-derived xenografts improved tissue regeneration compared with the naturally healed defects. No clinical complications associated with any implanted compound were noted.
Collapse
Affiliation(s)
- Lina Roldan
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
| | - Catalina Isaza
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
| | - Juan Ospina
- Centro de Investigación y Desarrollo Cárnico, Industrias de Alimentos Zenú S.A.S., Grupo Nutresa, Medellín 050044, Colombia;
| | - Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
| | - José Domínguez
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, PA 19122, USA; (C.M.); (S.O.)
- Bioengineering Department, College of Engineering, Temple University, Philadelphia, PA 191122, USA
| | - Santiago Correa
- Grupo de Investigación en Bioingeniería (GIB), Universidad EAFIT, Medellín 050022, Colombia; (L.R.); (C.I.)
- Escuela de Ciencias Aplicadas e Ingeniería, Universidad EAFIT, Medellín 050022, Colombia
| |
Collapse
|
30
|
Climie RE, Alastruey J, Mayer CC, Schwarz A, Laucyte-Cibulskiene A, Voicehovska J, Bianchini E, Bruno RM, Charlton PH, Grillo A, Guala A, Hallab M, Hametner B, Jankowski P, Königstein K, Lebedeva A, Mozos I, Pucci G, Puzantian H, Terentes-Printzios D, Yetik-Anacak G, Park C, Nilsson PM, Weber T. Vascular ageing: moving from bench towards bedside. Eur J Prev Cardiol 2023; 30:1101-1117. [PMID: 36738307 PMCID: PMC7614971 DOI: 10.1093/eurjpc/zwad028] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/20/2022] [Accepted: 01/12/2023] [Indexed: 02/05/2023]
Abstract
Prevention of cardiovascular disease (CVD) remains one of the largest public health challenges of our time. Identifying individuals at increased cardiovascular risk at an asymptomatic, sub-clinical stage is of paramount importance for minimizing disease progression as well as the substantial health and economic burden associated with overt CVD. Vascular ageing (VA) involves the deterioration in vascular structure and function over time and ultimately leads to damage in the heart, brain, kidney, and other organs. Vascular ageing encompasses the cumulative effect of all cardiovascular risk factors on the arterial wall over the life course and thus may help identify those at elevated cardiovascular risk, early in disease development. Although the concept of VA is gaining interest clinically, it is seldom measured in routine clinical practice due to lack of consensus on how to characterize VA as physiological vs. pathological and various practical issues. In this state-of-the-art review and as a network of scientists, clinicians, engineers, and industry partners with expertise in VA, we address six questions related to VA in an attempt to increase knowledge among the broader medical community and move the routine measurement of VA a little closer from bench towards bedside.
Collapse
Affiliation(s)
- Rachel E. Climie
- Menzies Institute for Medical Research, University of Tasmania, 17 Liverpool St, 7000 Hobart, Australia
- Sports Cardiology, Baker Heart and Diabetes Institute, 99 Commercial Rd, Melbourne 3000, Australia
- Integrative Epidemiology of Cardiovascular Disease, Université de Paris, INSERM, U970, Paris Cardiovascular Research Center (PARCC), 56 rue Leblanc, 75015 Paris, France
| | - Jordi Alastruey
- Department of Biomedical Engineering, School of Biomedical Engineering and Imaging Sciences, King’s College London, 249 Westminster Bridge Rd, London SE1 7EH, UK
| | - Christopher C. Mayer
- Medical Signal Analysis, Center for Health & Bioresources, AIT Austrian Institute of Technology, Giefinggasse 4, 1210 Vienna, Austria
| | - Achim Schwarz
- ALF Distribution GmbH, Stephanstrasse 19, 52064 Aachen, Germany
| | - Agne Laucyte-Cibulskiene
- Department of Clinical Sciences, Lund University, Skane University Hospital, Sölvegatan 19 - BMC F12, 221 84 Lund, Malmö, Sweden
- Faculty of Medicine, Vilnius University, M. K. C iurlionio g. 21, 03101 Vilnius, Lithuania
| | - Julija Voicehovska
- Department of Internal Diseases, Riga Stradins University, Dzirciema str. 16, Riga, L-1007, Latvia
- Nephrology and Renal Replacement Therapy Clinics, Riga East University Hospital, Hipokrata str. 2, Riga, LV-1079, Latvia
| | - Elisabetta Bianchini
- Institute of Clinical Physiology, Italian National Research Council (CNR), Via Moruzzi, 1, 56124 Pisa (PI), Italy
| | - Rosa-Maria Bruno
- Integrative Epidemiology of Cardiovascular Disease, Université de Paris, INSERM, U970, Paris Cardiovascular Research Center (PARCC), 56 rue Leblanc, 75015 Paris, France
| | - Peter H. Charlton
- Department of Public Health and Primary Care, University of Cambridge, Strangeways Research Laboratory, 2 Worts Causeway, Cambridge CB1 8RN, UK
| | - Andrea Grillo
- Medicina Clinica, Department of Medicine, Surgery and Health Sciences, University of Trieste, Strada di Fiume 447, 34149 Trieste, Italy
| | - Andrea Guala
- Vall d’Hebron Institut de Recerca (VHIR), Paseo de la Vall d’Hebron, 129, 08035 Barcelona, Spain
| | - Magid Hallab
- Clinique Bizet, 23 Georges Bizet, 75116 Paris, France
| | - Bernhard Hametner
- Medical Signal Analysis, Center for Health & Bioresources, AIT Austrian Institute of Technology, Giefinggasse 4, 1210 Vienna, Austria
| | - Piotr Jankowski
- Department of Internal Medicine and Geriatric Cardiology, Centre of Postgraduate Medical Education, 231 Czerniakowska St., 00-416 Warsaw, Poland
| | - Karsten Königstein
- Department of Sport, Exercise and Health (DSBG) University of Basel, Grosse Allee 6, 4052 Basel, Switzerland
| | - Anna Lebedeva
- Department of Internal Medicine and Cardiology, Dresden Heart Centre, Dresden University of Technology, Fetscher str. 76, 01307 Dresden, Germany
| | - Ioana Mozos
- Department of Functional Sciences-Pathophysiology, Center for Translational Research and Systems Medicine, ‘Victor Babes’ University of Medicine and Pharmacy, T. Vladimirescu Street 14, 300173 Timisoara, Romania
| | - Giacomo Pucci
- Unit of Internal Medicine, Terni University Hospital - Department of Medicine and Surgery, University of Perugia, Terni, Italy
| | - Houry Puzantian
- Hariri School of Nursing, American University of Beirut, P.O. Box 11-0236, Riad El Solh 1107 2020, Beirut, Lebanon
| | - Dimitrios Terentes-Printzios
- First Department of Cardiology, Hippokration Hospital, Medical School, National and Kapodistrian University of Athens, 114 Vasilissis Sofias Avenue, 11527 Athens, Greece
| | - Gunay Yetik-Anacak
- Department of Pharmacology, Faculty of Pharmacy, Acibadem Mehmet Ali Aydinlar University, Kayisdagi Cad. No:32 Atasehir, 34752 Istanbul, Turkey
| | - Chloe Park
- MRC Unit for Lifelong Health and Ageing at UCL, 1-19 Torrington Place, London WC1E 7HB, UK; and
| | - Peter M. Nilsson
- Department of Clinical Sciences, Lund University, Skane University Hospital, Sölvegatan 19 - BMC F12, 221 84 Lund, Malmö, Sweden
| | - Thomas Weber
