1
|
Amor M, Diaz M, Bianco V, Svecla M, Schwarz B, Rainer S, Pirchheim A, Schooltink L, Mukherjee S, Grabner GF, Beretta G, Lamina C, Norata GD, Hackl H, Kratky D. Identification of regulatory networks and crosstalk factors in brown adipose tissue and liver of a cold-exposed cardiometabolic mouse model. Cardiovasc Diabetol 2024; 23:298. [PMID: 39143620 PMCID: PMC11325583 DOI: 10.1186/s12933-024-02397-7] [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: 04/30/2024] [Accepted: 08/07/2024] [Indexed: 08/16/2024] Open
Abstract
BACKGROUND Activation of brown adipose tissue (BAT) has gained attention due to its ability to dissipate energy and counteract cardiometabolic diseases (CMDs). METHODS This study investigated the consequences of cold exposure on the BAT and liver proteomes of an established CMD mouse model based on LDL receptor-deficient (LdlrKO) mice fed a high-fat, high-sucrose, high-cholesterol diet for 16 weeks. We analyzed energy metabolism in vivo and performed untargeted proteomics on BAT and liver of LdlrKO mice maintained at 22 °C or 5 °C for 7 days. RESULTS We identified several dysregulated pathways, miRNAs, and transcription factors in BAT and liver of cold-exposed Ldlrko mice that have not been previously described in this context. Networks of regulatory interactions based on shared downstream targets and analysis of ligand-receptor pairs identified fibrinogen alpha chain (FGA) and fibronectin 1 (FN1) as potential crosstalk factors between BAT and liver in response to cold exposure. Importantly, genetic variations in the genes encoding FGA and FN1 have been associated with cardiometabolic-related phenotypes and traits in humans. DISCUSSION This study describes the key factors, pathways, and regulatory networks involved in the crosstalk between BAT and the liver in a cold-exposed CMD mouse model. These findings may provide a basis for future studies aimed at testing whether molecular mediators, as well as regulatory and signaling mechanisms involved in tissue adaption upon cold exposure, could represent a target in cardiometabolic disorders.
Collapse
Affiliation(s)
- Melina Amor
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Malena Diaz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Valentina Bianco
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Monika Svecla
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
- Department of Neurosurgery, Charité- Universitätsmedizin Berlin, Berlin, Germany
| | - Birgit Schwarz
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Silvia Rainer
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Anita Pirchheim
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Laszlo Schooltink
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Suravi Mukherjee
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Gernot F Grabner
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria
| | - Giangiacomo Beretta
- Department of Environmental Science and Policy, Università degli Studi di Milano, Milan, Italy
| | - Claudia Lamina
- Department of Genetics and Pharmacology, Institute of Genetic Epidemiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Giuseppe Danilo Norata
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Hubert Hackl
- Institute of Bioinformatics, Medical University of Innsbruck, Innsbruck, Austria
| | - Dagmar Kratky
- Gottfried Schatz Research Center, Molecular Biology and Biochemistry, Medical University of Graz, Neue Stiftingtalstrasse 6/4, Graz, 8010, Austria.
- BioTechMed-Graz, Graz, Austria.
| |
Collapse
|
2
|
Dziedzic A, Maciak K, Miller ED, Starosta M, Saluk J. Targeting Vascular Impairment, Neuroinflammation, and Oxidative Stress Dynamics with Whole-Body Cryotherapy in Multiple Sclerosis Treatment. Int J Mol Sci 2024; 25:3858. [PMID: 38612668 PMCID: PMC11011409 DOI: 10.3390/ijms25073858] [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: 01/31/2024] [Revised: 03/05/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
Multiple sclerosis (MS), traditionally perceived as a neurodegenerative disease, exhibits significant vascular alternations, including blood-brain barrier (BBB) disruption, which may predispose patients to increased cardiovascular risks. This vascular dysfunction is intricately linked with the infiltration of immune cells into the central nervous system (CNS), which plays a significant role in perpetuating neuroinflammation. Additionally, oxidative stress serves not only as a byproduct of inflammatory processes but also as an active contributor to neural damage. The synthesis of these multifaceted aspects highlights the importance of understanding their cumulative impact on MS progression. This review reveals that the triad of vascular damage, chronic inflammation, and oxidative imbalance may be considered interdependent processes that exacerbate each other, underscoring the need for holistic and multi-targeted therapeutic approaches in MS management. There is a necessity for reevaluating MS treatment strategies to encompass these overlapping pathologies, offering insights for future research and potential therapeutic interventions. Whole-body cryotherapy (WBCT) emerges as one of the potential avenues for holistic MS management approaches which may alleviate the triad of MS progression factors in multiple ways.
Collapse
Affiliation(s)
- Angela Dziedzic
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (K.M.)
| | - Karina Maciak
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (K.M.)
| | - Elżbieta Dorota Miller
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland; (E.D.M.); (M.S.)
| | - Michał Starosta
- Department of Neurological Rehabilitation, Medical University of Lodz, Milionowa 14, 93-113 Lodz, Poland; (E.D.M.); (M.S.)
| | - Joanna Saluk
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland; (A.D.); (K.M.)
| |
Collapse
|
3
|
Teległów A, Seremak J, Golec J, Marchewka J, Golec P, Marchewka U, Maciejczyk M, Golec E. The effect of sulfur baths on hemorheological properties of blood in patients with osteoarthritis. Sci Rep 2023; 13:7960. [PMID: 37198390 DOI: 10.1038/s41598-023-35264-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 05/15/2023] [Indexed: 05/19/2023] Open
Abstract
Balneotherapy is an effective treatment method in various diseases and commonly used treatment modality among patients with musculoskeletal disorders. Sulfur baths are known for healing properties however effect on rheological properties is unstudied. Thus the aim of our study was to determine the effect of sulfur balneotherapy on hemorheological blood indices. A total of 48 patients with osteoarthritis were enrolled to the study. Blood samples were collected twice, before and after 3-week time period. We evaluated complete blood count, fibrinogen, hs-CRP and blood rheology parameters such as elongation index (EI), half-time of total aggregation (T1/2) and aggregation index (AI) analyzed with the Lorrca Maxis. Mean age of studied cohort was 67 ± 5 years. After sulfur baths WBC count was significantly decreased is studied group (p = 0.021) as well as neutrophile count (p = 0.036). Red blood cell EIs were statistically higher after sulfur baths in shear stress ranging from 8.24 to 60.30 Pa. T1/2 was significantly higher (p = 0.031) and AI lower (p = 0.003) compared to baseline. No significant changes in fibrinogen and hs-CRP were observed. It is the first study that evaluate effect of sulfur balneotherapy on rheologic properties of blood. Sulfur water baths may improve erythrocyte deformability and aggregation parameters.
Collapse
Affiliation(s)
- Aneta Teległów
- Department of Health Promotion, Institute of Basic Sciences, University of Physical Education in Krakow, 31-571, Kraków, Poland.
| | | | - Joanna Golec
- Institute of Clinical Rehabilitation, University of Physical Education in Krakow, 31-571, Kraków, Poland
| | - Jakub Marchewka
- Institute of Clinical Rehabilitation, University of Physical Education in Krakow, 31-571, Kraków, Poland
- 5th Military Clinical Hospital, 30-901, Kraków, Poland
| | - Piotr Golec
- Individual Healthcare Centre, Kraków, Poland
| | | | - Marcin Maciejczyk
- Institute of Biomedical Sciences, University of Physical Education in Krakow, 31-571, Kraków, Poland
| | - Edward Golec
- Institute of Clinical Rehabilitation, University of Physical Education in Krakow, 31-571, Kraków, Poland
| |
Collapse
|
4
|
Sulimai N, Brown J, Lominadze D. Vascular Effects on Cerebrovascular Permeability and Neurodegeneration. Biomolecules 2023; 13:biom13040648. [PMID: 37189395 DOI: 10.3390/biom13040648] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 03/29/2023] [Accepted: 04/02/2023] [Indexed: 05/17/2023] Open
Abstract
Neurons and glial cells in the brain are protected by the blood brain barrier (BBB). The local regulation of blood flow is determined by neurons and signal conducting cells called astrocytes. Although alterations in neurons and glial cells affect the function of neurons, the majority of effects are coming from other cells and organs of the body. Although it seems obvious that effects beginning in brain vasculature would play an important role in the development of various neuroinflammatory and neurodegenerative pathologies, significant interest has only been directed to the possible mechanisms involved in the development of vascular cognitive impairment and dementia (VCID) for the last decade. Presently, the National Institute of Neurological Disorders and Stroke applies considerable attention toward research related to VCID and vascular impairments during Alzheimer's disease. Thus, any changes in cerebral vessels, such as in blood flow, thrombogenesis, permeability, or others, which affect the proper vasculo-neuronal connection and interaction and result in neuronal degeneration that leads to memory decline should be considered as a subject of investigation under the VCID category. Out of several vascular effects that can trigger neurodegeneration, changes in cerebrovascular permeability seem to result in the most devastating effects. The present review emphasizes the importance of changes in the BBB and possible mechanisms primarily involving fibrinogen in the development and/or progression of neuroinflammatory and neurodegenerative diseases resulting in memory decline.
Collapse
Affiliation(s)
- Nurul Sulimai
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| | - Jason Brown
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| | - David Lominadze
- Department of Surgery, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
- Department of Molecular Pharmacology and Physiology, College of Medicine, University of South Florida Morsani, Tampa, FL 33612, USA
| |
Collapse
|
5
|
Stohnii Y, Yatsenko T, Nikulina V, Kucheriavyi Y, Hrabovskyi O, Slominskyi O, Savchenko K, Garmanchuk L, Varbanets L, Tykhomyrov A, Chernyshenko V. Functional properties of individual sub-domains of the fibrin(ogen) αC-domains. BBA ADVANCES 2023; 3:100072. [PMID: 37082262 PMCID: PMC10074951 DOI: 10.1016/j.bbadva.2023.100072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/29/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Background Fibrinogen is a large polyfunctional plasma protein consisting of a number of structural and functional domains. Among them, two αC-domains, each formed by the amino acid residues Аα392-610, are involved in fibrin polymerization, activation of fibrinolysis, platelet aggregation, and interaction with different cell types. Previous study revealed that each fibrinogen αC-domain consists of the N-terminal and C-terminal sub-domains. The major objections of the present study were to test functional role of these sub-domains in the above mentioned processes. Methods To achieve these objections, we used specific proteases to prepare two truncated forms of fibrinogen, fibrinogen desAα505-610 and fibrinogen desAα414-610, missing their N-terminal and both N- and C-terminal sub-domains, respectively. Results Our study with these truncated forms using turbidity measurements and electron microscopy revealed that the N- and C-terminal subdomains both contribute to protofibril formation and their lateral aggregation into fibers during fibrin polymerization process. These two sub-domains also contributed to platelet aggregation with the N-terminal sub-domains playing a more significant role in this process. At the same time, the C-terminal sub-domains make the major contribution to the plasminogen activation process. Further, our experiments revealed that the C-terminal sub-domains are involved in endothelial cell viability and migration of cancer cells. Conclusions Thus, the results obtained establish the functional role of individual sub-domains of the αC-domains in fibrin polymerization, activation of fibrinolytic system, platelet aggregation, and cellular interactions. General significance The present study expands our understanding of the functional role of individual fibrinogen domains and their specific portions in various fibrin(ogen)-dependent processes.
Collapse
|
6
|
Piñeiro-Llanes J, Suzuki-Hatano S, Jain A, Pérez Medina VA, Cade WT, Pacak CA, Simmons CS. Matrix produced by diseased cardiac fibroblasts affects early myotube formation and function. Acta Biomater 2022; 152:100-112. [PMID: 36055608 PMCID: PMC10625442 DOI: 10.1016/j.actbio.2022.08.060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Revised: 08/25/2022] [Accepted: 08/25/2022] [Indexed: 11/28/2022]
Abstract
The extracellular matrix (ECM) provides both physical and chemical cues that dictate cell function and contribute to muscle maintenance. Muscle cells require efficient mitochondria to satisfy their high energy demand, however, the role the ECM plays in moderating mitochondrial function is not clear. We hypothesized that the ECM produced by stromal cells with mitochondrial dysfunction (Barth syndrome, BTHS) provides cues that contribute to metabolic dysfunction independent of muscle cell health. To test this, we harnessed the ECM production capabilities of human pluripotent stem-cell-derived cardiac fibroblasts (hPSC-CFs) from healthy and BTHS patients to fabricate cell-derived matrices (CDMs) with controlled topography, though we found that matrix composition from healthy versus diseased cells influenced myotube formation independent of alignment cues. To further investigate the effects of matrix composition, we then examined the influence of healthy- and BTHS-derived CDMs on myotube formation and metabolic function. We found that BTHS CDMs induced lower fusion index, lower ATP production, lower mitochondrial membrane potential, and higher ROS generation than the healthy CDMs. These findings imply that BTHS-derived ECM alone contributes to myocyte dysfunction in otherwise healthy cells. Finally, to investigate potential mechanisms, we defined the composition of CDMs produced by hPSC-CFs from healthy and BTHS patients using mass spectrometry and identified 15 ECM and related proteins that were differentially expressed in the BTHS-CDM compared to healthy CDM. Our results highlight that ECM composition affects skeletal muscle formation and metabolic efficiency in otherwise healthy cells, and our methods to generate patient-specific CDMs are a useful tool to investigate the influence of the ECM on disease progression and to investigate variability among diseased patients. STATEMENT OF SIGNIFICANCE: Muscle function requires both efficient metabolism to generate force and structured extracellular matrix (ECM) to transmit force, and we sought to examine the interactions between metabolism and ECM when metabolic disease is present. We fabricated patient-specific cell derived matrices (CDMs) with controlled topographic features to replicate the composition of healthy and mitochondrial-diseased (Barth syndrome) ECM. We found that disease-derived ECM negatively affects metabolic function of otherwise healthy myoblasts, and we identified several proteins in disease-derived ECM that may be mediating this dysfunction. We anticipate that our patient-specific CDM system could be fabricated with other topographies and cell types to study cell functions and diseases of interest beyond mitochondrial dysfunction and, eventually, be applied toward personalized medicine.
