1
|
Luo C, He S, Shi F, Zhou J, Shang L. The Role of TRAIL Signaling in Cancer: Searching for New Therapeutic Strategies. BIOLOGY 2024; 13:521. [PMID: 39056714 PMCID: PMC11274015 DOI: 10.3390/biology13070521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024]
Abstract
Cancer continues to pose a significant threat to global health, with its status as a leading cause of death remaining unchallenged. Within the realm of cancer research, the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) stands out as a critical player, having been identified in the 1990s as the tenth member of the TNF family. This review examines the pivotal role of TRAIL in cancer biology, focusing on its ability to induce apoptosis in malignant cells through both endogenous and exogenous pathways. We provide an in-depth analysis of TRAIL's intracellular signaling and intercellular communication, underscoring its potential as a selective anticancer agent. Additionally, the review explores TRAIL's capacity to reshape the tumor microenvironment, thereby influencing cancer progression and response to therapy. With an eye towards future developments, we discuss the prospects of harnessing TRAIL's capabilities for the creation of tailored, precision-based cancer treatments, aiming to enhance efficacy and improve patient survival rates.
Collapse
Affiliation(s)
- Cheng Luo
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Shan He
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Feng Shi
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
- Key Laboratory of Carcinogenesis of National Health Commission, Cancer Research Institute and School of Basic Medical Science, Xiangya School of Medicine, Central South University, Changsha 410078, China
| | - Jianhua Zhou
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| | - Li Shang
- Department of Pathology, National Clinical Research Center for Geriatric Disorders/Xiangya Hospital, Central South University, Changsha 410078, China; (C.L.); (J.Z.)
- Key Laboratory of Carcinogenesis and Cancer Invasion of Chinese Ministry of Education, Xiangya Hospital, Central South University, Changsha 410078, China; (S.H.); (F.S.)
| |
Collapse
|
2
|
Ding R, Huang L, Yan K, Sun Z, Duan J. New insight into air pollution-related cardiovascular disease: an adverse outcome pathway framework of PM2.5-associated vascular calcification. Cardiovasc Res 2024; 120:699-707. [PMID: 38636937 DOI: 10.1093/cvr/cvae082] [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: 11/17/2023] [Revised: 02/15/2024] [Accepted: 02/18/2024] [Indexed: 04/20/2024] Open
Abstract
Despite the air quality has been generally improved in recent years, ambient fine particulate matter (PM2.5), a major contributor to air pollution, remains one of the major threats to public health. Vascular calcification is a systematic pathology associated with an increased risk of cardiovascular disease. Although the epidemiological evidence has uncovered the association between PM2.5 exposure and vascular calcification, little is known about the underlying mechanisms. The adverse outcome pathway (AOP) concept offers a comprehensive interpretation of all of the findings obtained by toxicological and epidemiological studies. In this review, reactive oxygen species generation was identified as the molecular initiating event (MIE), which targeted subsequent key events (KEs) such as oxidative stress, inflammation, endoplasmic reticulum stress, and autophagy, from the cellular to the tissue/organ level. These KEs eventually led to the adverse outcome, namely increased incidence of vascular calcification and atherosclerosis morbidity. To the best of our knowledge, this is the first AOP framework devoted to PM2.5-associated vascular calcification, which benefits future investigations by identifying current limitations and latent biomarkers.