- Cardiology Department, Klinikum Wels-Grieskirchen, Grieskirchnerstrasse 42, 4600 Wels, Austria
| |
Collapse
|
31
|
Zhang W, Zhao J, Deng L, Ishimwe N, Pauli J, Wu W, Shan S, Kempf W, Ballantyne MD, Kim D, Lyu Q, Bennett M, Rodor J, Turner AW, Lu YW, Gao P, Choi M, Warthi G, Kim HW, Barroso MM, Bryant WB, Miller CL, Weintraub NL, Maegdefessel L, Miano JM, Baker AH, Long X. INKILN is a Novel Long Noncoding RNA Promoting Vascular Smooth Muscle Inflammation via Scaffolding MKL1 and USP10. Circulation 2023; 148:47-67. [PMID: 37199168 PMCID: PMC10330325 DOI: 10.1161/circulationaha.123.063760] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 04/14/2023] [Indexed: 05/19/2023]
Abstract
BACKGROUND Activation of vascular smooth muscle cell (VSMC) inflammation is vital to initiate vascular disease. The role of human-specific long noncoding RNAs in VSMC inflammation is poorly understood. METHODS Bulk RNA sequencing in differentiated human VSMCs revealed a novel human-specific long noncoding RNA called inflammatory MKL1 (megakaryoblastic leukemia 1) interacting long noncoding RNA (INKILN). INKILN expression was assessed in multiple in vitro and ex vivo models of VSMC phenotypic modulation as well as human atherosclerosis and abdominal aortic aneurysm. The transcriptional regulation of INKILN was verified through luciferase reporter and chromatin immunoprecipitation assays. Loss-of-function and gain-of-function studies and multiple RNA-protein and protein-protein interaction assays were used to uncover a mechanistic role of INKILN in the VSMC proinflammatory gene program. Bacterial artificial chromosome transgenic mice were used to study INKILN expression and function in ligation injury-induced neointimal formation. RESULTS INKILN expression is downregulated in contractile VSMCs and induced in human atherosclerosis and abdominal aortic aneurysm. INKILN is transcriptionally activated by the p65 pathway, partially through a predicted NF-κB (nuclear factor kappa B) site within its proximal promoter. INKILN activates proinflammatory gene expression in cultured human VSMCs and ex vivo cultured vessels. INKILN physically interacts with and stabilizes MKL1, a key activator of VSMC inflammation through the p65/NF-κB pathway. INKILN depletion blocks interleukin-1β-induced nuclear localization of both p65 and MKL1. Knockdown of INKILN abolishes the physical interaction between p65 and MKL1 and the luciferase activity of an NF-κB reporter. Furthermore, INKILN knockdown enhances MKL1 ubiquitination through reduced physical interaction with the deubiquitinating enzyme USP10 (ubiquitin-specific peptidase 10). INKILN is induced in injured carotid arteries and exacerbates ligation injury-induced neointimal formation in bacterial artificial chromosome transgenic mice. CONCLUSIONS These findings elucidate an important pathway of VSMC inflammation involving an INKILN/MKL1/USP10 regulatory axis. Human bacterial artificial chromosome transgenic mice offer a novel and physiologically relevant approach for investigating human-specific long noncoding RNAs under vascular disease conditions.
Collapse
Affiliation(s)
- Wei Zhang
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jinjing Zhao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Lin Deng
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Nestor Ishimwe
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Jessica Pauli
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany
| | - Wen Wu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Shengshuai Shan
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Wolfgang Kempf
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany
| | | | - David Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Qing Lyu
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Matthew Bennett
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Julie Rodor
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Adam W. Turner
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
| | - Yao Wei Lu
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Ping Gao
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Mihyun Choi
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Ganesh Warthi
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Ha Won Kim
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Margarida M Barroso
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - William B. Bryant
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Clint L. Miller
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, USA
| | - Neal L. Weintraub
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Lars Maegdefessel
- Department for Vascular and Endovascular Surgery, Klinikum rechts der Isar, Technical University Munich, Germany
- German Center for Cardiovascular Research (DZHK, partner site Munich), Germany
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Joseph M. Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Andrew H Baker
- Centre for Cardiovascular Science University of Edinburgh, Edinburgh, Scotland
| | - Xiaochun Long
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| |
Collapse
|
32
|
Regolo M, Sorce A, Vaccaro M, Colaci M, Stancanelli B, Natoli G, Motta M, Isaia I, Castelletti F, Giangreco F, Fichera D, Aparo P, Lanzafame A, Russo M, Santangelo N, Noto P, Malatino L. Assessing Humoral Immuno-Inflammatory Pathways Associated with Respiratory Failure in COVID-19 Patients. J Clin Med 2023; 12:4057. [PMID: 37373750 DOI: 10.3390/jcm12124057] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/12/2023] [Accepted: 06/13/2023] [Indexed: 06/29/2023] Open
Abstract
All severe cases of SARS-CoV-2 infections are characterized by a high risk of disease progression towards ARDS, leading to a bad outcome. Respiratory symptoms in COVID-19 patients often do not correspond to disease's worsening. In our sample, median age was 74 years (72-75) and 54% were men. The median period of hospitalization was 9 days. Firstly, we observed a significant asynchronous trend of neutrophil-to-lymphocyte ratio (NLR) and C-reactive protein (CRP) in 764 selected among 963 patients, who were consecutively recruited in two hospitals (Cannizzaro, S. Marco) in Catania, Italy. NLR values in deceased patients showed an increase from baseline over time. By contrast, CRP tended to fall from baseline to median day of hospitalization in all three subgroups, but steeply increased at the end of hospitalization only in ICU-admitted patients. Then, we evaluated the relationships between NLR and CRP as continuous variables with PaO2/FiO2 ratio (P/F). NLR was an independent predictor of mortality (HR: 1.77, p < 0.0001), while ICU admission was more significantly associated with CRP (HR: 1.70, p < 0.0001). Finally, age, neutrophils, CRP, and lymphocytes are significantly and directly linked to P/F, while the influence of inflammation on P/F, reflected by CRP, was also mediated by neutrophils.