Collapse
Affiliation(s)
- Janny Piñeiro-Llanes
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Silveli Suzuki-Hatano
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA
| | - Ananya Jain
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Valerie A Pérez Medina
- Department of Mechanical Engineering, University of Puerto Rico, Mayaguez 00682, Puerto Rico
| | - William Todd Cade
- Physical Therapy Division, Duke University, 311 Trent Drive, Durham, NC 27710, USA
| | - Christina A Pacak
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL 32610, USA; Neurology Department, Medical School, University of Minnesota, WMBB 4-188 2101 6th Street SE, Minneapolis 55455, USA
| | - Chelsey S Simmons
- J. Crayton Pruitt Family Department of Biomedical Engineering, Herbert Wertheim College of Engineering, University of Florida, Gainesville, FL 32611, USA; Department of Mechanical and Aerospace Engineering Herbert Wertheim College of Engineering, University of Florida.
| |
Collapse
|
7
|
Lin G, Hu M, Song J, Xu X, Liu H, Qiu L, Zhu H, Xu M, Geng D, Yang L, Huang G, He J, Wang Z. High Fibrinogen to Albumin Ratio: A Novel Marker for Risk of Stroke-Associated Pneumonia? Front Neurol 2022; 12:747118. [PMID: 35095715 PMCID: PMC8792987 DOI: 10.3389/fneur.2021.747118] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 12/14/2021] [Indexed: 11/13/2022] Open
Abstract
Background: Stroke-associated pneumonia (SAP) is associated with poor prognosis after acute ischemic stroke (AIS). Purpose: This study aimed to describe the parameters of coagulation function and evaluate the association between the fibrinogen-to-albumin ratio (FAR) and SAP in patients with AIS. Patients and methods: A total of 932 consecutive patients with AIS were included. Coagulation parameters were measured at admission. All patients were classified into two groups according to the optimal cutoff FAR point at which the sum of the specificity and sensitivity was highest. Propensity score matching (PSM) was performed to balance potential confounding factors. Univariate and multivariate logistic regression analyses were applied to identify predictors of SAP. Results: A total of 100 (10.7%) patients were diagnosed with SAP. The data showed that fibrinogen, FAR, and D-dimer, prothrombin time (PT), activated partial thromboplastin time (aPTT) were higher in patients with SAP, while albumin was much lower. Patients with SAP showed a significantly increased FAR when compared with non-SAP (P < 0.001). Patients were assigned to groups of high FAR (≥0.0977) and low FAR (<0.0977) based on the optimal cut-off value. Propensity score matching analysis further confirmed the association between FAR and SAP. After adjusting for confounding and risk factors, multivariate regression analysis showed that the high FAR (≥0.0977) was an independent variable predicting the occurrence of SAP (odds ratio =2.830, 95% CI = 1.654–4.840, P < 0.001). In addition, the FAR was higher in the severe pneumonia group when it was assessed by pneumonia severity index (P = 0.008). Conclusions: High FAR is an independent potential risk factor of SAP, which can help clinicians identify high-risk patients with SAP after AIS.
Collapse
Affiliation(s)
- Gangqiang Lin
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minlei Hu
- Department of Neurology, The First Hospital of Jiaxing, Jiaxing, China
| | - Jiaying Song
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Xueqian Xu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Haiwei Liu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Linan Qiu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hanyu Zhu
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Minjie Xu
- School of Mental Health, Wenzhou Medical University, Wenzhou, China
| | - Dandan Geng
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lexuan Yang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Guiqian Huang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Guiqian Huang
| | - Jincai He
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Jincai He
| | - Zhen Wang
- Department of Neurology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Zhen Wang
| |
Collapse
|
8
|
Teległów A, Romanovski V, Skowron B, Mucha D, Tota Ł, Rosińczuk J, Mucha D. The Effect of Extreme Cold on Complete Blood Count and Biochemical Indicators: A Case Study. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 19:424. [PMID: 35010684 PMCID: PMC8744862 DOI: 10.3390/ijerph19010424] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 11/17/2022]
Abstract
Regular exposure to a cold factor-cold water swimming or ice swimming and cold air-results in an increased tolerance to cold due to numerous adaptive mechanisms in humans. Due to the lack of scientific reports on the effects of extremely low outdoor temperatures on the functioning of the human circulatory system, the aim of this study was to evaluate complete blood count and biochemical blood indices in multiple Guinness world record holder Valerjan Romanovski, who was exposed to extremely cold environment from -5 °C to -37 °C for 50 days in Rovaniemi (a city in northern Finland). Valerjan Romanovski proved that humans can function in extremely cold temperatures. Blood from the subject was collected before and after the expedition. The subject was found to have abnormalities for the following blood indices: testosterone increases by 60.14%, RBC decreases by 4.01%, HGB decreases by 3.47%, WBC decreases by 21.53%, neutrocytes decrease by 17.31%, PDW increases by 5.31%, AspAT increases by 52.81%, AlAT increase by 68.75%, CK increases by 8.61%, total cholesterol decreases by 5.88%, HDL increases by 28.18%. Percentage changes in other complete blood count and biochemical indices were within standard limits. Long-term exposure of the subject (50 days) to extreme cold stress had no noticeable negative effect on daily functioning.
Collapse
Affiliation(s)
- Aneta Teległów
- Department of Rehabilitation in Internal Diseases, Institute of Clinical Rehabilitation, Faculty of Motor Rehabilitation, University of Physical Education, 31-571 Krakow, Poland;
| | - Valerjan Romanovski
- Non-Governmental Organization and Association Oswajamy Żywioły, 25-607 Kielce, Poland;
| | - Beata Skowron
- Medical Department Diagnostyka S.A., 31-864 Krakow, Poland;
| | - Dawid Mucha
- Institute of Health Sciences, Podhale State College of Applied Science in Nowy Targ, 34-400 Nowy Targ, Poland;
| | - Łukasz Tota
- Department of Physiology and Biochemistry, Institute of Biomedical Sciences, Faculty of Physical Education and Sport, University of Physical Education, 31-571 Krakow, Poland;
| | - Joanna Rosińczuk
- Department of Nursing and Obstetrics, Division of Internal Medicine Nursing, Faculty of Health Sciences, Wroclaw Medical University, 51-618 Wroclaw, Poland
| | - Dariusz Mucha
- Department of Biological Regeneration and Correction of Posture Defects, Institute of Biomedical Sciences, Faculty of Physical Education and Sport, University of Physical Education, 31-571 Krakow, Poland;
| |
Collapse
|
9
|
Teległów A, Marchewka J, Ptaszek B, Mardyła M, Mazur-Kurach P, Rychlik U, Wójcik E, Golec J, Dąbrowski Z, Marchewka A. Assessment of morphological, biochemical and rheological blood indicators in men after a 24-hour stay in a thermo-climatic chamber -50°C. REHABILITACJA MEDYCZNA 2021. [DOI: 10.5604/01.3001.0015.5114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Purpose: The aim of the study was to evaluate morphological, biochemical, and rheological blood indicators in men staying in a cryochamber at a temperature of -50°C for 24 hours. In 2018, a scientific-survival project ‘Taming the Frost’ was conducted at the Technoclimatic Research and Working Machines Laboratory of the Cracow University of Technology, under the scientific patronage of the Rector of the Cracow University of Technology, Prof. Jan Kazior, PhD, as well as the Rector of the University of Physical Education in Krakow, the late Prof. Aleksander Tyka, PhD, and the Vice-Rector for Science, Prof. Anna Marchewka, PhD.
Material and methods: The blood for the tests was collected from an ulnar vein in fasting participants by a qualified nurse, in the morning, before entering the cryochamber and after 24 hours, i.e. on leaving the cryochamber. Morphological, biochemical, and rheological blood indicators were evaluated. The study group of the scientific-survival project ‘Taming the Frost’ involved men (n=6) who stayed in a cryochamber at a temperature of -50°C for 24 hours. For each participant, a 5-ml blood sample was placed in a tube (BD Vacutainer) with EDTA K2 anticoagulant for blood morphology and blood rheological evaluations in the Blood Physiology Laboratory of the Central Research and Development Laboratory, University of Physical Education in Krakow. Another 5-ml blood sample was placed in a Vacuette tube with a clotting activator for the remaining biochemical analyses in the Department of Clinical Biochemistry of the Krakow Branch of Maria Skłodowska-Curie National Research Institute of Oncology.
Results: After leaving the cryochamber with a temperature of –50°C, the participants presented statistically significant increases in monocyte count and high-density lipoprotein and creatine kinase values, as well as decreases in IgA, total cholesterol, and triglycerides. In the assessment of blood rheological indicators, statistically significant increases in the elongation index at the shear stress of 0.30, 0.58, 1.13, and 2.19 Pa and decreases in the elongation index at the shear stress of 31.03 and 60.3 Pa were observed. For red cell aggregation indicators, a statistically significant increase in total aggregation time was reported. The other indicators exhibited a significance level of p>0.05.
Conclusions: Staying in a cryochamber at a temperature of -50°C for 24 hours did not exert a negative impact on morphological, biochemical, or rheological blood indicators, which implies the subjects’ adaptation to the arranged conditions.
Collapse
Affiliation(s)
- Aneta Teległów
- Institute of Basic Sciences, University of Physical Education in Krakow, Krakow, Poland / Zakład Promocji Zdrowia, Instytut Nauk Podstawowych, Wydział Rehabilitacji Ruchowej, Akademia Wychowania Fizycznego w Krakowie
| | - Jakub Marchewka
- Institute of Clinical Rehabilitation, University of Physical Education in Krakow, Krakow, Poland / Instytut Rehabilitacji Klinicznej, Wydział Rehabilitacji Ruchowej, Akademia Wychowania Fizycznego w Krakowie
| | - Bartłomiej Ptaszek
- Institute of Applied Sciences, University of Physical Education in Krakow, Krakow, Poland / Instytut Nauk Stosowanych, Akademia Wychowania Fizycznego w Krakowie
| | - Mateusz Mardyła
- Institute of Biomedical Sciences, University of Physical Education in Krakow, Poland / Instytut Nauk Biomedycznych, Akademia Wychowania Fizycznego w Krakowie
| | - Paulina Mazur-Kurach
- Institute of Biomedical Sciences, University of Physical Education in Krakow, Poland / Instytut Nauk Biomedycznych, Akademia Wychowania Fizycznego w Krakowie
| | - Urszula Rychlik
- Maria Sklodowska-Curie National Research Institute of Oncology in Krakow, Poland / Narodowy Instytut Onkologii im. M. Skłodowskiej-Curie, Państwowy Instytut Badawczy, Oddział w Krakowie
| | - Ewa Wójcik
- Retired employee Maria Sklodowska-Curie National Research Institute of Oncology in Krakow, Poland / Emerytowany pracownik Narodowego Instytutu Onkologii im. M. Skłodowskiej-Curie, Państwowy Instytut Badawczy, Oddział w Krakowie
| | - Joanna Golec
- Institute of Clinical Rehabilitation, University of Physical Education in Krakow, Krakow, Poland / Instytut Rehabilitacji Klinicznej, Wydział Rehabilitacji Ruchowej, Akademia Wychowania Fizycznego w Krakowie
| | - Zbigniew Dąbrowski
- Retired employee University of Physical Education in Krakow, Poland / Emerytowany pracownik Akademii Wychowania Fizycznego w Krakowie
| | - Anna Marchewka
- Institute of Clinical Rehabilitation, University of Physical Education in Krakow, Krakow, Poland / Instytut Rehabilitacji Klinicznej, Wydział Rehabilitacji Ruchowej, Akademia Wychowania Fizycznego w Krakowie
| |
Collapse
|
10
|
Effect of Whole-Body Cryotherapy on Morphological, Rheological and Biochemical Indices of Blood in People with Multiple Sclerosis. J Clin Med 2021; 10:jcm10132833. [PMID: 34198961 PMCID: PMC8268166 DOI: 10.3390/jcm10132833] [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: 05/22/2021] [Revised: 06/19/2021] [Accepted: 06/21/2021] [Indexed: 11/17/2022] Open
Abstract
The aim of this study was to examine and assess the impact of a series of 20 whole-body cryotherapy (WBC) treatments on the biochemical and rheological indices of blood in people with multiple sclerosis. In this prospective controlled study, the experimental group consisted of 15 women aged 34-55 (mean age, 41.53 ± 6.98 years) with diagnosed multiple sclerosis who underwent a series of whole-body cryotherapy treatments. The first control group consisted of 20 women with diagnosed multiple sclerosis. This group had no intervention in the form of whole-body cryotherapy. The second control group consisted of 15 women aged 30-49 years (mean age, 38.47 ± 6.0 years) without neurological diseases and other chronic diseases who also underwent the whole-body cryotherapy treatment. For the analysis of the blood indices, venous blood was taken twice (first, on the day of initiation of whole-body cryotherapy treatments and, second, after a series of 20 cryotherapy treatments). The blood counts were determined using an ABX MICROS 60 hematological analyzer (USA). The LORCA analyzer (Laser-Optical Rotational Cell Analyzer, RR Mechatronics, the Netherlands) was used to study the aggregation and deformability of erythrocytes. The total protein serum measurement was performed using a Cobas 6000 analyzer, Roche and a Proteinogram-Minicap Sebia analyzer. Fibrinogen determinations were made using a Bio-Ksel, Chrom-7 camera. Statistically significant differences and changes after WBC in the levels of red blood cells (RBC), hemoglobin (HGB), hematocrit (HCT), elongation index, total extend of aggregation (AMP), and proteins (including fibrinogen) were observed. However, there was no significant effect of a series of 20 WBC treatments on changes in blood counts, rheology, and biochemistry in women with multiple sclerosis. Our results show that the use of WBC has a positive effect on the rheological properties of the blood of healthy women.