Collapse
Affiliation(s)
- Ruiyang Ding
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Linyuan Huang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Kanglin Yan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| | - Junchao Duan
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, No. 10 Xitoutiao, You'anmen Wai, Fengtai District, Beijing 100069, PR China
| |
Collapse
|
3
|
Didier R, Garnier L, Duloquin G, Meloux A, Sagnard A, Graber M, Dogon G, Benali K, Pommier T, Laurent G, Vergely C, Bejot Y, Guenancia C. Distribution of atrial cardiomyopathy markers and association with atrial fibrillation detected after ischaemic stroke in the SAFAS study. Stroke Vasc Neurol 2024; 9:165-173. [PMID: 37429637 PMCID: PMC11103154 DOI: 10.1136/svn-2023-002447] [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: 03/06/2023] [Accepted: 06/27/2023] [Indexed: 07/12/2023] Open
Abstract
BACKGROUND Atrial cardiomyopathy (AC) is an emerging concept explaining the pathophysiology of cardioembolic strokes in absence of atrial fibrillation (AF). A definition based on the presence of electrical abnormality (P-wave terminal force in lead V1 (PTFV1) >5000 µV×ms), N-Terminal pro-B-type natriuretic peptide (NT pro BNP) >250 pg/mL and/or indexed left atrial diameter (LADI) >3 cm/m² is currently tested in the ARCADIA (AtRial Cardiopathy and Antithrombotic Drugs In prevention After cryptogenic stroke) trial. We set out to estimate the prevalence of AC as defined in the ARCADIA trial, its determinants and its association with AF detected after stroke (AFDAS). METHODS Stepwise screening for silent Atrial Fibrillation After Stroke (SAFAS) study prospectively included 240 ischaemic stroke patients. AC markers were complete for 192 of them and 9 were not included in this analysis because AF had been diagnosed on admission. RESULTS A total of 183 patients were analysed, of whom 57% (104 patients) met the AC criteria (79 NT-proBNP, 47 PTFV1, 4 LADI). In the multivariate logistic regression, C reactive protein >3 mg/L (OR (95% CI) 2.60 (1.30 to 5.21), p=0.007) and age (OR (95% CI) 1.07 (1.04 to 1.10), p<0.001) were found to be independently associated with AC. After 6 months of follow-up, AFDAS was detected in 33% of AC patients and in 14% of the remaining ones (p=0.003). However, AC was not independently associated with AFDAS, contrary to left atrial volume index (>34 mL/m2, OR 2.35 (CI 1.09 to 5.06) p=0029). CONCLUSION AC as defined in ARCADIA is mostly based on NT pro BNP elevation (76% of patients) and is associated with age and inflammation. Moreover, AC was not independently associated with AFDAS at follow-up. The ARCADIA trial, which compares aspirin to apixaban in patients with embolic strokes of undetermined source with AC markers and must, therefore be analysed in the light of these limitations. TRIAL REGISTRATION NUMBER NCT03570060.
Collapse
Affiliation(s)
| | - Lucie Garnier
- Dijon Stroke Registry, Department of Neurology, University Hospital Centre Dijon, Dijon, France
| | - Gauthier Duloquin
- Dijon Stroke Registry, Department of Neurology, University Hospital Centre Dijon, Dijon, France
| | | | | | - Mathilde Graber
- Dijon Stroke Registry, Department of Neurology, University Hospital Centre Dijon, Dijon, France
| | | | - Karim Benali
- Cardiology, CHU Saint Etienne, Saint Etienne, France
| | - Thibaut Pommier
- Cardiology, CHU Dijon Bourgogne, Dijon, France
- PEC 2, Université de Bourgogne, Dijon, France
| | - Gabriel Laurent
- Cardiology, CHU Dijon Bourgogne, Dijon, France
- PEC 2, Université de Bourgogne, Dijon, France
| | | | - Yannick Bejot
- Dijon Stroke Registry, Department of Neurology, University Hospital Centre Dijon, Dijon, France
- PEC 2, Université de Bourgogne, Dijon, France
| | - Charles Guenancia
- Cardiology, CHU Dijon Bourgogne, Dijon, France
- PEC 2, Université de Bourgogne, Dijon, France
| |
Collapse
|
4
|
Suh SH, Oh TR, Choi HS, Kim CS, Bae EH, Ma SK, Oh KH, Lee KB, Jeong JC, Jung JY, Kim SW. Circulating osteoprotegerin levels and cardiovascular outcomes in patients with pre-dialysis chronic kidney disease: results from the KNOW-CKD study. Sci Rep 2024; 14:4136. [PMID: 38374135 PMCID: PMC10876961 DOI: 10.1038/s41598-024-54335-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 02/12/2024] [Indexed: 02/21/2024] Open
Abstract
While the relationship between circulating osteoprotegerin (OPG) and cardiovascular events is well-established in the general population, its association with cardiovascular risks in chronic kidney disease (CKD) patients remains less robust. This study hypothesized that elevated circulating OPG levels might be associated with an increased risk of major adverse cardiac events (MACE) in CKD patients, a total of 2,109 patients with CKD stages 1 through pre-dialysis 5 from the KNOW-CKD cohort were categorized into quartiles based on serum OPG levels. The primary outcome of the study was 3-point MACE, defined as a composite of nonfatal myocardial infarction, nonfatal stroke, or cardiac death. The median follow-up duration was 7.9 years. The cumulative incidence of 3-point MACE significantly varied across serum OPG levels in Kaplan-Meier curve analysis (P < 0.001, log-rank test), with the highest incidence observed in the 4th quartile. Cox regression analysis indicated that, relative to the 1st quartile, the risk of 3-point MACE was significantly higher in the 3rd (adjusted hazard ratio 2.901, 95% confidence interval 1.009 to 8.341) and the 4th quartiles (adjusted hazard ratio 4.347, 95% confidence interval 1.410 to 13.395). In conclusion, elevated circulating OPG levels are associated with adverse cardiovascular outcomes in pre-dialysis CKD patients.