Collapse
Affiliation(s)
- Matteo Regolo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Alessandra Sorce
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Medical Specialties, "G. D'Alessandro" (PROMISE), Unit of Nephrology and Hypertension, European Society of Hypertension Excellence Centre, University of Palermo, 90133 Palermo, Italy
| | - Mauro Vaccaro
- Department of Emergency Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Michele Colaci
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Benedetta Stancanelli
- Unit of Internal Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Giuseppe Natoli
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Massimo Motta
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Ivan Isaia
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Federica Castelletti
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
| | - Federica Giangreco
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Daniela Fichera
- Department of Emergency Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Paola Aparo
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Alessandra Lanzafame
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Mario Russo
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Nicola Santangelo
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| | - Paola Noto
- Department of Emergency Medicine, San Marco-Polyclinic Academic Hospital, 95121 Catania, Italy
| | - Lorenzo Malatino
- Department of Clinical and Experimental Medicine, University of Catania, 95123 Catania, Italy
- Academic Unit of Internal Medicine, Cannizzaro Hospital, 95126 Catania, Italy
| |
Collapse
|
33
|
Longtine AG, Venkatasubramanian R, Zigler MC, Lindquist AJ, Mahoney SA, Greenberg NT, VanDongen NS, Ludwig KR, Moreau KL, Seals DR, Clayton ZS. Female C57BL/6N mice are a viable model of aortic aging in women. Am J Physiol Heart Circ Physiol 2023; 324:H893-H904. [PMID: 37115626 PMCID: PMC10202480 DOI: 10.1152/ajpheart.00120.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 04/29/2023]
Abstract
The aorta stiffens with aging in both men and women, which predicts cardiovascular mortality. Aortic wall structural and extracellular matrix (ECM) remodeling, induced in part by chronic low-grade inflammation, contribute to aortic stiffening. Male mice are an established model of aortic aging. However, there is little information regarding whether female mice are an appropriate model of aortic aging in women, which we aimed to elucidate in the present study. We assessed two strains of mice and found that in C57BL/6N mice, in vivo aortic stiffness (pulse wave velocity, PWV) was higher with aging in both sexes, whereas in B6D2F1 mice, PWV was higher in old versus young male mice, but not in old versus young female mice. Because the age-related stiffening that occurs in men and women was reflected in male and female C57BL/6N mice, we examined the mechanisms of stiffening in this strain. In both sexes, aortic modulus of elasticity (pin myography) was lower in old mice, occurred in conjunction with and was related to higher plasma levels of the elastin-degrading enzyme matrix metalloproteinase-9 (MMP-9), and was accompanied by higher numbers of aortic elastin breaks and higher abundance of adventitial collagen-1. Plasma levels of the inflammatory cytokines interferon-γ, interleukin 6, and monocyte chemoattractant protein-1 were higher in both sexes of old mice. In conclusion, female C57BL/6N mice exhibit aortic stiffening, reduced modulus of elasticity and structural/ECM remodeling, and associated increases in MMP-9 and systemic inflammation with aging, and thus are an appropriate model of aortic aging in women.NEW & NOTEWORTHY Our study demonstrates that with aging, female C57BL/6N mice exhibit higher in vivo aortic stiffness, reduced modulus of elasticity, aortic wall structural and extracellular matrix remodeling, and elevations in systemic inflammation. These changes are largely reflective of those that occur with aging in women. Thus, female C57BL/6N mice are a viable model of human aortic aging and the utility of these animals should be considered in future biomedical investigations.
Collapse
Affiliation(s)
- Abigail G Longtine
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | | | - Melanie C Zigler
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Alexandra J Lindquist
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nathan T Greenberg
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Nicholas S VanDongen
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Katelyn R Ludwig
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Kerrie L Moreau
- University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado, United States
| |
Collapse
|
34
|
Pitzer Mutchler A, Huynh L, Patel R, Lam T, Bain D, Jamison S, Kirabo A, Ray EC. The role of dietary magnesium deficiency in inflammatory hypertension. Front Physiol 2023; 14:1167904. [PMID: 37293263 PMCID: PMC10244581 DOI: 10.3389/fphys.2023.1167904] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 05/10/2023] [Indexed: 06/10/2023] Open
Abstract
Nearly 30% of adults consume less than the estimated average daily requirement of magnesium (Mg2+), and commonly used medications, such as diuretics, promote Mg2+ deficiency. Higher serum Mg2+ levels, increased dietary Mg2+ in-take, and Mg2+ supplementation are each associated with lower blood pressure, suggesting that Mg2+-deficiency contributes to the pathogenesis of hypertension. Antigen-presenting cells, such as monocytes and dendritic cells, are well-known to be involved in the pathogenesis of hypertension. In these cells, processes implicated as necessary for increased blood pressure include activation of the NLRP3 inflammasome, IL-1β production, and oxidative modification of fatty acids such as arachidonic acid, forming isolevuglandins (IsoLGs). We hypothesized that increased blood pressure in response to dietary Mg2+-depletion leads to increased NLRP3, IL-1β, and IsoLG production in antigen presenting cells. We found that a Mg2+-depleted diet (0.01% Mg2+ diet) increased blood pressure in mice compared to mice fed a 0.08% Mg2+ diet. Mg2+-depleted mice did not exhibit an increase in total body fluid, as measured by quantitative magnetic resonance. Plasma IL-1β concentrations were increased (0.13 ± 0.02 pg/mL vs. 0.04 ± 0.02 pg/mL). Using flow cytometry, we observed increased NLRP3 and IL-1β expression in antigen-presenting cells from spleen, kidney, and aorta. We also observed increased IsoLG production in antigen-presenting cells from these organs. Primary culture of CD11c+ dendritic cells confirmed that low extracellular Mg2+ exerts a direct effect on these cells, stimulating IL-1β and IL-18 production. The present findings show that NLRP3 inflammasome activation and IsoLG-adduct formation are stimulated when dietary Mg2+ is depleted. Interventions and increased dietary Mg2+ consumption may prove beneficial in decreasing the prevalence of hypertension and cardiovascular disease.