Collapse
|
11
|
Szczurek W, Gąsior M, Skrzypek M, Romuk E, Szyguła-Jurkiewicz B. Factors Associated with Ineffectiveness of Sildenafil Treatment in Patients with End-Stage Heart Failure and Elevated Pulmonary Vascular Resistance. J Clin Med 2020; 9:jcm9113539. [PMID: 33147835 PMCID: PMC7692635 DOI: 10.3390/jcm9113539] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/29/2020] [Accepted: 10/30/2020] [Indexed: 01/12/2023] Open
Abstract
Introduction: Elevated pulmonary vascular resistance (PVR) unresponsive to vasodilator treatment is a marker of heart failure (HF) severity, and an important predictor of poor results of heart transplantation (HT). Objective: We sought to analyze factors associated with ineffectiveness of sildenafil treatment in end-stage HF patients with elevated PVR with particular emphasis placed on tenascin-C (TNC) serum concentrations. Patients and Methods: The study is an analysis of 132 end-stage HF patients referred for HT evaluation in the Cardiology Department between 2015 and 2018. TNC was measured by sandwich enzyme-linked immunosorbent assay (Human TNC, SunRedBio Technology, Shanghai, China). The endpoint was PVR > 3 Wood units after the six-month sildenafil therapy. Results: The median age was 58 years, and 90.2% were men. PVR >3 Wood units after 6 months of sildenafil treatment were found in 36.6% patients. The multivariable logistic regression analysis confirmed that TNC (OR = 1.004 (1.002–1.006), p = 0.0003), fibrinogen (OR= 1.019 (1.005–1.033), p = 0.085), creatinine (OR =1.025 (1.004–1.047), p = 0.0223) and right ventricular end-diastolic dimension (RVEDd) (OR = 1.279 (1.074–1.525), p = 0.0059) were independently associated with resistance to sildenafil treatment. Area under the ROC curves indicated an acceptable power of TNC (0.9680 (0.9444–0.9916)), fibrinogen (0.8187 (0.7456–0.8917)) and RVEDd (0.7577 (0.6723–0.8431)), as well as poor strength of creatinine (0.6025 (0.4981–0.7070)) for ineffectiveness of sildenafil treatment. Conclusions: Higher concentrations of TNC, fibrinogen and creatinine, as well as a larger RVEDd are independently associated with the ineffectiveness of sildenafil treatment. TNC has the strongest predictive power, sensitivity and specificity for evaluation of resistance to sildenafil treatment.
Collapse
Affiliation(s)
- Wioletta Szczurek
- Silesian Center for Heart Diseases in Zabrze, 41-800 Zabrze, Poland
- Correspondence: ; Tel.: +48-694-138-970 or +48-323-733-860
| | - Mariusz Gąsior
- 3rd Department of Cardiology, School of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (M.G.); (B.S.-J.)
| | - Michał Skrzypek
- Department of Biostatistics, School of Public Health in Bytom, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Ewa Romuk
- Department of Biochemistry, School of Medicine with the Division of Dentistry, Medical University of Silesia, 41-800 Zabrze, Poland;
| | - Bożena Szyguła-Jurkiewicz
- 3rd Department of Cardiology, School of Medical Sciences in Zabrze, Medical University of Silesia, 40-055 Katowice, Poland; (M.G.); (B.S.-J.)
| |
Collapse
|
12
|
Zadeh FJ, Mohammadtaghizadeh M, Bahadori H, Saki N, Rezaeeyan H. The role of exogenous Fibrinogen in cardiac surgery: stop bleeding or induce cardiovascular disease. Mol Biol Rep 2020; 47:8189-8198. [PMID: 33026614 DOI: 10.1007/s11033-020-05880-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
The surgical treatment contributes to broad variety of cardiovascular diseases (CVD). Due to many involved factors in preoperative bleeding, it is almost difficult to perform better Haemostatic approach. Fibrinogen is a major blood glycoprotein and a coagulation factor which decreases postoperative bleeding. It has a potential role in platelet activation and bleeding inhibition; it may reflect the inflammatory responses and be related to the endothelial dysfunction. Fibrinogen can act as a pro-inflammatory element via increasing some inflammatory markers including IL-6, tumor necrosis factor-α (TNF-α), monocyte chemo attractant protein (MCP-1), macrophage inflammatory protein-1 (MIP-1a and b), matrix metalloproteinase (MMP-1 and MMP-9) and Toll-like Receptors (TLRs); through activation of these factors, fibrinogen may induce some inflammatory mechanisms such as focal adhesion kinase (FAK), mitogen-activated protein kinases (MAPK) and nuclear factor κB (NF-κB) pathways. It may cause endothelial dysfunction by increasing P and E-selection, intercellular adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule-1 (VCAM-1) levels which activate MAPK and NF-κB pathways. This factor is also associated with increased exocytosed von Willebrand factor (vWF) as well as activation of Rho-GTPase mechanism. All of these data demonstrate the dual role of fibrinogen in cardiac surgeries, bleeding inhibition and CVD. Therefore, identifying the CVD factors is helpful for designing preventive strategies and alternative drugs.
Collapse
Affiliation(s)
- Fatemeh Javaherforoosh Zadeh
- Department of Cardiac Anesthesia, Ahvaz Anesthesiology and Pain Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Hojatolah Bahadori
- Department of Cardiac Anesthesia, Ahvaz Anesthesiology and Pain Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hadi Rezaeeyan
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. .,High Institute for Education and Research in Transfusion Medicine, Tehran, Iran.
| |
Collapse
|
13
|
Sulimai N, Lominadze D. Fibrinogen and Neuroinflammation During Traumatic Brain Injury. Mol Neurobiol 2020; 57:4692-4703. [PMID: 32776201 DOI: 10.1007/s12035-020-02012-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 07/08/2020] [Indexed: 12/14/2022]
Abstract
Many neurodegenerative diseases such as Alzheimer's disease (AD), multiple sclerosis, and traumatic brain injury (TBI) are associated with systemic inflammation. Inflammation itself results in increased blood content of fibrinogen (Fg), called hyperfibrinogenemia (HFg). Fg is not only considered an acute phase protein and a marker of inflammation, but has been shown that it can cause inflammatory responses. Fibrin deposits have been associated with memory reduction in neuroinflammatory diseases such as AD and TBI. Reduction in short-term memory has been seen during the most common form of TBI, mild-to-moderate TBI. Fibrin deposits have been found in brains of patients with mild-to-moderate TBI. The vast majority of the literature emphasizes the role of fibrin-activated microglia as the mediator in the neuroinflammation pathway. However, the recent discovery that astrocytes, which constitute approximately 30% of the cells in the mammalian central nervous system, manifest different reactive states warrants further investigations in the causative role of HFg in astrocyte-mediated neuroinflammation. Our previous study showed that Fg deposited in the vasculo-astrocyte interface-activated astrocytes. However, little is known of how Fg directly affects astrocytes and neurons. In this review, we summarize studies that show the effect of Fg on different types of cells in the vasculo-neuronal unit. We will also discuss the possible mechanism of HFg-induced neuroinflammation during TBI.
Collapse
Affiliation(s)
- Nurul Sulimai
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA
| | - David Lominadze
- Departments of Surgery, University of South Florida Morsani College of Medicine, MDC-4024, 12901 Bruce B. Downs Blvd, Tampa, FL, 33612, USA.
- Molecular Pharmacology and Physiology, University of South Florida Morsani College of Medicine, Tampa, FL, 33612, USA.
| |
Collapse
|
14
|
The human liver matrisome - Proteomic analysis of native and fibrotic human liver extracellular matrices for organ engineering approaches. Biomaterials 2020; 257:120247. [PMID: 32739662 DOI: 10.1016/j.biomaterials.2020.120247] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Revised: 07/05/2020] [Accepted: 07/16/2020] [Indexed: 02/06/2023]
Abstract
The production of biomaterials that endow significant morphogenic and microenvironmental cues for the constitution of cell integration and regeneration remains a key challenge in the successful implementation of functional organ replacements. Despite the vast development in the production of biological and architecturally native matrices, the complex compositions and pivotal figures by which the human matrisome mediates many of its essential functions are yet to be defined. Here we present a thorough analysis of the native human liver proteomic landscape using decellularization and defatting protocols to create extracellular matrix scaffolds of natural origin that can further be used in both bottom-up and top-down approaches in tissue engineering based organ replacements. Furthermore, by analyzing human liver extracellular matrices in different stages of fibrosis and cirrhosis, we have identified distinct attributes of these tissues that could potentially be exploited therapeutically and thus require further investigation. The general experimental pipeline presented in this study is applicable to any type of tissue and can be widely used for different approaches in regenerative medicine and in the construction of novel biomaterials for organ engineering approaches.
Collapse
|
15
|
Charkviani M, Muradashvili N, Lominadze D. Vascular and non-vascular contributors to memory reduction during traumatic brain injury. Eur J Neurosci 2019; 50:2860-2876. [PMID: 30793398 PMCID: PMC6703968 DOI: 10.1111/ejn.14390] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Revised: 02/06/2019] [Accepted: 02/07/2019] [Indexed: 01/09/2023]
Abstract
Traumatic brain injury (TBI) is an increasing health problem. It is a complex, progressive disease that consists of many factors affecting memory. Studies have shown that increased blood-brain barrier (BBB) permeability initiates pathological changes in neuro-vascular network but the role of cerebrovascular dysfunction and its mediated mechanisms associated with memory reduction during TBI are still not well understood. Changes in BBB, inflammation, extravasation of blood plasma components, activation of neuroglia lead to neurodegeneration. Extravasated proteins such as amyloid-beta, fibrinogen, and cellular prion protein may form degradation resistant complexes that can lead to neuronal dysfunction and degeneration. They also have the ability to activate astrocytes, and thus, can be involved in memory impairment. Understanding the triggering mechanisms and the places they originate in vasculature or in extravascular tissue may help to identify potential therapeutic targets to ameliorate memory reduction during TBI. The goal of this review is to discuss conceptual mechanisms that lead to short-term memory reduction during non-severe TBI considering distinction between vascular and non-vascular effects on neurons. Some aspects of these mechanisms need to be confirmed further. Therefore, we hope that the discussion presented bellow may lead to experiments that may clarify the triggering mechanisms of memory reduction after head trauma.
Collapse
Affiliation(s)
- Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
- Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, KY, USA
| |
Collapse
|
16
|
Li T, Wang F, Peng R, Pei S, Hou Z, Lu B, Cong X, Chen X. Sex-related differences in the association between plasma fibrinogen and non-calcified or mixed coronary atherosclerotic plaques. Biol Sex Differ 2018; 9:51. [PMID: 30518417 PMCID: PMC6282270 DOI: 10.1186/s13293-018-0210-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 11/22/2018] [Indexed: 12/19/2022] Open
Abstract
Background Plasma fibrinogen (FIB) has been demonstrated to be a risk factor for cardiovascular disease. Patients with non-calcified plaque (NCP) or mix plaque (MP) have a higher risk of poor outcomes. However, the association between FIB and the presence of NCP or MP (NCP/MP) remains unclear, and if present, whether sex has any impact on this association remains unknown. The aim of this study was to investigate the role of FIB in predicting the presence of NCP/MP and evaluate whether sex has any impact on this association. Methods A total of 329 subjects were recruited, and the clinical and laboratory data were collected. Plasma FIB was detected by enzyme-linked immunosorbent assay. According to whether they had coronary atherosclerotic plaques and the characteristics of the most stenotic plaque, we divided them into three groups: no plaque (NP), calcified plaque (CP), and NCP/MP. Results Patients with NCP/MP had significantly higher FIB level in females, but not in males. Multiple logistic regression analysis showed that FIB was an independent risk factor for the presence of NCP/MP (odds ratio [OR] = 3.677, 95% CI 1.539–8.785, P = 0.003) in females. Receiver operating characteristic (ROC) curve analysis showed that the optimal cut-off value FIB for predicting the presence of NCP/MP was 3.41 g/L (area under curve [AUC] = 0.73, 95% CI 0.63–0.82, P < 0.001) in females. Conclusions FIB is independently associated with the presence of NCP/MP in females, but not in males. These results suggest that the potential significance of FIB-lowering regimens in females with NCP/MP.