Collapse
Affiliation(s)
- Sang Heon Suh
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Tae Ryom Oh
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Hong Sang Choi
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Chang Seong Kim
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Eun Hui Bae
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Seong Kwon Ma
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea
| | - Kook-Hwan Oh
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea
| | - Kyu-Beck Lee
- Division of Nephrology, Department of Internal Medicine, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Jong Cheol Jeong
- Division of Nephrology, Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Ji Yong Jung
- Division of Nephrology, Department of Internal Medicine, Gachon University Gil Medical Center, Incheon, Republic of Korea
| | - Soo Wan Kim
- Department of Internal Medicine, Chonnam National University Medical School and Chonnam National University Hospital, 42 Jebongro, Gwangju, 61469, Korea.
| |
Collapse
|
5
|
Nakamizo T, Cologne J, Kishi T, Takahashi T, Inoue M, Ryukaku H, Hayashi T, Kusunoki Y, Fujiwara S, Ohishi W. Reliability, stability during long-term storage, and intra-individual variation of circulating levels of osteopontin, osteoprotegerin, vascular endothelial growth factor-A, and interleukin-17A. Eur J Med Res 2024; 29:133. [PMID: 38368424 PMCID: PMC10873926 DOI: 10.1186/s40001-024-01722-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 02/09/2024] [Indexed: 02/19/2024] Open
Abstract
BACKGROUND Studies in many populations have reported associations between circulating cytokine levels and various physiological or pathological conditions. However, the reliability of cytokine measurements in population studies, which measure cytokines in multiple assays over a prolonged period, has not been adequately examined; nor has stability during sample storage or intra-individual variation been assessed. METHODS We assessed (1) analytical reliability in short- and long-term repeated measurements; (2) stability and analytical reliability during long-term sample storage, and (3) variability within individuals over seasons, of four cytokines-osteopontin (OPN), osteoprotegerin (OPG), vascular endothelial growth factor-A (VEGF-A), and interleukin-17A (IL-17A). Measurements in plasma or serum samples were made with commercial kits according to standard procedures. Estimation was performed by fitting a random or mixed effects linear model on the log scale. RESULTS In repeated assays over a short period, OPN, OPG, and VEGF-A had acceptable reliability, with intra- and inter-assay coefficients of variation (CV) less than 0.11. Reliability of IL-17A was poor, with inter- and intra-assay CV 0.85 and 0.43, respectively. During long-term storage, OPG significantly decayed (- 33% per year; 95% confidence interval [- 54, - 3.7]), but not OPN or VEGF-A (- 0.3% or - 6.3% per year, respectively). Intra- and inter-assay CV over a long period were comparable to that in a short period except for a slight increase in inter-assay CV of VEGF-A. Within-individual variation was small for OPN and VEGF-A, with intra-class correlations (ICC) 0.68 and 0.83, respectively, but large for OPG (ICC 0.11). CONCLUSIONS We conclude that OPN and VEGF-A can be reliably measured in a large population, that IL-17A is suitable only for small experiments, and that OPG should be assessed with caution due to degradation during storage and intra-individual variation. The overall results of our study illustrate the need for validation under relevant conditions when measuring circulating cytokines in population studies.