Collapse
Affiliation(s)
- Ashley Pitzer Mutchler
- Vanderbilt University Department of Medicine, Division of Clinical Pharmacology, Nashville, TN, United States
| | - Linh Huynh
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Ritam Patel
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Tracey Lam
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| | - Daniel Bain
- University of Pittsburgh Department of Geology, Pittsburgh, PA, United States
| | - Sydney Jamison
- Meharry Medical College Nashville, Nashville, TN, United States
| | - Annet Kirabo
- Vanderbilt University Department of Medicine, Division of Clinical Pharmacology, Nashville, TN, United States
| | - Evan C. Ray
- University of Pittsburgh Department of Medicine, Renal-Electrolyte Division, Pittsburgh, PA, United States
| |
Collapse
|
35
|
Zhao X, Kiyozuka K, Konishi A, Kawabata-Iwakawa R, Minamishima YA, Obinata H. Actin-binding protein Filamin B regulates the cell-surface retention of endothelial sphingosine 1-phosphate receptor 1. J Biol Chem 2023:104851. [PMID: 37220855 PMCID: PMC10300261 DOI: 10.1016/j.jbc.2023.104851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 05/09/2023] [Accepted: 05/11/2023] [Indexed: 05/25/2023] Open
Abstract
Sphingosine 1-phosphate receptor 1 (S1PR1) is a G protein-coupled receptor essential for vascular development and postnatal vascular homeostasis. When exposed to sphingosine 1-phosphate (S1P) in the blood of ∼1 μM, S1PR1 in endothelial cells retains cell-surface localization, while lymphocyte S1PR1 shows almost complete internalization, suggesting the cell-surface retention of S1PR1 is endothelial cell-specific. To identify regulating factors that function to retain S1PR1 on the endothelial cell surface, here we utilized an enzyme-catalyzed proximity labeling technique followed by proteomic analyses. We identified Filamin B (FLNB), an actin-binding protein involved in F-actin cross-linking, as a candidate regulating protein. We show FLNB knockdown by RNA interference induced massive internalization of S1PR1 into early endosomes, which was partially ligand-dependent and required receptor phosphorylation. Further investigation showed FLNB was also important for the recycling of internalized S1PR1 back to the cell surface. FLNB knockdown did not affect the localization of S1PR3, another S1P receptor subtype expressed in endothelial cells, nor did it affect localization of ectopically expressed β2-adrenergic receptor. Functionally, we show FLNB knockdown in endothelial cells impaired S1P-induced intracellular phosphorylation events and directed cell migration and enhancement of the vascular barrier. Taken together, our results demonstrate that FLNB is a novel regulator critical for S1PR1 cell-surface localization and thereby proper endothelial cell function.
Collapse
Affiliation(s)
- Xian Zhao
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Keisuke Kiyozuka
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Akimitsu Konishi
- Department of Biochemistry, Gunma University Graduate School of Medicine, Gunma, Japan
| | - Reika Kawabata-Iwakawa
- Division of Integrated Oncology Research, Gunma University Initiative for Advanced Research, Gunma University, Gunma, Japan
| | | | - Hideru Obinata
- Education and Research Support Center, Gunma University Graduate School of Medicine, Gunma, Japan.
| |
Collapse
|
36
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
37
|
Badimon A, Torrente D, Norris EH. Vascular Dysfunction in Alzheimer's Disease: Alterations in the Plasma Contact and Fibrinolytic Systems. Int J Mol Sci 2023; 24:7046. [PMID: 37108211 PMCID: PMC10138543 DOI: 10.3390/ijms24087046] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 03/30/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disease, affecting millions of people worldwide. The classical hallmarks of AD include extracellular beta-amyloid (Aβ) plaques and neurofibrillary tau tangles, although they are often accompanied by various vascular defects. These changes include damage to the vasculature, a decrease in cerebral blood flow, and accumulation of Aβ along vessels, among others. Vascular dysfunction begins early in disease pathogenesis and may contribute to disease progression and cognitive dysfunction. In addition, patients with AD exhibit alterations in the plasma contact system and the fibrinolytic system, two pathways in the blood that regulate clotting and inflammation. Here, we explain the clinical manifestations of vascular deficits in AD. Further, we describe how changes in plasma contact activation and the fibrinolytic system may contribute to vascular dysfunction, inflammation, coagulation, and cognitive impairment in AD. Given this evidence, we propose novel therapies that may, alone or in combination, ameliorate AD progression in patients.