Collapse
Affiliation(s)
- Tiewei Li
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Fang Wang
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China.
| | - Rui Peng
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Shengqiang Pei
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Zhihui Hou
- Department of Radiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Bin Lu
- Department of Radiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Xiangfeng Cong
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China
| | - Xi Chen
- Department of Clinical Laboratory Center, State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, 167 Beilishi Street, Xi-Cheng District, Beijing, 100037, China.
| |
Collapse
|
17
|
Wang H, Zheng C, Xu X, Zhao Y, Lu Y, Liu Z. Fibrinogen links podocyte injury with Toll-like receptor 4 and is associated with disease activity in FSGS patients. Nephrology (Carlton) 2018; 23:418-429. [PMID: 28407405 DOI: 10.1111/nep.13046] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 02/24/2017] [Accepted: 03/15/2017] [Indexed: 12/19/2022]
Abstract
AIM Fibrinogen (Fg) is reported to participate in inflammation through Toll-like receptor 4 (TLR4). However, it remains unknown whether Fg might induce podocyte damage through TLR4 and be related to disease activity in patients with focal segmental glomerulosclerosis (FSGS). METHODS We observed Fg-induced alterations in actin and apoptosis in cultured human podocytes transfected with or without TLR4 siRNA. Expression of TLR4, phospho-p38 MAPK and phospho-NF-κB p65 was evaluated by quantitative reverse transcription polymerase chain reaction (qRT-PCR) or western blotting, and we analysed urinary Fg levels in adriamycin-treated mice and double immunofluorescence staining for TLR4, Fg and podocin. Urinary Fg changes were also analyzed in FSGS patients under prednisone treatment. RESULTS First, Fg dose-dependently induced actin damage and apoptosis in cultured human podocytes, with an Fg-induced increase in TLR4 expression, and TLR4 siRNA transfection prevented these effects. TLR4 knockdown inhibited activation of p38 MAPK and NF-κB p65 in podocytes. Elevated urinary Fg levels were positively correlated with albuminuria in adriamycin-treated mice, in which Fg and TLR4 colocalized and exhibited increased expression in podocytes. Additionally, elevated urinary Fg levels were positively correlated with 24-h proteinuria and foot process width in FSGS patients. Urinary Fg levels were significantly decreased in patients with complete remission but not in those without remission. CONCLUSIONS Fg induced podocytes injury via the TLR4-p38 MAPK-NF-κB p65 pathway. In FSGS patients, urinary Fg levels reflect therapeutic response to prednisone and disease activity.
Collapse
Affiliation(s)
- Hongtian Wang
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Chunxia Zheng
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Xiaodong Xu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yue Zhao
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Yinghui Lu
- National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| | - Zhihong Liu
- Division of Nephrology, Jinling Hospital, Southern Medical University, Nanjing, China.,National Clinical Research Center of Kidney Diseases, Jinling Hospital, Nanjing University School of Medicine, Nanjing, China
| |
Collapse
|
18
|
Clark VD, Layson A, Charkviani M, Muradashvili N, Lominadze D. Hyperfibrinogenemia-mediated astrocyte activation. Brain Res 2018; 1699:158-165. [PMID: 30153459 DOI: 10.1016/j.brainres.2018.08.023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 08/10/2018] [Accepted: 08/22/2018] [Indexed: 12/26/2022]
Abstract
Fibrinogen (Fg)-containing plaques are associated with memory loss during various inflammatory neurodegenerative diseases such as Alzheimer's disease, multiple sclerosis, stroke, and traumatic brain injury. However, mechanisms of its action in neurovascular unit are not clear. As Fg is a high molecular weight blood protein and cannot translocate far from the vessel after extravasation, we hypothesized that it may interact with astrocytes first causing their activation. Cultured mouse cortical astrocytes were treated with Fg in the presence or absence of function-blocking anti-mouse intercellular adhesion molecule 1 (ICAM-1) antibody, or with medium alone (control). Expressions of ICAM-1 and tyrosine receptor kinase B (TrkB) as markers of astrocyte activation, and phosphorylation of TrkB (pTrkB) were assessed. Fg dose-dependently increased activation of astrocytes defined by their shape change, retraction of processes, and enhanced expressions of ICAM-1 and TrkB, and increased pTrkB. Blocking of ICAM-1 function ameliorated these Fg effects. Data suggest that Fg interacts with astrocytes causing overexpression of ICAM-1 and TrkB, and TrkB phosphorylation, and thus, astrocyte activation. Since TrkB is known to be involved in neurodegeneration, interaction of Fg with astrocytes and the resultant activation of TrkB can be a possible mechanism involved in memory reduction, which were observed in previous studies and were associated with formation of complexes of Fg deposited in extravascular space with proteins such as Amyloid beta or prion, the proteins involved in development of dementia.
Collapse
Affiliation(s)
- Vincent D Clark
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Ailey Layson
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Mariam Charkviani
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA; Department of Basic Medicine, Caucasus International University, Tbilisi, Georgia
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA; Kentucky Spinal Cord Research Center, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
19
|
Nath KA, Belcher JD, Nath MC, Grande JP, Croatt AJ, Ackerman AW, Katusic ZS, Vercellotti GM. Role of TLR4 signaling in the nephrotoxicity of heme and heme proteins. Am J Physiol Renal Physiol 2018; 314:F906-F914. [PMID: 28978536 PMCID: PMC6031913 DOI: 10.1152/ajprenal.00432.2017] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 09/29/2017] [Accepted: 10/01/2017] [Indexed: 12/23/2022] Open
Abstract
Destabilized heme proteins release heme, and free heme is toxic. Heme is now recognized as an agonist for the Toll-like receptor-4 (TLR4) receptor. This study examined whether the TLR4 receptor mediates the nephrotoxicity of heme, specifically, the effects of heme on renal blood flow and inflammatory responses. We blocked TLR4 signaling by the specific antagonist TAK-242. Intravenous administration of heme to mice promptly reduced renal blood flow, an effect attenuated by TAK-242. In vitro, TAK-242 reduced heme-elicited activation of NF-κB and its downstream gene monocyte chemoattractant protein-1(MCP-1); in contrast, TAK-242 failed to reduce heme-induced activation of the anti-inflammatory transcription factor Nrf2 and its downstream gene heme oxygenase-1 (HO-1). TAK-242 did not reduce heme-induced renal MCP-1 upregulation in vivo. TAK-242 did not reduce dysfunction and histological injury in the glycerol model of heme protein-induced acute kidney injury (AKI), findings corroborated by studies in TLR4+/+ and TLR4-/- mice. We conclude that 1) acute heme-mediated renal vasoconstriction occurs through TLR4 signaling; 2) proinflammatory effects of heme in renal epithelial cells involve TLR4 signaling, whereas the anti-inflammatory effects of heme do not; 3) TLR4 signaling does not mediate the proinflammatory effects of heme in the kidney; and 4) major mechanisms underlying glycerol-induced, heme protein-mediated AKI do not involve TLR4 signaling. These findings in the glycerol model are in stark contrast with findings in virtually all other AKI models studied to date and emphasize the importance of TLR4-independent pathways of heme protein-mediated injury in this model. Finally, these studies urge caution when using observations derived in vitro to predict what occurs in vivo.
Collapse
Affiliation(s)
- Karl A Nath
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - John D Belcher
- Division of Hematology, Oncology and Transplantation, University of Minnesota , Minneapolis, Minnesota
| | - Meryl C Nath
- Department of Pathology, Mayo Clinic , Rochester, Minnesota
| | | | - Anthony J Croatt
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | - Allan W Ackerman
- Division of Nephrology and Hypertension, Mayo Clinic , Rochester, Minnesota
| | | | - Gregory M Vercellotti
- Division of Hematology, Oncology and Transplantation, University of Minnesota , Minneapolis, Minnesota
| |
Collapse
|
20
|
Petersen MA, Ryu JK, Chang KJ, Etxeberria A, Bardehle S, Mendiola AS, Kamau-Devers W, Fancy SPJ, Thor A, Bushong EA, Baeza-Raja B, Syme CA, Wu MD, Rios Coronado PE, Meyer-Franke A, Yahn S, Pous L, Lee JK, Schachtrup C, Lassmann H, Huang EJ, Han MH, Absinta M, Reich DS, Ellisman MH, Rowitch DH, Chan JR, Akassoglou K. Fibrinogen Activates BMP Signaling in Oligodendrocyte Progenitor Cells and Inhibits Remyelination after Vascular Damage. Neuron 2017; 96:1003-1012.e7. [PMID: 29103804 DOI: 10.1016/j.neuron.2017.10.008] [Citation(s) in RCA: 110] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 08/30/2017] [Accepted: 10/04/2017] [Indexed: 12/20/2022]
Abstract
Blood-brain barrier (BBB) disruption alters the composition of the brain microenvironment by allowing blood proteins into the CNS. However, whether blood-derived molecules serve as extrinsic inhibitors of remyelination is unknown. Here we show that the coagulation factor fibrinogen activates the bone morphogenetic protein (BMP) signaling pathway in oligodendrocyte progenitor cells (OPCs) and suppresses remyelination. Fibrinogen induces phosphorylation of Smad 1/5/8 and inhibits OPC differentiation into myelinating oligodendrocytes (OLs) while promoting an astrocytic fate in vitro. Fibrinogen effects are rescued by BMP type I receptor inhibition using dorsomorphin homolog 1 (DMH1) or CRISPR/Cas9 activin A receptor type I (ACVR1) knockout in OPCs. Fibrinogen and the BMP target Id2 are increased in demyelinated multiple sclerosis (MS) lesions. Therapeutic depletion of fibrinogen decreases BMP signaling and enhances remyelination in vivo. Targeting fibrinogen may be an upstream therapeutic strategy to promote the regenerative potential of CNS progenitors in diseases with remyelination failure.
Collapse
Affiliation(s)
- Mark A Petersen
- Gladstone Institutes, San Francisco, CA, USA; Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Jae Kyu Ryu
- Gladstone Institutes, San Francisco, CA, USA
| | - Kae-Jiun Chang
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Ainhoa Etxeberria
- Department of Neurology, University of California, San Francisco, CA, USA
| | | | | | - Wanjiru Kamau-Devers
- Gladstone Institutes, San Francisco, CA, USA; Berkeley City College, Berkeley, CA, USA
| | - Stephen P J Fancy
- Department of Pediatrics, University of California, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA; Newborn Brain Research Institute, University of California, San Francisco, CA, USA
| | - Andrea Thor
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA, USA
| | - Eric A Bushong
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA, USA
| | | | | | - Michael D Wu
- Gladstone Institutes, San Francisco, CA, USA; Department of Anesthesia, University of California, San Francisco, CA, USA
| | | | | | - Stephanie Yahn
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Lauriane Pous
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jae K Lee
- Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Christian Schachtrup
- Institute of Anatomy and Cell Biology, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Hans Lassmann
- Center for Brain Research, Medical University of Vienna, Vienna, Austria
| | - Eric J Huang
- Department of Pathology, University of California, San Francisco, CA, USA
| | - May H Han
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Martina Absinta
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Reich
- Translational Neuroradiology Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Mark H Ellisman
- National Center for Microscopy and Imaging Research, Center for Research in Biological Systems, University of California, San Diego, La Jolla, CA, USA; Department of Neurosciences, University of California, San Diego, La Jolla, California, USA; Salk Institute for Biological Studies, La Jolla, San Diego, California, USA
| | - David H Rowitch
- Department of Pediatrics, University of California, San Francisco, CA, USA; Department of Neurosurgery, Eli and Edythe Broad Institute for Stem Cell Research and Regeneration Medicine, University of California, San Francisco, CA, USA; Department of Paediatrics, University of Cambridge, Cambridge, UK
| | - Jonah R Chan
- Department of Neurology, University of California, San Francisco, CA, USA
| | - Katerina Akassoglou
- Gladstone Institutes, San Francisco, CA, USA; Department of Neurology, University of California, San Francisco, CA, USA.
| |
Collapse
|
21
|
Miranda Acuña J, Hidalgo de la Cruz M, Ros AL, Tapia SP, Martínez Ginés ML, de Andrés Frutos CD. Elevated plasma fibrinogen levels in multiple sclerosis patients during relapse. Mult Scler Relat Disord 2017; 18:157-160. [PMID: 29141800 DOI: 10.1016/j.msard.2017.09.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 09/25/2017] [Accepted: 09/27/2017] [Indexed: 01/03/2023]
Abstract
Fibrinogen is a protein that plays a key role in blood coagulation and thrombosis and it is involved in several inflammatory processes; in multiple sclerosis (MS) may be related with blood-brain barrier (BBB) disruption. We analysed the relationship between plasma fibrinogen levels and the presence of active lesions on magnetic resonance imaging (MRI) during relapses of multiple sclerosis (MS) and clinically isolated syndrome (CIS) patients. METHODS We collected data of patients admitted to a tertiary-care hospital with relapse of MS and CIS from 2008 to 2013 and we analysed the relation between plasma fibrinogen levels (normal: 200mg/dl-417mg/dl) and the presence of active lesions on brain or spinal MRI. RESULTS A total of 58 patients were included, 45 (77%) CIS patients, 12 (21%) relapsing-remitting MS (RR-MS) and 1 (2%) active progressive MS (P-MS). 17 patients had high plasma fibrinogen levels (> 417mg/dl). Among total of patients with elevated plasma fibrinogen, 15 (88%) had active lesions on MRI and only 2 (12%) did not have active lesion on MRI (p = 0,013) with a specificity of 95%. All patients with fibrinogen > 417mg/dl were CIS (p = < 0.05). DISCUSSION We observed that high plasma fibrinogen levels had a high specificity high specificity, but low sensitivity (only 40%) for detection of active lesions on MRI during relapses of MS. These findings support the hypothesis that fibrinogen could play an important role on the development of MS lesions, however, additional studies are needed to confirm these results.