Collapse
Affiliation(s)
| | | | - Takeshi Kishi
- Division of Clinical Laboratories, RERF, Hiroshima, Japan
| | - Tetsuya Takahashi
- Faculty of Rehabilitation, Hiroshima International University, Hiroshima, Japan
| | - Mayumi Inoue
- Division of Clinical Laboratories, RERF, Hiroshima, Japan
| | | | | | | | - Saeko Fujiwara
- Faculty of Pharmacy, Yasuda Women's University, Hiroshima, Japan
| | - Waka Ohishi
- Department of Clinical Studies, RERF, Hiroshima, Japan
| |
Collapse
|
6
|
Chen YL, Huang PY, Tsai JP, Wang JH, Hsu BG. Serum Osteoprotegerin Levels and the Vascular Reactivity Index in Patients with Hypertension. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1794. [PMID: 37893512 PMCID: PMC10608475 DOI: 10.3390/medicina59101794] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 09/28/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023]
Abstract
Background and Objectives: Osteoprotegerin (OPG), a soluble glycoprotein found in serum, has been associated with both the presence and severity of atherosclerosis. OPG is regarded as the mediator in the process of vascular endothelial dysfunction. Impaired endothelial function has an intimate link with hypertension (HTN) and is associated with significant morbidity and mortality. This study was to investigate the connection between OPG and endothelial dysfunction in patients having HTN. Materials and Methods: There are 102 patients with HTN included. For the purpose of determining the levels of OPG, a commercial enzyme-linked immunosorbent test kit was applied. The vascular reactivity index (VRI), which is assessed via the digital thermal monitoring, provides information on endothelial function. Results: Ten patients with HTN (9.8%) were classified as having poor vascular reactivity (VRI < 1.0), 46 HTN patients (45.1%) as having intermediate vascular reactivity (1.0 ≤ VRI < 2.0), and 46 HTN patients (45.1%) were classified as having high vascular reactivity (VRI ≥ 2.0). A greater serum OPG level (p < 0.001) and older age (p = 0.022) were linked to impaired vascular reactivity. The estimated glomerular filtration rate (r = 0.196, p = 0.048) was positively correlated with VRI values in hypertensive participants, while advanced age (r = -0.222, p = 0.025) and the log-transformed OPG level (log-OPG, r = -0.357, p < 0.001) were negatively correlated with VRI. Serum log-OPG level was shown to be strongly and independently correlated with VRI values in HTN individuals after multivariable forward stepwise linear regression analysis (β = -0.357, adjusted R2 change = 0.119, p < 0.001). Conclusions: In patients with HTN, serum OPG levels were adversely correlated with VRI and probably had a role in endothelial dysfunction.
Collapse
Affiliation(s)
- Yen-Liang Chen
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
| | - Po-Yu Huang
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan; (P.-Y.H.); (J.-P.T.)