Collapse
Affiliation(s)
| | | | - Erin H. Norris
- Patricia and John Rosenwald Laboratory of Neurobiology and Genetics, The Rockefeller University, 1230 York Avenue, New York, NY 10065, USA
| |
Collapse
|
38
|
Mo S, Wang Y, Yuan X, Wu W, Zhao H, Wei H, Qin H, Jiang H, Qin S. Identification of common signature genes and pathways underlying the pathogenesis association between nonalcoholic fatty liver disease and atherosclerosis. Front Cardiovasc Med 2023; 10:1142296. [PMID: 37063958 PMCID: PMC10098172 DOI: 10.3389/fcvm.2023.1142296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/07/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundAtherosclerosis (AS) is one of the leading causes of the cardio-cerebral vascular incident. The constantly emerging evidence indicates a close association between nonalcoholic fatty liver disease (NAFLD) and AS. However, the exact molecular mechanisms underlying the correlation between these two diseases remain unclear. This study proposed exploring the common signature genes, pathways, and immune cells among AS and NAFLD.MethodsThe common differentially expressed genes (co-DEGs) with a consistent trend were identified via bioinformatic analyses of the Gene Expression Omnibus (GEO) datasets GSE28829 and GSE49541, respectively. Further, the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses were performed. We utilized machine learning algorithms of lasso and random forest (RF) to identify the common signature genes. Then the diagnostic nomogram models and receiver operator characteristic curve (ROC) analyses were constructed and validated with external verification datasets. The gene interaction network was established via the GeneMANIA database. Additionally, gene set enrichment analysis (GSEA), gene set variation analysis (GSVA), and immune infiltration analysis were performed to explore the co-regulated pathways and immune cells.ResultsA total of 11 co-DEGs were identified. GO and KEGG analyses revealed that co-DEGs were mainly enriched in lipid catabolic process, calcium ion transport, and regulation of cytokine. Moreover, three common signature genes (PLCXD3, CCL19, and PKD2) were defined. Based on these genes, we constructed the efficiently predictable diagnostic models for advanced AS and NAFLD with the nomograms, evaluated with the ROC curves (AUC = 0.995 for advanced AS, 95% CI 0.971–1.0; AUC = 0.973 for advanced NAFLD, 95% CI 0.938–0.998). In addition, the AUC of the verification datasets had a similar trend. The NOD-like receptors (NLRs) signaling pathway might be the most crucial co-regulated pathway, and activated CD4 T cells and central memory CD4 T cells were significantly excessive infiltration in advanced NAFLD and AS.ConclusionWe identified three common signature genes (PLCXD3, CCL19, and PKD2), co-regulated pathways, and shared immune features of NAFLD and AS, which might provide novel insights into the molecular mechanism of NAFLD complicated with AS.
Collapse
Affiliation(s)
- Shuangyang Mo
- Gastroenterology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Gastroenterology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Yingwei Wang
- Gastroenterology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Xin Yuan
- Cardiovascular Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Wenhong Wu
- Gastroenterology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Huaying Zhao
- Gastroenterology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Haixiao Wei
- Gastroenterology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Haiyan Qin
- Gastroenterology Department, Liuzhou Peoples’ Hospital Affiliated to Guangxi Medical University, Liuzhou, China
| | - Haixing Jiang
- Gastroenterology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Correspondence: Shanyu Qin Haixing Jiang
| | - Shanyu Qin
- Gastroenterology Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Correspondence: Shanyu Qin Haixing Jiang
| |
Collapse
|
39
|
Froldi G. The Use of Medicinal Plants in Blood Vessel Diseases: The Influence of Gender. Life (Basel) 2023; 13:life13040866. [PMID: 37109395 PMCID: PMC10147070 DOI: 10.3390/life13040866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/17/2023] [Accepted: 03/23/2023] [Indexed: 04/29/2023] Open
Abstract
Data available in the literature on the use of herbal products to treat inflammation-related vascular diseases were considered in this study, while also assessing the influence of gender. To this end, the articles published in PubMed over the past 10 years that described the use of plant extracts in randomized clinical trials studying the effectiveness in vascular pathologies were analyzed. The difference in efficacy of plant-derived preparations in female and male subjects was always considered when reporting. The safety profiles of the selected plants were described, reporting unwanted effects in humans and also by searching the WHO database (VigiBase®). The medicinal plants considered were Allium sativum, Campomanesia xanthocarpa, Sechium edule, Terminalia chebula. Additionally, an innovative type of preparation consisting of plant-derived nanovesicles was also reported.
Collapse
Affiliation(s)
- Guglielmina Froldi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
40
|
Aortic Stiffness: A Major Risk Factor for Multimorbidity in the Elderly. J Clin Med 2023; 12:jcm12062321. [PMID: 36983321 PMCID: PMC10058400 DOI: 10.3390/jcm12062321] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/14/2023] [Accepted: 03/15/2023] [Indexed: 03/19/2023] Open
Abstract
Multimorbidity, the coexistence of multiple health conditions in an individual, has emerged as one of the greatest challenges facing health services, and this crisis is partly driven by the aging population. Aging is associated with increased aortic stiffness (AoStiff), which in turn is linked with several morbidities frequently affecting and having disastrous consequences for the elderly. These include hypertension, ischemic heart disease, heart failure, atrial fibrillation, chronic kidney disease, anemia, ischemic stroke, and dementia. Two or more of these disorders (multimorbidity) often coexist in the same elderly patient and the specific multimorbidity pattern depends on several factors including sex, ethnicity, common morbidity routes, morbidity interactions, and genomics. Regular exercise, salt restriction, statins in patients at high atherosclerotic risk, and stringent blood pressure control are interventions that delay progression of AoStiff and most likely decrease multimorbidity in the elderly.
Collapse
|
41
|
Ahn S, Howie-Esquivel J, Davis EM, Chung ML, Lobo JM, Logan JG. Association of disrupted sleep with 24-hour blood pressure variability in caregivers. Heart Lung 2023; 60:45-51. [PMID: 36905754 DOI: 10.1016/j.hrtlng.2023.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 02/21/2023] [Accepted: 02/26/2023] [Indexed: 03/11/2023]
Abstract
BACKGROUND A growing body of research highlights the negative impact of caregiving on cardiovascular disease (CVD) risk. OBJECTIVES This study aimed to examine associations of psychological symptoms and sleep quality with 24-hour blood pressure variability (BPV), which is an independent predictor of CVD, among family caregivers of community-dwelling individuals with chronic illness. METHODS For this cross-sectional study, we assessed caregiving burden and depressive symptoms using questionnaires and 7-day sleep quality (i.e., number of awakenings, wake after sleep onset, sleep efficiency) using an actigraph. The participants carried out a 24-hour ambulatory BP monitoring for systolic and diastolic BPV over 24 h and during awake/sleep times. We performed Pearson's correlations and multiple linear regression. RESULTS The analytic sample consisted of 30 caregivers (25 female; mean age 62 years). The number of awakenings during sleep was positively correlated with systolic BPV-awake (r = 0.426, p = 0.019) and diastolic BPV-awake (r = 0.422, p = 0.020). Sleep efficiency was negatively correlated with diastolic BPV-awake (r = -0.368, p = 0.045). Caregiving burden and depressive symptoms were not correlated with BPV. After controlling for age and mean arterial pressure, the number of awakenings was significantly associated with increased systolic BPV-24 h (β = 0.194, p = 0.018) and systolic BPV-awake (β = 0.280, p = 0.002), respectively. CONCLUSIONS Caregivers' disrupted sleep may play a role in increased CVD risk. While these findings should be confirmed in large clinical studies, improving sleep quality would need to be considered in CVD prevention for caregivers.