Collapse
Affiliation(s)
- Jahir Miranda Acuña
- Department of Neurology, Hospital General Universitario Gregorio Marañon, Madrid, Spain.
| | | | - Alberto Lozano Ros
- Department of Neurology, Hospital General Universitario Gregorio Marañon, Madrid, Spain
| | - Saskia Prasca Tapia
- Hemostasis and Thrombosis Division, Children's Hospital Los Angeles, Los Angeles, CA, USA
| | | | | |
Collapse
|
22
|
Cseplo P, Vamos Z, Torok O, Ivic I, Toth A, Buki A, Koller A. Hemolyzed Blood Elicits a Calcium Antagonist and High CO 2 Reversible Constriction via Elevation of [Ca 2+] i in Isolated Cerebral Arteries. J Neurotrauma 2017; 34:529-534. [PMID: 27018759 DOI: 10.1089/neu.2015.4365] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
During acute subarachnoid hemorrhage, blood is hemolyzed, which is followed by a significant cerebrovascular spasm resulting in a serious clinical condition. Interestingly, however, the direct vasomotor effect of perivascular hemolyzed blood (HB) has not yet been characterized, preventing the assessment of contribution of vasoconstrictor mechanisms deriving from brain tissue and/or blood and development of possible treatments. We hypothesized that perivascular HB reduces the diameter of the cerebral arteries (i.e., basilar artery [BA]; middle cerebral artery [MCA]) by elevating vascular tissue [Ca2+]i level. Vasomotor responses were measured by videomicroscopy and intracellular Ca2+ by the Fura2-AM ratiometric method. Adding HB to the vessel chamber reduced the diameter significantly (BA: from 264 ± 7 to 164 ± 11 μm; MCA: from 185 ± 15 to 155 ± 14 μm), which was reversed to control level by wash-out of HB. Potassium chloride (KCl), HB, serum, hemolyzed red blood cell (RBC), plasma, and platelet suspension (PLTs) elicited significant constrictions of isolated basilar arteries. There was a significant increase in K+ concentration in hemolyzed HB (7.02 ± 0.22 mmol/L) compared to Krebs' solution (6.20 ± 0.01 mmol/L). Before HB, acetylcholine (ACh), sodium-nitroprussid (SNP), nifedipin, and CO2 elicited substantial dilations in cerebral arteries. In contrast, in the presence of HB dilations to ACh, SNP decreased, but not to nifedipine and CO2. After washout of HB, nitric oxide-mediated dilations remained significantly reduced compared to control. HB significantly increased the ratiometric Ca signal, which returned to control level after washout. In conclusion, perivascular hemolyzed blood elicits significant-nifedipine and high CO2 reversible-constrictions of isolated BAs and MCAs, primarily by increasing intracellular Ca2+, findings that can contribute to the refinement of local treatment of subarachnoid hemorrhage.
Collapse
Affiliation(s)
- Peter Cseplo
- 1 Institute of Translational Medicine, University of Pecs , Medical School, Pecs, Hungary .,2 Department of Central Anesthesiology and Intensive Therapy, Petz Aladar County Teaching Hospital , Gyor, Hungary .,6 Szentagothai Research Centre, University of Pecs , Pecs, Hungary
| | - Zoltan Vamos
- 3 Department of Anaesthesiology and Intensive Therapy, University of Pecs , Medical School, Pecs, Hungary
| | - Orsolya Torok
- 1 Institute of Translational Medicine, University of Pecs , Medical School, Pecs, Hungary
| | - Ivan Ivic
- 1 Institute of Translational Medicine, University of Pecs , Medical School, Pecs, Hungary
| | - Attila Toth
- 4 Institute of Cardiology, Division of Clinical Physiology, Faculty of Medicine, University of Debrecen , Debrecen, Hungary
| | - Andras Buki
- 5 Department of Neurosurgery, University of Pecs , Medical School, Pecs, Hungary .,6 Szentagothai Research Centre, University of Pecs , Pecs, Hungary .,7 MTA-PTE Clinical Neuroscience MR Research Group , Pecs, Hungary
| | - Akos Koller
- 1 Institute of Translational Medicine, University of Pecs , Medical School, Pecs, Hungary .,5 Department of Neurosurgery, University of Pecs , Medical School, Pecs, Hungary .,6 Szentagothai Research Centre, University of Pecs , Pecs, Hungary .,8 Institute of Natural Sciences, University of Physical Education , Budapest, Hungary .,9 Department of Physiology, New York Medical College , Valhalla, New York
| |
Collapse
|
23
|
Diabetic Microvascular Disease and Pulmonary Fibrosis: The Contribution of Platelets and Systemic Inflammation. Int J Mol Sci 2016; 17:ijms17111853. [PMID: 27834824 PMCID: PMC5133853 DOI: 10.3390/ijms17111853] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/29/2022] Open
Abstract
Diabetes is strongly associated with systemic inflammation and oxidative stress, but its effect on pulmonary vascular disease and lung function has often been disregarded. Several studies identified restrictive lung disease and fibrotic changes in diabetic patients and in animal models of diabetes. While microvascular dysfunction is a well-known complication of diabetes, the mechanisms leading to diabetes-induced lung injury have largely been disregarded. We described the potential involvement of diabetes-induced platelet-endothelial interactions in perpetuating vascular inflammation and oxidative injury leading to fibrotic changes in the lung. Changes in nitric oxide synthase (NOS) activation and decreased NO bioavailability in the diabetic lung increase platelet activation and vascular injury and may account for platelet hyperreactivity reported in diabetic patients. Additionally, the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway has been reported to mediate pancreatic islet damage, and is implicated in the onset of diabetes, inflammation and vascular injury. Many growth factors and diabetes-induced agonists act via the JAK/STAT pathway. Other studies reported the contribution of the JAK/STAT pathway to the regulation of the pulmonary fibrotic process but the role of this pathway in the development of diabetic lung fibrosis has not been considered. These observations may open new therapeutic perspectives for modulating multiple pathways to mitigate diabetes onset or its pulmonary consequences.
Collapse
|
24
|
Lu P, Liu J, Pang X. Pravastatin inhibits fibrinogen- and FDP-induced inflammatory response via reducing the production of IL-6, TNF-α and iNOS in vascular smooth muscle cells. Mol Med Rep 2015; 12:6145-51. [PMID: 26238934 DOI: 10.3892/mmr.2015.4149] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 07/10/2015] [Indexed: 11/05/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory response of the arterial wall to pro‑atherosclerotic factors. As an inflammatory marker, fibrinogen directly participates in the pathogenesis of atherosclerosis. Our previous study demonstrated that fibrinogen and fibrin degradation products (FDP) produce a pro‑inflammatory effect on vascular smooth muscle cells (VSMCs) through inducing the production of interleukin‑6 (IL‑6), tumor necrosis factor‑α (TNF‑α) and inducible nitric oxide synthase (iNOS). In the present study, the effects of pravastatin on fibrinogen‑ and FDP‑induced expression of IL‑6, TNF‑α and iNOS were observed in VSMCs. The results showed that pravastatin dose‑dependently inhibited fibrinogen‑ and FDP‑stimulated expression of IL‑6, TNF‑α and iNOS in VSMCs at the mRNA and protein level. The maximal inhibition of protein expression of IL‑6, TNF‑α and iNOS was 46.9, 42.7 and 49.2% in fibrinogen‑stimulated VSMCs, and 50.2, 49.8 and 53.6% in FDP‑stimulated VSMCs, respectively. This suggests that pravastatin has the ability to relieve vascular inflammation via inhibiting the generation of IL‑6, TNF‑α and iNOS. The results of the present study may aid in further explaining the beneficial effects of pravastatin on atherosclerosis and related cardiovascular diseases. In addition, they suggest that application of pravastatin may be beneficial for prevention of atherosclerosis formation in hyperfibrinogenemia.
Collapse
Affiliation(s)
- Peipei Lu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Juntian Liu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaoming Pang
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
25
|
Wang J, Li H, He J, Li B, Bao Q, Zhang X, Lv Z, Zhang Y, Han J, Ai D, Zhu Y. 20-Hydroxyeicosatetraenoic acid involved in endothelial activation and thrombosis. Am J Physiol Heart Circ Physiol 2015; 308:H1359-67. [DOI: 10.1152/ajpheart.00802.2014] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2014] [Accepted: 03/04/2015] [Indexed: 12/27/2022]
Abstract
Endothelial cells play an important role in the process of coagulation and the function of platelets. We have previously reported that 20-hydroxyeicosatetraenoic acid (20-HETE), a metabolite of arachidonic acid, increased platelet aggregation and induced hemostasis. The purpose of the present study is to investigate whether 20-HETE-mediated endothelial activation has effect on the coagulation and platelet aggregation. C57Bl/6 mice were treated with PBS or 20-HETE (20 μg/kg) for 2 h, and then we performed a carotid artery or femoral artery thrombosis model by FeCl3. Detection of blood flow indicated that 20-HETE pretreatment accelerated formation of thrombus in both common carotid artery and femoral artery. In vitro, the secretion and expression of von Willebrand factor (vWF) in cultured human umbilical vein endothelial cells (HUVECs) with 20-HETE stimulation were increased, subsequently. The protein level of vWF in HUVECs was decreased at 1 h but increased with prolonged treatment with 20-HETE (>4 h). In contrast, vWF in the culture medium was increased under administration of 20-HETE at 1 h. As a result, adhesion of platelets on HUVECs was significantly increased by 20-HETE. In HUVECs, the extracellular signal-regulated kinase (ERK) pathway was activated by 20-HETE in a dose-dependent manner, and the inhibitors of ERK and L-type Ca2+channel blocked the release of vWF mediated by 20-HETE. In conclusion, 20-HETE instigates endothelial activation and induces the expression and secretion of vWF via the activation of ERK and calcium channel and therefore triggers thrombosis.
Collapse
Affiliation(s)
- Jiaxing Wang
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
| | - Hua Li
- Department of Pharmacology, College of Pharmacy, Dalian Medical University, Dalian, Liaoning, China
| | - Jinlong He
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Bochuan Li
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Qiankun Bao
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Xu Zhang
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Zhizhen Lv
- Institute of Vascular Medicine of Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Beijing, China; and
| | - Youyi Zhang
- Institute of Vascular Medicine of Peking University Third Hospital, Key Laboratory of Molecular Cardiovascular Sciences of Ministry of Education, Beijing, China; and
| | - Jingyan Han
- Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing, China
| | - Ding Ai
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| | - Yi Zhu
- Department of Physiology and Pathophysiology, Peking University Health Science Center, Beijing, China
- Collaborative Innovation Center of Tianjin for Medical Epigenetics and Department of Physiology and Pathophysiology, Tianjin Medical University, Tianjin, China
| |
Collapse
|
26
|
Wang S, Liu J, Wu DI, Pang X, Zhao J, Zhang X. Pro-inflammatory effect of fibrinogen on vascular smooth muscle cells by regulating the expression of PPARα, PPARγ and MMP-9. Biomed Rep 2015; 3:513-518. [PMID: 26171158 DOI: 10.3892/br.2015.459] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Accepted: 04/02/2015] [Indexed: 11/05/2022] Open
Abstract
Atherosclerosis is a chronic inflammatory disease in the vessel. As one of the inflammatory markers, fibrinogen has been indicated in formation and progression of atherosclerosis. However, it is completely unclear whether fibrinogen produces a pro-inflammatory effect on vascular smooth muscle cells (VSMCs). The purpose of the present study was to observe the effect of fibrinogen on the expression of peroxisome proliferator-activated receptors-α (PPARα), PPARγ and matrix metalloproteinase-9 (MMP-9) in VSMCs. Rat VSMCs were cultured and fibrinogen was used as a stimulant for PPARα, PPARγ and MMP-9 expression. mRNA expression of PPARα, PPARγ and MMP-9 was identified with the reverse transcription polymerase chain reaction. Protein production of PPARα and PPARγ was examined by western blot analysis and the MMP-9 level in the supernatant of VSMCs was measured with the enzyme-linked immunosorbent assay. The results showed that fibrinogen downregulated mRNA and protein expression of PPARα and PPARγ, and upregulated mRNA and protein generation of MMP-9 in VSMCs in time- and concentration-dependent manners. The maximal inhibition of protein expression of PPARα and PPARγ was 71.8 and 79.9%, respectively. The maximal release of MMP-9 was 4 times over the control. The results suggest that fibrinogen exerts a pro-inflammatory effect on VSMCs through inhibiting the expression of anti-inflammatory cytokine PPARα and PPARγ and stimulating the production of pro-inflammatory cytokine MMP-9. The findings provide new evidence for the pro-inflammatory and pro-atherosclerotic effects of fibrinogen.