| | - Jen-Pi Tsai
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan; (P.-Y.H.); (J.-P.T.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Ji-Hung Wang
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Cardiology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| | - Bang-Gee Hsu
- Department of Internal Medicine, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan;
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan
| |
Collapse
|
7
|
Sayed SZ, Abd El-Hafez AH, Abu El-Ela MA, Mourad MAF, Mousa SO. OPG/RANK/RANKL axis relation to cardiac iron-overload in children with transfusion-dependent thalassemia. Sci Rep 2023; 13:12568. [PMID: 37532711 PMCID: PMC10397306 DOI: 10.1038/s41598-023-39596-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/27/2023] [Indexed: 08/04/2023] Open
Abstract
OPG/RANK/RANKL axis was reportedly involved in initiating various diseases, especially bone and cardiovascular diseases. This study aimed to assess the relationship between some OPG, RANK, and RANKL polymorphisms and alleles and iron-overload-induced cardiomyopathy in children with transfusion-dependent thalassemia (TDT). This study included 80 TDT children and 80 age and sex-matched controls. Real-time PCR was done for rs207318 polymorphism for the OPG gene and rs1805034, rs1245811, and rs75404003 polymorphisms for the RANK gene, and rs9594782 and rs2277438 polymorphisms for the RANKL gene. Cardiac T2* MRI and ejection fraction (EF) were done to assess the myocardial iron status and cardiac function. In this study, there were no significant differences in frequencies of the studied polymorphisms between cases and controls (p > 0.05 in all). In TDT children, OPG rs2073618 (G > C) had a significant relation to myocardial iron overload (p = 0.02). Its C allele had significantly more frequent normal EF than its G allele (p = 0.04). RANK rs75404403 (C > DEL) had a significant relation to cardiac dysfunction (p = 0.02). Moreover, the C allele of that gene had significantly more frequent affected EF than its DEL allele (p = 0.02). The A allele of RANKL rs2277438 (G > A) had significantly less frequent severe cardiac iron overload than the G allele (p = 0.04). In conclusion, the OPG/ RANK/RANKL genes may act as genetic markers for iron-induced cardiomyopathy in TDT children. Some of the studied genes' polymorphisms and alleles were significantly related to myocardial iron overload and cardiac dysfunction in TDT children.
Collapse
Affiliation(s)
- Samira Zein Sayed
- Department of Pediatrics, Faculty of Medicine, Minia University, El Minya, Egypt
| | | | | | | | - Suzan Omar Mousa
- Department of Pediatrics, Faculty of Medicine, Minia University, El Minya, Egypt.
| |
Collapse
|
8
|
Meier C, Eastell R, Pierroz DD, Lane NE, Al-Daghri N, Suzuki A, Napoli N, Mithal A, Chakhtoura M, Fuleihan GEH, Ferrari S. Biochemical Markers of Bone Fragility in Patients with Diabetes. A Narrative Review by the IOF and the ECTS. J Clin Endocrinol Metab 2023; 108:dgad255. [PMID: 37155585 PMCID: PMC10505554 DOI: 10.1210/clinem/dgad255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 04/24/2023] [Accepted: 05/03/2023] [Indexed: 05/10/2023]
Abstract
CONTEXT The risk of fragility fractures is increased in both type 1 and type 2 diabetes. Numerous biochemical markers reflecting bone and/or glucose metabolism have been evaluated in this context. This review summarizes current data on biochemical markers in relation to bone fragility and fracture risk in diabetes. METHODS Literature review by a group of experts from the International Osteoporosis Foundation (IOF) and European Calcified Tissue Society (ECTS) focusing on biochemical markers, diabetes, diabetes treatments and bone in adults. RESULTS Although bone resorption and bone formation markers are low and poorly predictive of fracture risk in diabetes, osteoporosis drugs seem to change bone turnover markers in diabetics similarly to non-diabetics, with similar reductions in fracture risk. Several other biochemical markers related to bone and glucose metabolism have been correlated with BMD and/or fracture risk in diabetes, including osteocyte-related markers such as sclerostin, HbA1c and advanced glycation end products (AGEs), inflammatory markers and adipokines, as well as IGF-1 and calciotropic hormones. CONCLUSION Several biochemical markers and hormonal levels related to bone and/or glucose metabolism have been associated with skeletal parameters in diabetes. Currently, only HbA1c levels seem to provide a reliable estimate of fracture risk, while bone turnover markers could be used to monitor the effects of anti-osteoporosis therapy.