Collapse
Affiliation(s)
- Soojung Ahn
- School of Nursing, Vanderbilt University, Nashville, TN, USA.
| | | | - Eric M Davis
- Division of Pulmonary and Critical Care, Department of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Misook L Chung
- College of Nursing, University of Kentucky, Lexington, KY, USA
| | - Jennifer M Lobo
- Department of Public Health Sciences, University of Virginia, Charlottesville, VA, USA
| | - Jeongok G Logan
- School of Nursing, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
42
|
Liu YH, Chen SC, Lee WH, Chen YC, Huang JC, Wu PY, Hung CH, Kuo CH, Su HM. Components of the Complete Blood Count as a Risk Predictor for Incident Hypertension in a Large Taiwanese Population Follow-up Study. Circ J 2023; 87:456-462. [PMID: 36261331 DOI: 10.1253/circj.cj-22-0512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND Previous studies investigating the relationship between hypertension (HT) and hematological parameters report inconsistent results, and most them included a small number of participants or only conducted a cross-sectional analysis of 1 or 2 hematological factors. Moreover, no large cohort follow-up studies have investigated this topic. The aim of this longitudinal study was to explore associations between components of the complete blood count (CBC) and incident HT using data from a large Taiwanese biobank. METHODS AND RESULTS Hematological parameters including white blood cell (WBC) count, red blood cell (RBC) count, hemoglobin, hematocrit (HCT), and platelet count were evaluated. We included 21,293 participants who did not have HT at baseline and followed them for a mean period of 3.9 years. During follow-up, 3,002 participants with new-onset HT (defined as incident HT) were identified. Univariable analysis revealed that high WBC count, high RBC count, high hemoglobin, high HCT, and low platelet count were associated with incident HT. Multivariable analysis after adjusting potential confounding factors found high WBC count (odds ratio [OR], 1.057; 95% confidence interval [CI], 1.028 to 1.087; P<0.001) and high HCT (OR, 1.023; 95% CI, 1.010 to 1.036; P<0.001) were still significantly associated with incident HT. CONCLUSIONS High WBC count and high HCT were associated with incident HT.
Collapse
Affiliation(s)
- Yi-Hsueh Liu
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital
| | - Szu-Chia Chen
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University
- Research Center for Environmental Medicine, Kaohsiung Medical University
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
| | - Wen-Hsien Lee
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital
| | - Ying-Chih Chen
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital
| | - Jiun-Chi Huang
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
| | - Pei-Yu Wu
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
| | - Chih-Hsing Hung
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University
- Research Center for Environmental Medicine, Kaohsiung Medical University
| | - Chao-Hung Kuo
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
| | - Ho-Ming Su
- Faculty of Medicine, College of Medicine, Kaohsiung Medical University
- Department of Internal Medicine, Kaohsiung Municipal Siaogang Hospital
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University Hospital
| |
Collapse
|
43
|
Ehlers H, Nicolas A, Schavemaker F, Heijmans JPM, Bulst M, Trietsch SJ, van den Broek LJ. Vascular inflammation on a chip: A scalable platform for trans-endothelial electrical resistance and immune cell migration. Front Immunol 2023; 14:1118624. [PMID: 36761747 PMCID: PMC9903066 DOI: 10.3389/fimmu.2023.1118624] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023] Open
Abstract
The vasculature system plays a critical role in inflammation processes in the body. Vascular inflammatory mechanisms are characterized by disruption of blood vessel wall permeability together with increased immune cell recruitment and migration. There is a critical need to develop models that fully recapitulate changes in vascular barrier permeability in response to inflammatory conditions. We developed a scalable platform for parallel measurements of trans epithelial electrical resistance (TEER) in 64 perfused microfluidic HUVEC tubules under inflammatory conditions. Over 250 tubules where exposed to Tumor necrosis factor alpha (TNFα) and interferon gamma (INF-γ) or human peripheral blood mononuclear cells. The inflammatory response was quantified based on changes TEER and expression of ICAM and VE-cadherin. We observed changes in barrier function in the presence of both inflammatory cytokines and human peripheral blood mononuclear cells, characterized by decreased TEER values, increase in ICAM expression as well changes in endothelial morphology. OrganoPlate 3-lane64 based HUVEC tubules provide a valuable tool for inflammatory studies in an automation compatible manner. Continuous TEER measurements enable long term, sensitive assays for barrier studies. We propose the use of our platform as a powerful tool for modelling endothelial inflammation in combination with immune cell interaction that can be used to screen targets and drugs to treat chronic vascular inflammation.