Collapse
Affiliation(s)
- Shuyue Wang
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Juntian Liu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - D I Wu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaoming Pang
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Jingjing Zhao
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| | - Xiaolu Zhang
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
27
|
Muradashvili N, Benton RL, Saatman KE, Tyagi SC, Lominadze D. Ablation of matrix metalloproteinase-9 gene decreases cerebrovascular permeability and fibrinogen deposition post traumatic brain injury in mice. Metab Brain Dis 2015; 30:411-26. [PMID: 24771110 PMCID: PMC4213324 DOI: 10.1007/s11011-014-9550-3] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/15/2014] [Indexed: 12/14/2022]
Abstract
Traumatic brain injury (TBI) is accompanied with enhanced matrix metalloproteinase-9 (MMP-9) activity and elevated levels of plasma fibrinogen (Fg), which is a known inflammatory agent. Activation of MMP-9 and increase in blood content of Fg (i.e. hyperfibrinogenemia, HFg) both contribute to cerebrovascular disorders leading to blood brain barrier disruption. It is well-known that activation of MMP-9 contributes to vascular permeability. It has been shown that at an elevated level (i.e. HFg) Fg disrupts blood brain barrier. However, mechanisms of their actions during TBI are not known. Mild TBI was induced in wild type (WT, C57BL/6 J) and MMP-9 gene knockout (Mmp9(-/-)) homozygous, mice. Pial venular permeability to fluorescein isothiocyanate-conjugated bovine serum albumin in pericontusional area was observed 14 days after injury. Mice memory was tested with a novel object recognition test. Increased expression of Fg endothelial receptor intercellular adhesion protein-1 and formation of caveolae were associated with enhanced activity of MMP-9 causing an increase in pial venular permeability. As a result, an enhanced deposition of Fg and cellular prion protein (PrP(C)) were found in pericontusional area. These changes were attenuated in Mmp9(-/-) mice and were associated with lesser loss of short-term memory in these mice than in WT mice. Our data suggest that mild TBI-induced increased cerebrovascular permeability enhances deposition of Fg-PrP(C) and loss of memory, which is ameliorated in the absence of MMP-9 activity. Thus, targeting MMP-9 activity and blood level of Fg can be a possible therapeutic remedy to diminish vasculo-neuronal damage after TBI.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - Richard L. Benton
- Department of Anatomical Sciences and Neurobiology and Kentucky Spinal Cord Injury Research Center (KSCIRC), University of Louisville, School of Medicine, Louisville, KY
| | - Kathryn E. Saatman
- Department of Physiology and Neurosurgery and Spinal Cord & Brain Injury Research Center (SCoBIRC), University of Kentucky, Lexington, KY, USA
| | - Suresh C. Tyagi
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
| | - David Lominadze
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY
- Corresponding Author: David Lominadze, Ph. D., University of Louisville, Dept. of Physiology & Biophysics, School of Medicine, Bldg. A, Room 1115, 500 South Preston Street, Louisville, KY 40202, Phone (502) 852-4902, Fax (502) 852-6239,
| |
Collapse
|
28
|
Nigro C, Raciti GA, Leone A, Fleming TH, Longo M, Prevenzano I, Fiory F, Mirra P, D'Esposito V, Ulianich L, Nawroth PP, Formisano P, Beguinot F, Miele C. Methylglyoxal impairs endothelial insulin sensitivity both in vitro and in vivo. Diabetologia 2014; 57:1485-94. [PMID: 24759959 DOI: 10.1007/s00125-014-3243-7] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Accepted: 03/27/2014] [Indexed: 10/25/2022]
Abstract
AIMS/HYPOTHESIS Insulin exerts a direct action on vascular cells, thereby affecting the outcome and progression of diabetic vascular complications. However, the mechanism through which insulin signalling is impaired in the endothelium of diabetic individuals remains unclear. In this work, we have evaluated the role of the AGE precursor methylglyoxal (MGO) in generating endothelial insulin resistance both in cells and in animal models. METHODS Time course experiments were performed on mouse aortic endothelial cells (MAECs) incubated with 500 μmol/l MGO. The glyoxalase-1 inhibitor S-p-bromobenzylglutathione-cyclopentyl-diester (SpBrBzGSHCp2) was used to increase the endogenous levels of MGO. For the in vivo study, an MGO solution was administrated i.p. to C57BL/6 mice for 7 weeks. RESULTS MGO prevented the insulin-dependent activation of the IRS1/protein kinase Akt/endothelial nitric oxide synthase (eNOS) pathway, thereby blunting nitric oxide (NO) production, while extracellular signal-regulated kinase (ERK1/2) activation and endothelin-1 (ET-1) release were increased by MGO in MAECs. Similar results were obtained in MAECs treated with SpBrBzGSHCp2. In MGO- and SpBrBzGSHCp2-exposed cells, inhibition of ERK1/2 decreased IRS1 phosphorylation on S616 and rescued insulin-dependent Akt activation and NO generation, indicating that MGO inhibition of the IRS1/Akt/eNOS pathway is mediated, at least in part, by ERK1/2. Chronic administration of MGO to C57BL/6 mice impaired whole-body insulin sensitivity and induced endothelial insulin resistance. CONCLUSIONS/INTERPRETATION MGO impairs the action of insulin on the endothelium both in vitro and in vivo, at least in part through an ERK1/2-mediated mechanism. These findings may be instrumental in developing novel strategies for preserving endothelial function in diabetes.
Collapse
Affiliation(s)
- Cecilia Nigro
- Institute of Experimental Endocrinology and Oncology 'G. Salvatore', National Council of Research, Via Pansini 5, 80131, Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Muradashvili N, Khundmiri SJ, Tyagi R, Gartung A, Dean WL, Lee MJ, Lominadze D. Sphingolipids affect fibrinogen-induced caveolar transcytosis and cerebrovascular permeability. Am J Physiol Cell Physiol 2014; 307:C169-79. [PMID: 24829496 DOI: 10.1152/ajpcell.00305.2013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Inflammation-induced vascular endothelial dysfunction can allow plasma proteins to cross the vascular wall, causing edema. Proteins may traverse the vascular wall through two main pathways, the paracellular and transcellular transport pathways. Paracellular transport involves changes in endothelial cell junction proteins, while transcellular transport involves caveolar transcytosis. Since both processes are associated with filamentous actin formation, the two pathways are interconnected. Therefore, it is difficult to differentiate the prevailing role of one or the other pathway during various pathologies causing an increase in vascular permeability. Using a newly developed dual-tracer probing method, we differentiated transcellular from paracellular transport during hyperfibrinogenemia (HFg), an increase in fibrinogen (Fg) content. Roles of cholesterol and sphingolipids in formation of functional caveolae were assessed using a cholesterol chelator, methyl-β-cyclodextrin, and the de novo sphingolipid synthesis inhibitor myriocin. Fg-induced formation of functional caveolae was defined by association and colocalization of Na+-K+-ATPase and plasmalemmal vesicle-associated protein-1 with use of Förster resonance energy transfer and total internal reflection fluorescence microscopy, respectively. HFg increased permeability of the endothelial cell layer mainly through the transcellular pathway. While MβCD blocked Fg-increased transcellular and paracellular transport, myriocin affected only transcellular transport. Less pial venular leakage of albumin was observed in myriocin-treated HFg mice. HFg induced greater formation of functional caveolae, as indicated by colocalization of Na+-K+-ATPase with plasmalemmal vesicle-associated protein-1 by Förster resonance energy transfer and total internal reflection fluorescence microscopy. Our results suggest that elevated blood levels of Fg alter cerebrovascular permeability mainly by affecting caveolae-mediated transcytosis through modulation of de novo sphingolipid synthesis.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Syed Jalal Khundmiri
- Kidney Disease Program, Department of Medicine, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Reeta Tyagi
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Allison Gartung
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan
| | - William L Dean
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Louisville, Louisville, Kentucky; and
| | - Menq-Jer Lee
- Department of Pathology, School of Medicine, Wayne State University, Detroit, Michigan
| | - David Lominadze
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky;
| |
Collapse
|
30
|
Fibrinogen plasma concentration is an independent marker of haemodynamic impairment in chronic thromboembolic pulmonary hypertension. Sci Rep 2014; 4:4808. [PMID: 24770447 PMCID: PMC5381222 DOI: 10.1038/srep04808] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 03/24/2014] [Indexed: 01/09/2023] Open
Abstract
Fibrinogen has a crucial role in both inflammation and coagulation, two processes pivotal for the pathogenesis of pulmonary hypertension. We therefore aimed to investigate whether fibrinogen plasma concentrations a) are elevated in pulmonary arterial hypertension (PAH) and chronic thromboembolic pulmonary hypertension (CTEPH) and b) may serve as a novel biomarker for haemodynamic impairment. In a dual-centre, retrospective analysis including 112 patients with PAH (n = 52), CTEPH (n = 49) and a control cohort of patients with suspected PAH ruled out by right heart catheterisation (n = 11), we found fibrinogen plasma concentrations to be increased in patients with PAH (4.1 ± 1.4 g/l) and CTEPH (4.3 ± 1.2 g/l) compared to control patients (3.4 ± 0.5 g/l, p = 0.0035 and p = 0.0004, respectively). In CTEPH patients but not in PAH patients fibrinogen was associated with haemodynamics (p < 0.036) and functional parameters (p < 0.041). Furthermore, fibrinogen was linked to disease severity (WHO functional class, p = 0.017) and independently predicted haemodynamic impairment specifically in CTEPH (p < 0.016). Therefore, fibrinogen seems to represent an important factor in CTEPH pathophysiology and may have the potential to guide clinical diagnosis and therapy.
Collapse
|
31
|
Endothelin receptor polymorphisms in the cardiovascular system: potential implications for therapy and screening. Heart Fail Rev 2014; 19:743-58. [DOI: 10.1007/s10741-014-9426-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
32
|
Bürgin-Maunder CS, Brooks PR, Russell FD. Omega-3 fatty acids modulate Weibel-Palade body degranulation and actin cytoskeleton rearrangement in PMA-stimulated human umbilical vein endothelial cells. Mar Drugs 2013; 11:4435-50. [PMID: 24217286 PMCID: PMC3853737 DOI: 10.3390/md11114435] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2013] [Revised: 10/15/2013] [Accepted: 10/22/2013] [Indexed: 01/03/2023] Open
Abstract
Long chain omega-3 polyunsaturated fatty acids (LC n-3 PUFAs) produce cardiovascular benefits by improving endothelial function. Endothelial cells store von Willebrand factor (vWF) in cytoplasmic Weibel-Palade bodies (WPBs). We examined whether LC n-3 PUFAs regulate WPB degranulation using cultured human umbilical vein endothelial cells (HUVECs). HUVECs were incubated with or without 75 or 120 µM docosahexaenoic acid or eicosapentaenoic acid for 5 days at 37 °C. WPB degranulation was stimulated using phorbol 12-myristate 13-acetate (PMA), and this was assessed by immunocytochemical staining for vWF. Actin reorganization was determined using phalloidin-TRITC staining. We found that PMA stimulated WPB degranulation, and that this was significantly reduced by prior incubation of cells with LC n-3 PUFAs. In these cells, WPBs had rounded rather than rod-shaped morphology and localized to the perinuclear region, suggesting interference with cytoskeletal remodeling that is necessary for complete WPB degranulation. In line with this, actin rearrangement was altered in cells containing perinuclear WPBs, where cells exhibited a thickened actin rim in the absence of prominent cytoplasmic stress fibers. These findings indicate that LC n-3 PUFAs provide some protection against WBP degranulation, and may contribute to an improved understanding of the anti-thrombotic effects previously attributed to LC n-3 PUFAs.