Collapse
Affiliation(s)
- Haley Ehlers
- Mimetas B.V., Leiden, Netherlands,Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | - Arnaud Nicolas
- Mimetas B.V., Leiden, Netherlands,Leiden Academic Centre for Drug Research, Leiden University, Leiden, Netherlands
| | | | | | | | | | | |
Collapse
|
44
|
Zhang W, Zhao J, Deng L, Ishimwe N, Pauli J, Wu W, Shan S, Kempf W, Ballantyne MD, Kim D, Lyu Q, Bennett M, Rodor J, Turner AW, Lu YW, Gao P, Choi M, Warthi G, Kim HW, Barroso MM, Bryant WB, Miller CL, Weintraub NL, Maegdefessel L, Miano JM, Baker AH, Long X. INKILN is a novel long noncoding RNA promoting vascular smooth muscle inflammation via scaffolding MKL1 and USP10. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.07.522948. [PMID: 36711681 PMCID: PMC9881896 DOI: 10.1101/2023.01.07.522948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Background Activation of vascular smooth muscle cells (VSMCs) inflammation is vital to initiate vascular disease. However, the role of human-specific long noncoding RNAs (lncRNAs) in VSMC inflammation is poorly understood. Methods Bulk RNA-seq in differentiated human VSMCs revealed a novel human-specific lncRNA called IN flammatory M K L1 I nteracting L ong N oncoding RNA ( INKILN ). INKILN expression was assessed in multiple in vitro and ex vivo models of VSMC phenotypic modulation and human atherosclerosis and abdominal aortic aneurysm (AAA) samples. The transcriptional regulation of INKILN was determined through luciferase reporter system and chromatin immunoprecipitation assay. Both loss- and gain-of-function approaches and multiple RNA-protein and protein-protein interaction assays were utilized to uncover the role of INKILN in VSMC proinflammatory gene program and underlying mechanisms. Bacterial Artificial Chromosome (BAC) transgenic (Tg) mice were utilized to study INKLIN expression and function in ligation injury-induced neointimal formation. Results INKILN expression is downregulated in contractile VSMCs and induced by human atherosclerosis and abdominal aortic aneurysm. INKILN is transcriptionally activated by the p65 pathway, partially through a predicted NF-κB site within its proximal promoter. INKILN activates the proinflammatory gene expression in cultured human VSMCs and ex vivo cultured vessels. Mechanistically, INKILN physically interacts with and stabilizes MKL1, a key activator of VSMC inflammation through the p65/NF-κB pathway. INKILN depletion blocks ILIβ-induced nuclear localization of both p65 and MKL1. Knockdown of INKILN abolishes the physical interaction between p65 and MKL1, and the luciferase activity of an NF-κB reporter. Further, INKILN knockdown enhances MKL1 ubiquitination, likely through the reduced physical interaction with the deubiquitinating enzyme, USP10. INKILN is induced in injured carotid arteries and exacerbates ligation injury-induced neointimal formation in BAC Tg mice. Conclusions These findings elucidate an important pathway of VSMC inflammation involving an INKILN /MKL1/USP10 regulatory axis. Human BAC Tg mice offer a novel and physiologically relevant approach for investigating human-specific lncRNAs under vascular disease conditions.
Collapse
|
45
|
A Transient Inflammatory Response Induced by Lipopolysaccharide Infusion Lowers Markers of Endogenous Cholesterol and Bile Acid Synthesis in Healthy Normocholesterolemic Young Men. Biomedicines 2023; 11:biomedicines11010126. [PMID: 36672634 PMCID: PMC9855383 DOI: 10.3390/biomedicines11010126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 12/27/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
Inflammation is associated with changes in plasma lipids, lipoproteins, and cholesterol efflux capacity (CEC). It is unknown if the changes in lipids and lipoproteins during inflammation are related to changes in cholesterol absorption, synthesis, and bile acid synthesis. We, therefore, examined the effects of acute lipopolysaccharide (LPS)-induced transient systemic inflammation on lipids, lipoproteins, CEC, and markers of cholesterol metabolism. We also evaluated whether markers for cholesterol metabolism at baseline predict the intensity of the inflammatory response. Eight healthy young subjects received LPS infusion, and blood was sampled for the following 24 h. In addition to lipids, lipoproteins, and CEC, we also measured markers for cholesterol absorption and synthesis, bile acid synthesis, and inflammation. Compared with baseline, plasma total cholesterol, low-density lipoprotein cholesterol, and CEC decreased, while triglycerides increased in the 24 h following LPS infusion. TC-standardized levels of cholesterol synthesis markers (lathosterol, lanosterol, and desmosterol) and a bile acid synthesis marker (7α-OH-cholesterol) also decreased, with no changes in cholesterol absorption markers (campesterol, sitosterol, and cholestanol). Baseline TC-standardized levels of desmosterol and 7α-OH-cholesterol were positively correlated with concentrations of various inflammatory markers. Changes in TC-standardized desmosterol and 7α-OH-cholesterol were negatively correlated with concentrations of inflammatory markers. LPS infusion reduced endogenous cholesterol synthesis and bile acid synthesis in healthy young men.
Collapse
|
46
|
Salas LA, Kelsey KT. Hypertension Impacts Peripheral Blood Leukocyte Composition. Hypertension 2023; 80:54-56. [PMID: 36475861 PMCID: PMC9752179 DOI: 10.1161/hypertensionaha.122.20422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Lucas A Salas
- Department of Epidemiology, Geisel School of Medicine at Dartmouth, Dartmouth Cancer Center, Lebanon, NH (L.A.S.)
| | - Karl T Kelsey
- Department of Epidemiology and Pathology and Laboratory Medicine, Brown University, Providence, RI (K.T.K.)
| |
Collapse
|
47
|
Photoplethysmographic Measurement of Arterial Stiffness in Polish Patients with Long-COVID-19 Syndrome-The Results of a Cross-Sectional Study. Diagnostics (Basel) 2022; 12:diagnostics12123189. [PMID: 36553195 PMCID: PMC9777579 DOI: 10.3390/diagnostics12123189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/30/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022] Open
Abstract
The coronavirus disease 2019 (COVID-19) is associated with an increase in the incidence of cardiovascular diseases (CVD) that persists even several months after the onset of infection. COVID-19 may also have an impact on arterial stiffness, which is a risk factor for CVD. We aimed to analyze if and to what extent arterial stiffness measured by photoplethysmography differed among COVID-19 convalescents depending on the acute phase severity and time elapsed since disease onset. A total of 225 patients (mean age 58.98 ± 8.57 years, 54.7% women) were analyzed after COVID-19 hospitalization at the Cardiac Rehabilitation Department of the Ustron Health Resort (Poland). In the entire study population, no differences were found in the mean values of stiffness index (SI) and reflection index (RI) depending on the severity of the acute COVID-19 and the time since the onset of the disease. There were no differences in the heart rate (HR) according to the severity of acute COVID-19; the mean HR was higher in patients who had COVID-19 less than 12 weeks before the study than in convalescents more than 24 weeks after the acute disease (p = 0.002). The mean values of SI and RI were higher in men than in women (p < 0.001), while the heart rate (HR) was similar in both sexes (p = 0.286). However, multiple linear regression analyses after adjusting for factors influencing arterial stiffness, i.e., sex, age, body mass index, smoking status, hypertension, diabetes, the severity of the acute COVID-19, and the time from the disease onset, confirmed that age, sex, time from disease onset, and diabetes are the most important determinants that could influence arterial stiffness.