Collapse
Affiliation(s)
- Corinna S Bürgin-Maunder
- Inflammation and Healing Research Cluster, School of Health and Sport Sciences, University of the Sunshine Coast, Maroochydore, Queensland 4556, Australia.
| | | | | |
Collapse
|
33
|
Abstract
At least 468 individual genes have been manipulated by molecular methods to study their effects on the initiation, promotion, and progression of atherosclerosis. Most clinicians and many investigators, even in related disciplines, find many of these genes and the related pathways entirely foreign. Medical schools generally do not attempt to incorporate the relevant molecular biology into their curriculum. A number of key signaling pathways are highly relevant to atherogenesis and are presented to provide a context for the gene manipulations summarized herein. The pathways include the following: the insulin receptor (and other receptor tyrosine kinases); Ras and MAPK activation; TNF-α and related family members leading to activation of NF-κB; effects of reactive oxygen species (ROS) on signaling; endothelial adaptations to flow including G protein-coupled receptor (GPCR) and integrin-related signaling; activation of endothelial and other cells by modified lipoproteins; purinergic signaling; control of leukocyte adhesion to endothelium, migration, and further activation; foam cell formation; and macrophage and vascular smooth muscle cell signaling related to proliferation, efferocytosis, and apoptosis. This review is intended primarily as an introduction to these key signaling pathways. They have become the focus of modern atherosclerosis research and will undoubtedly provide a rich resource for future innovation toward intervention and prevention of the number one cause of death in the modern world.
Collapse
Affiliation(s)
- Paul N Hopkins
- Cardiovascular Genetics, Department of Internal Medicine, University of Utah, Salt Lake City, Utah, USA.
| |
Collapse
|
34
|
Abstract
Traumatic brain injury (TBI) has been associated with various neurological disorders. However, the role of cerebrovascular dysfunction and its mechanisms associated with TBI are still not well understood. Inflammation is the main cause of vascular dysfunction. It affects properties of blood components and the vascular wall leading to changes in blood flow and in interaction of blood components and vascular endothelium exacerbating microcirculatory complications during inflammatory diseases. One of the markers of inflammation is a plasma adhesion protein, fibrinogen (Fg). At elevated levels, Fg can also cause inflammatory responses. One of the manifestations of inflammatory responses is an increase in microvascular permeability leading to accumulation of plasma proteins in the subendothelial matrix and causing vascular remodelling. This has a most devastating effect on cerebral circulation after TBI that is accompanied with an elevation of plasma level of Fg and with an increased cerebrovascular permeability in injury penumbra impairing the normal healing process. This study reviews cerebrovascular alterations after TBI, considers the consequences of increased blood-brain barrier permeability, defines the role of elevated level of Fg and discusses the potential mechanisms of its action leading to vascular dysfunction, which subsequently can cause impairment in neuronal function. Thus, possible mechanisms of vasculo-neuronal dysfunction after TBI are considered.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine , Louisville, KY , USA
| | | |
Collapse
|
35
|
Qipshidze N, Metreveli N, Mishra PK, Lominadze D, Tyagi SC. Hydrogen sulfide mitigates cardiac remodeling during myocardial infarction via improvement of angiogenesis. Int J Biol Sci 2012; 8:430-41. [PMID: 22419888 PMCID: PMC3303169 DOI: 10.7150/ijbs.3632] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Accepted: 01/12/2012] [Indexed: 11/17/2022] Open
Abstract
Exogenous hydrogen sulfide (H2S) leads to down-regulation of inflammatory responses and provides myocardial protection during acute ischemia/reperfusion injury; however its role during chronic heart failure (CHF) due to myocardial infarction (MI) is yet to be unveiled. We previously reported that H2S inhibits antiangiogenic factors such, as endostatin and angiostatin, but a little is known about its effect on parstatin (a fragment of proteinase-activated receptor-1, PAR-1). We hypothesize that H2S inhibits parstatin formation and promotes VEGF activation, thus promoting angiogenesis and significantly limiting the extent of MI injury. To verify this hypothesis MI was created in 12 week-old male mice by ligation of left anterior descending artery (LAD). Sham surgery was performed except LAD ligation. After the surgery mice were treated with sodium hydrogen sulfide (30 μmol/l NaHS, a donor for H2S, in drinking water) for 4 weeks. The LV tissue was analyzed for VEGF, flk-1 and flt-1, endostatin, angiostatin and parstatin. The expression of VEGF, flk-1 and flt-1 were significantly increased in treated mice while the level of endostatin, angiostatin and parstatin were decreased compared to in untreated mice. The echocardiography in mice treated with H2S showed the improvement of heart function compared to in untreated mice. The X-ray and Doppler blood flow measurements showed enhancement of cardiac-angiogenesis in mice treated with H2S. This observed cytoprotection was associated with an inhibition of anti-angiogenic proteins and stimulation of angiogenic factors. We established that administration of H2S at the time of MI ameliorated infarct size and preserved LV function during development of MI in mice. These results suggest that H2S is cytoprotective and angioprotective during evolution of MI.
Collapse
Affiliation(s)
- Natia Qipshidze
- Department of Physiology, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | | | | | | | | |
Collapse
|
36
|
Abe N, Okuhira M, Tsutsui C, Murata Y, Nakamura Y. Cytotoxicity of benzyl isothiocyanate in normal renal proximal tubular cells and its modulation by glutathione. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:1887-1892. [PMID: 22296293 DOI: 10.1021/jf2052042] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
In the present study, we examined the toxicity of benzyl ITC (BITC) and its urinary mercapturic acid metabolite (BITC-NAC), using a normal renal proximal tubular cell line, pig LLC-PK1. BITC increased cell death with an IC(50) value of about 7 μM, whereas the cytotoxic effect of BITC-NAC was five times weaker than that of BITC. We observed a significant necrosis of the compounds on LLC-PK1 cells with oxidative stress. In the presence of 5 mM glutathione (GSH), comparable to physiological levels, the cytotoxicity of BITC-NAC as well as BITC was significantly reduced. Furthermore, the increase in intracellular GSH levels by pretreatment with NAC before the BITC treatment resulted in inhibition of the BITC-induced necrotic events as well as intracellular oxidative stress. These results suggest that GSH is a determinant of cellular resistance against the BITC-mediated and oxidative stress-dependent cytotoxicity in renal proximal tubular cells.
Collapse
Affiliation(s)
- Naomi Abe
- Graduate School of Natural Science and Technology, Okayama University, Okayama, Japan
| | | | | | | | | |
Collapse
|
37
|
Qipshidze N, Tyagi N, Metreveli N, Lominadze D, Tyagi SC. Autophagy mechanism of right ventricular remodeling in murine model of pulmonary artery constriction. Am J Physiol Heart Circ Physiol 2012; 302:H688-96. [PMID: 22101525 PMCID: PMC3353777 DOI: 10.1152/ajpheart.00777.2011] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 11/03/2011] [Indexed: 01/24/2023]
Abstract
Although right ventricular failure (RVF) is the hallmark of pulmonary arterial hypertension (PAH), the mechanism of RVF is unclear. Development of PAH-induced RVF is associated with an increased reactive oxygen species (ROS) production. Increases in oxidative stress lead to generation of nitro-tyrosine residues in tissue inhibitor of metalloproteinase (TIMPs) and liberate active matrix metalloproteinase (MMPs). To test the hypothesis that an imbalance in MMP-to-TIMP ratio leads to interstitial fibrosis and RVF and whether the treatment with folic acid (FA) alleviates ROS generation, maintains MMP/TIMP balance, and regresses interstitial fibrosis, we used a mouse model of pulmonary artery constriction (PAC). After surgery mice were given FA in their drinking water (0.03 g/l) for 4 wk. Production of ROS in the right ventricle (RV) was measured using oxidative fluorescent dye. The level of MMP-2, -9, and -13 and TIMP-4, autophagy marker (p62), mitophagy marker (LC3A/B), collagen interstitial fibrosis, and ROS in the RV wall was measured. RV function was measured by Millar catheter. Treatment with FA decreased the pressure to 35 mmHg from 50 mmHg in PAC mice. Similarly, RV volume in PAC mice was increased compared with the Sham group. A robust increase of ROS was observed in RV of PAC mice, which was decreased by treatment with FA. The protein level of MMP-2, -9, and -13 was increased in RV of PAC mice in comparison with that in the sham-operated mice, whereas supplementation with FA abolished this effect and mitigated MMPs levels. The protein level of TIMP-4 was decreased in RV of PAC mice compared with the Sham group. Treatment with FA helped PAC mice to improve the level of TIMP-4. To further support the claim of mitophagy occurrence during RVF, the levels of LC3A/B and p62 were measured by Western blot and immunohistochemistry. LC3A/B was increased in RV of PAC mice. Similarly, increased p62 protein level was observed in RV of PAC mice. Treatment with FA abolished this effect in PAC mice. These results suggest that FA treatment improves MMP/TIMP balance and ameliorates mitochondrial dysfunction that results in protection of RV failure during pulmonary hypertension.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Animals
- Autophagy/drug effects
- Autophagy/physiology
- Biomarkers/metabolism
- Disease Models, Animal
- Folic Acid/pharmacology
- Heart Ventricles/metabolism
- Heart Ventricles/pathology
- Heart Ventricles/physiopathology
- Hypertension, Pulmonary/drug therapy
- Hypertension, Pulmonary/metabolism
- Hypertension, Pulmonary/pathology
- Hypertension, Pulmonary/physiopathology
- Hypertrophy, Right Ventricular/drug therapy
- Hypertrophy, Right Ventricular/metabolism
- Hypertrophy, Right Ventricular/pathology
- Hypertrophy, Right Ventricular/physiopathology
- Male
- Matrix Metalloproteinase 13/metabolism
- Matrix Metalloproteinase 2/metabolism
- Matrix Metalloproteinase 9/metabolism
- Mice
- Mice, Inbred C57BL
- Pulmonary Artery/metabolism
- Pulmonary Artery/pathology
- Pulmonary Artery/physiopathology
- Reactive Oxygen Species/metabolism
- Tissue Inhibitor of Metalloproteinases/metabolism
- Ventricular Remodeling/drug effects
- Ventricular Remodeling/physiology
- Vitamin B Complex/pharmacology
- Tissue Inhibitor of Metalloproteinase-4
Collapse
Affiliation(s)
- Natia Qipshidze
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40202, USA.
| | | | | | | | | |
Collapse
|
38
|
Muradashvili N, Qipshidze N, Munjal C, Givvimani S, Benton RL, Roberts AM, Tyagi SC, Lominadze D. Fibrinogen-induced increased pial venular permeability in mice. J Cereb Blood Flow Metab 2012; 32:150-63. [PMID: 21989482 PMCID: PMC3256415 DOI: 10.1038/jcbfm.2011.144] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Elevated blood level of Fibrinogen (Fg) is commonly associated with vascular dysfunction. We tested the hypothesis that at pathologically high levels, Fg increases cerebrovascular permeability by activating matrix metalloproteinases (MMPs). Fibrinogen (4 mg/mL blood concentration) or equal volume of phosphate-buffered saline (PBS) was infused into male wild-type (WT; C57BL/6J) or MMP-9 gene knockout (MMP9-/-) mice. Pial venular leakage of fluorescein isothiocyanate-bovine serum albumin to Fg or PBS alone and to topically applied histamine (10(-5) mol/L) were assessed. Intravital fluorescence microscopy and image analysis were used to assess cerebrovascular protein leakage. Pial venular macromolecular leakage increased more after Fg infusion than after infusion of PBS in both (WT and MMP9-/-) mice but was more pronounced in WT compared with MMP9-/- mice. Expression of vascular endothelial cadherin (VE-cadherin) was less and plasmalemmal vesicle-associated protein-1 (PV-1) was greater in Fg-infused than in PBS-infused both mice groups. However, in MMP9-/- mice, VE-cadherin expression was greater and PV-1 expression was less than in WT mice. These data indicate that at higher levels, Fg compromises microvascular integrity through activation of MMP-9 and downregulation of VE-cadherin and upregulation of PV-1. Our results suggest that elevated blood level of Fg could have a significant role in cerebrovascular dysfunction and remodeling.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Davalos D, Akassoglou K. Fibrinogen as a key regulator of inflammation in disease. Semin Immunopathol 2011; 34:43-62. [PMID: 22037947 DOI: 10.1007/s00281-011-0290-8] [Citation(s) in RCA: 621] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 10/03/2011] [Indexed: 12/11/2022]
Abstract
The interaction of coagulation factors with the perivascular environment affects the development of disease in ways that extend beyond their traditional roles in the acute hemostatic cascade. Key molecular players of the coagulation cascade like tissue factor, thrombin, and fibrinogen are epidemiologically and mechanistically linked with diseases with an inflammatory component. Moreover, the identification of novel molecular mechanisms linking coagulation and inflammation has highlighted factors of the coagulation cascade as new targets for therapeutic intervention in a wide range of inflammatory human diseases. In particular, a proinflammatory role for fibrinogen has been reported in vascular wall disease, stroke, spinal cord injury, brain trauma, multiple sclerosis, Alzheimer's disease, rheumatoid arthritis, bacterial infection, colitis, lung and kidney fibrosis, Duchenne muscular dystrophy, and several types of cancer. Genetic and pharmacologic studies have unraveled pivotal roles for fibrinogen in determining the extent of local or systemic inflammation. As cellular and molecular mechanisms for fibrinogen functions in tissues are identified, the role of fibrinogen is evolving from a marker of vascular rapture to a multi-faceted signaling molecule with a wide spectrum of functions that can tip the balance between hemostasis and thrombosis, coagulation and fibrosis, protection from infection and extensive inflammation, and eventually life and death. This review will discuss some of the main molecular links between coagulation and inflammation and will focus on the role of fibrinogen in inflammatory disease highlighting its unique structural properties, cellular targets, and signal transduction pathways that make it a potent proinflammatory mediator and a potential therapeutic target.
Collapse
Affiliation(s)
- Dimitrios Davalos
- Gladstone Institute of Neurological Disease, University of California, San Francisco, San Francisco, CA 94158, USA
| | | |
Collapse
|
40
|
Muradashvili N, Tyagi N, Tyagi R, Munjal C, Lominadze D. Fibrinogen alters mouse brain endothelial cell layer integrity affecting vascular endothelial cadherin. Biochem Biophys Res Commun 2011; 413:509-14. [PMID: 21920349 DOI: 10.1016/j.bbrc.2011.07.133] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2011] [Accepted: 07/31/2011] [Indexed: 12/24/2022]
Abstract
Many inflammatory diseases are associated with elevated blood concentration of fibrinogen (Fg) leading to vascular dysfunction. We showed that pathologically high (4 mg/ml) content of Fg disrupts integrity of endothelial cell (EC) layer and causes macromolecular leakage affecting tight junction proteins. However, role of adherence junction proteins, particularly vascular endothelial cadherin (VE-cadherin) and matrix metalloproteinase-9 (MMP-9) in this process is not clear. We tested the hypothesis that at high levels Fg affects integrity of mouse brain endothelial cell (MBEC) monolayer through activation of MMP-9 and downregulation of VE-cadherin expression and in part its translocation to the cytosol. The effect of Fg on cultured MBEC layer integrity was assessed by measuring transendothelial electrical resistance. Cellular expression and translocation of VE-cadherin were assessed by Western blot and immunohistochemical analyses, (respectively). Our results suggest that high content of Fg decreased VE-cadherin expression at protein and mRNA levels. Fg induced translocation of VE-cadherin to cytosol, which led to disruption of cell-to-cell interaction and cell to subendothelial matrix attachment. Fg-induced alterations in cell layer integrity and their attachment were diminished during inhibition of MMP-9 activity. Thus Fg compromises EC layer integrity causing downregulation and translocation of VE-cadherin and through MMP-9 activation. These results suggest that increased level of Fg could play a significant role in vascular dysfunction and remodeling.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology and Biophysics, University of Louisville, School of Medicine, Louisville, KY 40202, USA
| | | | | | | | | |
Collapse
|
41
|
Lu PP, Liu JT, Liu N, Guo F, Ji YY, Pang X. Pro-inflammatory effect of fibrinogen and FDP on vascular smooth muscle cells by IL-6, TNF-α and iNOS. Life Sci 2011; 88:839-45. [PMID: 21439977 DOI: 10.1016/j.lfs.2011.03.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2010] [Revised: 02/15/2011] [Accepted: 03/03/2011] [Indexed: 02/08/2023]
Abstract
AIMS Atherosclerosis is a chronic inflammatory response of the arterial wall to multiple endothelial injuries. As one of the inflammatory markers, fibrinogen has been implicated in pathogenesis of atherosclerosis. But, it is not completely understood whether atherogenesis of fibrinogen is related to its pro-inflammatory effect on vascular smooth muscle cells (VSMCs). The purpose of the present study was to observe effects of fibrinogen and fibrin degradation products (FDP) on interleukin-6 (IL-6), tumor necrosis factor alpha (TNF-α), and inducible nitric oxide synthase (iNOS) generation in rat VSMCs. MAIN METHODS Rat VSMCs were cultured, and fibrinogen and FDP were used as stimulants for IL-6, TNF-α, and iNOS. IL-6 and TNF-α level in the supernatant were measured by ELISA, mRNA expression of IL-6, TNF-α, and iNOS were assayed with RT-PCR, and protein expression of iNOS was detected with western blot and immunocytochemistry. KEY FINDINGS Fibrinogen and FDP both significantly stimulated mRNA and protein expressions of IL-6, TNF-α and iNOS in VSMCs in time- and concentration-dependent ways. The pro-inflammatory potency of FDP is higher than fibrinogen, which seems to mean that smaller fragments of the protein have greater pro-inflammatory activity. Fibrinogen and FDP promote more protein expressions of IL-6 and TNF-α compared to iNOS, suggesting that fibrinogen and FDP produce a pro-inflammatory effect on VSMCs mainly by IL-6 and TNF-α. SIGNIFICANCE These findings are helpful to better understand pro-inflammatory effect of fibrinogen on VSMCs involved in atherogenesis, and imply a therapeutic strategy targeting hyperfibrinogenemia in atherosclerosis.
Collapse
Affiliation(s)
- Pei-pei Lu
- Department of Pharmacology, Xi'an Jiaotong University School of Medicine, PR China
| | | | | | | | | | | |
Collapse
|
42
|
Qipshidze N, Tyagi N, Sen U, Givvimani S, Metreveli N, Lominadze D, Tyagi SC. Folic acid mitigated cardiac dysfunction by normalizing the levels of tissue inhibitor of metalloproteinase and homocysteine-metabolizing enzymes postmyocardial infarction in mice. Am J Physiol Heart Circ Physiol 2010; 299:H1484-93. [PMID: 20802128 DOI: 10.1152/ajpheart.00577.2010] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Myocardial infarction (MI) results in significant metabolic derangement, causing accumulation of metabolic by product, such as homocysteine (Hcy). Hcy is a nonprotein amino acid generated during nucleic acid methylation and demethylation of methionine. Folic acid (FA) decreases Hcy levels by remethylating the Hcy to methionine, by 5-methylene tetrahydrofolate reductase (5-MTHFR). Although clinical trials were inconclusive regarding the role of Hcy in MI, in animal models, the levels of 5-MTHFR were decreased, and FA mitigated the MI injury. We hypothesized that FA mitigated MI-induced injury, in part, by mitigating cardiac remodeling during chronic heart failure. Thus, MI was induced in 12-wk-old male C57BL/J mice by ligating the left anterior descending artery, and FA (0.03 g/l in drinking water) was administered for 4 wk after the surgery. Cardiac function was assessed by echocardiography and by a Millar pressure-volume catheter. The levels of Hcy-metabolizing enzymes, cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), and 5-MTHFR, were estimated by Western blot analyses. The results suggest that FA administered post-MI significantly improved cardiac ejection fraction and induced tissue inhibitor of metalloproteinase, CBS, CSE, and 5-MTHFR. We showed that FA supplementation resulted in significant improvement of myocardial function after MI. The study eluted the importance of homocysteine (Hcy) metabolism and FA supplementation in cardiovascular disease.
Collapse
Affiliation(s)
- Natia Qipshidze
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, Kentucky 40202, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Thorin E, Clozel M. The cardiovascular physiology and pharmacology of endothelin-1. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2010; 60:1-26. [PMID: 21081213 PMCID: PMC3693982 DOI: 10.1016/b978-0-12-385061-4.00001-5] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
One year after the discovery in 1980 that the endothelium was obligatory for acetylcholine to relax isolated arteries, it was clearly shown that the endothelium could also promote contraction. In 1988, Dr Yanagisawa's group identified endothelin-1 (ET-1) as the first endothelium-derived contracting factor. The circulating levels of this short (21 amino acids) peptide were quickly determined in humans and it was reported that in most cardiovascular diseases, circulating levels of ET-1 were increased and ET-1 was then recognized as a likely mediator of pathological vasoconstriction in human. The discovery of two receptor subtypes in 1990, ET(A) and ET(B), permitted optimization of bosentan, which entered clinical development in 1993, and was offered to patients with pulmonary arterial hypertension in 2001. In this report, we discuss the physiological and pathophysiological role of endothelium-derived ET-1, the pharmacology of its two receptors, focusing on the regulation of the vascular tone and as much as possible in humans. The coronary bed will be used as a running example, but references to the pulmonary, cerebral, and renal circulation will also be made. Many of the cardiovascular complications associated with aging and cardiovascular risk factors are initially attributable, at least in part, to endothelial dysfunction, particularly dysregulation of the vascular function associated with an imbalance in the close interdependence of NO and ET-1, in which the implication of the ET(B) receptor may be central.
Collapse
Affiliation(s)
- Eric Thorin
- Department of Surgery, Montreal Heart Institute, Research Center, Université de Montréal, Montreal, Quebec, Canada
| | | |
Collapse
|
44
|
Lominadze D, Dean WL, Tyagi SC, Roberts AM. Mechanisms of fibrinogen-induced microvascular dysfunction during cardiovascular disease. Acta Physiol (Oxf) 2010; 198:1-13. [PMID: 19723026 DOI: 10.1111/j.1748-1716.2009.02037.x] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Fibrinogen (Fg) is a high molecular weight plasma adhesion protein and a biomarker of inflammation. Many cardiovascular and cerebrovascular disorders are accompanied by increased blood content of Fg. Increased levels of Fg result in changes in blood rheological properties such as increases in plasma viscosity, erythrocyte aggregation, platelet thrombogenesis, alterations in vascular reactivity and compromises in endothelial layer integrity. These alterations exacerbate the complications in peripheral blood circulation during cardiovascular diseases such as hypertension, diabetes and stroke. In addition to affecting blood viscosity by altering plasma viscosity and erythrocyte aggregation, growing experimental evidence suggests that Fg alters vascular reactivity and impairs endothelial cell layer integrity by binding to its endothelial cell membrane receptors and activating signalling mechanisms. The purpose of this review is to discuss experimental data, which demonstrate the effects of Fg causing vascular dysfunction and to offer possible mechanisms for these effects, which could exacerbate microcirculatory complications during cardiovascular diseases accompanied by increased Fg content.
Collapse
Affiliation(s)
- D Lominadze
- Department of Physiology and Biophysics, School of Medicine, University of Louisville, Louisville, KY 40292, USA.
| | | | | | | |
Collapse
|
45
|
Patibandla PK, Tyagi N, Dean WL, Tyagi SC, Roberts AM, Lominadze D. Fibrinogen induces alterations of endothelial cell tight junction proteins. J Cell Physiol 2009; 221:195-203. [PMID: 19507189 DOI: 10.1002/jcp.21845] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
We previously showed that an elevated content of fibrinogen (Fg) increased formation of filamentous actin and enhanced endothelial layer permeability. In the present work we tested the hypothesis that Fg binding to endothelial cells (ECs) alters expression of actin-associated endothelial tight junction proteins (TJP). Rat cardiac microvascular ECs were grown in gold plated chambers of an electrical cell-substrate impedance system, 8-well chambered, or in 12-well plates. Confluent ECs were treated with Fg (2 or 4 mg/ml), Fg (4 mg/ml) with mitogen-activated protein kinase (MEK) kinase inhibitors (PD98059 or U0126), Fg (4 mg/ml) with anti-ICAM-1 antibody or BQ788 (endothelin type B receptor blocker), endothelin-1, endothelin-1 with BQ788, or medium alone for 24 h. Fg induced a dose-dependent decrease in EC junction integrity as determined by transendothelial electrical resistance (TEER). Western blot analysis and RT-PCR data showed that the higher dose of Fg decreased the contents of TJPs, occludin, zona occluden-1 (ZO-1), and zona occluden-2 (ZO-2) in ECs. Fg-induced decreases in contents of the TJPs were blocked by PD98059, U0126, or anti-ICAM-1 antibody. While BQ788 inhibited endothelin-1-induced decrease in TEER, it did not affect Fg-induced decrease in TEER. These data suggest that Fg increases EC layer permeability via the MEK kinase signaling pathway by affecting occludin, ZO-1, and ZO-2, TJPs, which are bound to actin filaments. Therefore, increased binding of Fg to its major EC receptor, ICAM-1, during cardiovascular diseases may increase microvascular permeability by altering the content and possibly subcellular localization of endothelial TJPs.
Collapse
Affiliation(s)
- Phani K Patibandla
- Department of Biochemistry and Molecular Biology, University of Louisville, School of Medicine, Louisville, Kentucky 40202, USA
| | | | | | | | | | | |
Collapse
|
46
|
Bae JJ, Kim JH, Kim H, Kim HS. Expression of Endothelin-1 by Stimulation with CXCL8 in Mouse Peritoneal Macrophages. ACTA ACUST UNITED AC 2009. [DOI: 10.4167/jbv.2009.39.3.205] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- Jei-Jun Bae
- Department of Obstetrics and Gynecology, College of Medicine, Yeungnam University, Daegu, Korea
| | - Jung-Hae Kim
- Department of Microbiology, College of Medicine, Yeungnam University, Daegu, Korea
| | - Hoon Kim
- Department of Emergency Medicine, College of Medicine, Inha University, Incheon, Korea
| | - Hee-Sun Kim
- Department of Microbiology, College of Medicine, Yeungnam University, Daegu, Korea
| |
Collapse
|