Collapse
|
48
|
Mossa EAM, Sayed KM, Awny I, Mohamed NA, Ali T, Hemdan SB, Helaly AA, Abdellatif MG, Farag RM, Alsmman AH, Mounir A. Expression of poly(ADP-ribose) polymerase-1 gene and optical coherence tomography angiographic parameters among patients with multiple sclerosis. BMJ Open Ophthalmol 2022. [DOI: 10.1136/bmjophth-2022-001157] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Back ground/aimsTo analyse different parameters of the macula, disc and their vascular affection using optical coherence tomography (OCT) and angiography (OCT-A) in patients with multiple sclerosis (MS) correlating these changes to PARP-1 gene expression in blood.MethodsThis cross-sectional study included 80 eyes of the clinically diagnosed relapsing-remitting phenotype of MS. The study included three groups; group (A) included 40 eyes of 20 patients with MS with a history of optic neuritis (MS+ON), group (B) included 40 eyes of 20 patients with MS without a history of ON (MS-ON) and group (C) (the control group) consisted of 40 eyes of 20 matched participants not suffering from any ocular or systemic disease. OCT and OCT-A, RTVue (Optovue, Fermont, California, USA) were done for all eyes for evaluating the macular and disc changes. Qualitative real-time PCR for estimation of PARP1 gene expression level was performed for all patients.ResultsPARP-1 gene expression level showed a significant difference in comparing the three groups, with the highest level being for the (ON+) group (p<0.0009). Significant negative correlations were found between PARP-1 gene expression level and central macular thickness, total macular volume and full foveal vessel density thickness. ROC curve constructed by plotting the area under the receiver operating characteristic curve value was (0.9) for PARP-1 gene expression level.ConclusionsPARP-1 may play an important role in the development of the ON cascade in patients with MS and may be a biomarker for diagnosing and a potential molecular target of ON in MS patients’ therapy. In addition to the OCT and OCT-angio changes that could be detected retrospectively, PARP-1 gene expression level could be considered a prospective detector to complete the full-blown picture of MS (ON+) early and prevent blindness.
Collapse
|
49
|
Miceli G, Basso MG, Rizzo G, Pintus C, Tuttolomondo A. The Role of the Coagulation System in Peripheral Arterial Disease: Interactions with the Arterial Wall and Its Vascular Microenvironment and Implications for Rational Therapies. Int J Mol Sci 2022; 23:14914. [PMID: 36499242 PMCID: PMC9739112 DOI: 10.3390/ijms232314914] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/19/2022] [Accepted: 11/23/2022] [Indexed: 11/30/2022] Open
Abstract
Peripheral artery disease (PAD) is a clinical manifestation of atherosclerotic disease with a large-scale impact on the economy and global health. Despite the role played by platelets in the process of atherogenesis being well recognized, evidence has been increasing on the contribution of the coagulation system to the atherosclerosis formation and PAD development, with important repercussions for the therapeutic approach. Histopathological analysis and some clinical studies conducted on atherosclerotic plaques testify to the existence of different types of plaques. Likely, the role of coagulation in each specific type of plaque can be an important determinant in the histopathological composition of atherosclerosis and in its future stability. In this review, we analyze the molecular contribution of inflammation and the coagulation system on PAD pathogenesis, focusing on molecular similarities and differences between atherogenesis in PAD and coronary artery disease (CAD) and discussing the possible implications for current therapeutic strategies and future perspectives accounting for molecular inflammatory and coagulation targets. Understanding the role of cross-talking between coagulation and inflammation in atherosclerosis genesis and progression could help in choosing the right patients for future dual pathway inhibition strategies, where an antiplatelet agent is combined with an anticoagulant, whose role, despite pathophysiological premises and trials' results, is still under debate.
Collapse
Affiliation(s)
- Giuseppe Miceli
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Maria Grazia Basso
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Giuliana Rizzo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Chiara Pintus
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), Università degli Studi di Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
- Internal Medicine and Stroke Care Ward, University Hospital Policlinico “P. Giaccone”, 90100 Palermo, Italy
| |
Collapse
|
50
|
Reliability of Rodent and Rabbit Models in Preeclampsia Research. Int J Mol Sci 2022; 23:ijms232214344. [PMID: 36430816 PMCID: PMC9696504 DOI: 10.3390/ijms232214344] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
In vivo studies on the pathology of gestation, including preeclampsia, often use small mammals such as rabbits or rodents, i.e., mice, rats, hamsters, and guinea pigs. The key advantage of these animals is their short reproductive cycle; in addition, similar to humans, they also develop a haemochorial placenta and present a similar transformation of maternal spiral arteries. Interestingly, pregnant dams also demonstrate a similar reaction to inflammatory factors and placentally derived antiangiogenic factors, i.e., soluble fms-like tyrosine kinase 1 (sFlt-1) or soluble endoglin-1 (sEng), as preeclamptic women: all animals present an increase in blood pressure and usually proteinuria. These constitute the classical duet that allows for the recognition of preeclampsia. However, the time of initiation of maternal vessel remodelling and the depth of trophoblast invasion differs between rabbits, rodents, and humans. Unfortunately, at present, no known animal replicates a human pregnancy exactly, and hence, the use of rabbit and rodent models is restricted to the investigation of individual aspects of human gestation only. This article compares the process of placentation in rodents, rabbits, and humans, which should be considered when planning experiments on preeclampsia; these aspects might determine the success, or failure, of the study. The report also reviews the rodent and rabbit models used to investigate certain aspects of the pathomechanism of human preeclampsia, especially those related to incorrect trophoblast invasion, placental hypoxia, inflammation, or maternal endothelial dysfunction.
Collapse
|