Collapse
Affiliation(s)
- Christian Meier
- Department of Endocrinology, Diabetology and Metabolism, University Hospital Basel, 4031 Basel, Switzerland
| | - Richard Eastell
- Academic Unit of Bone Metabolism, Mellanby Centre for Bone Research, University of Sheffield, S57AU Sheffield, UK
| | | | - Nancy E Lane
- Department of Medicine and Rheumatology, Davis School of Medicine, University of California, Sacramento, CA 95817, USA
| | - Nasser Al-Daghri
- Department of Biochemistry, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
| | - Atsushi Suzuki
- Department of Endocrinology, Diabetes and Metabolism, Fujita Health University, Toyoake, Aichi 470-1192, Japan
| | - Nicola Napoli
- Unit of Endocrinology and Diabetes, Department of Medicine, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Ambrish Mithal
- Institute of Diabetes and Endocrinology, Max Healthcare, Saket, New Delhi 110017, India
| | - Marlene Chakhtoura
- Department of Internal Medicine, Division of Endocrinology, Calcium Metabolism and Osteoporosis Program, WHO Collaborating Center for Metabolic Bone Disorders, American University of Beirut Medical Center, Riad El Solh, Beirut 6044, Lebanon
| | - Ghada El-Hajj Fuleihan
- Department of Internal Medicine, Division of Endocrinology, Calcium Metabolism and Osteoporosis Program, WHO Collaborating Center for Metabolic Bone Disorders, American University of Beirut Medical Center, Riad El Solh, Beirut 6044, Lebanon
| | - Serge Ferrari
- Service and Laboratory of Bone Diseases, Geneva University Hospital and Faculty of Medicine, 1205 Geneva, Switzerland
| |
Collapse
|
9
|
Monir D, Osama A, Saad AE, Negm M, Abd El-Razek R. Role of osteoprotegerin rs3102735 gene polymorphism in acute ischemic stroke patients. THE EGYPTIAN JOURNAL OF NEUROLOGY, PSYCHIATRY AND NEUROSURGERY 2023; 59:49. [DOI: 10.1186/s41983-023-00652-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 04/01/2023] [Indexed: 09/02/2023] Open
Abstract
Abstract
Background
Ischemic stroke ranks third among leading causes of death and disability. Both endothelial and vascular smooth muscle cells generate osteoprotegerin (OPG). Ischemic stroke and its severity may be enhanced by the OPG rs3102735 gene polymorphism. Our research aims to investigate OPG rs3102735 gene polymorphism role in ischemic stroke risk and to assess its association with stroke severity at presentation and degree of vascular stenosis and evaluate its potential as a predictor of stroke severity. Fifty people with acute ischemic stroke as well as fifty controls were studied. The NIHSS and ASPECTS were utilized to evaluate stroke severity and the infarction size, respectively. All subjects underwent extracranial carotid duplex study and molecular assessment for genotyping of OPG rs3102735) gene polymorphism.
Results
Stroke patients had markedly higher concentrations of OPG in the plasma than controls (311.60 ± 109.48 versus 240.20 ± 75.96 mmol/ml, p = 0.001). The optimal plasma OPG cutoff value for the predicting the occurrence of stroke was determined to be > 250 mmol/ml, the 95% confidence interval (CI) was (0.625–0.843), sensitivity was 68% and specificity was 72%. Ischemic stroke had a significantly different genotype distribution for the OPG rs3102735 gene polymorphism than did controls (36 CC, 13 CT, and 1 TT) versus (28 CC, 15 CT, and 7 TT) respectively. Stroke patients had a significantly greater CC + CT genotype than controls did (P = 0.041), also they had a higher propensity for carrying the C allele than the T allele (P = 0.017). Carotid intima medium thickness and the NIHSS both had positive correlations with OPG serum level (r = 0.39, p = 0.02 and r = 0.4, p = 0.02, respectively), whereas ASPECTS had an inversed correlation (r = − 0.65, p = 0.001).
Conclusions
The current study shows that as an independent risk factor, increased plasma OPG level, may participate in the atherothrombotic ischemic stroke pathophysiology, in addition, genetic variants in the OPG gene (rs3102735) are a separate risk factor for large artery atherosclerosis and plasma OPG level can serve as a biomarker to determine the severity of a stroke.
Collapse
|
10
|
Differential angiogenesis of bone and muscle endothelium in aging and inflammatory processes. Commun Biol 2023; 6:126. [PMID: 36721025 PMCID: PMC9889796 DOI: 10.1038/s42003-023-04515-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 01/20/2023] [Indexed: 02/01/2023] Open
Abstract
Different tissues have different endothelial features, however, the implications of this heterogeneity in pathological responses are not clear yet. "Inflamm-aging" has been hypothesized as a possible trigger of diseases, including osteoarthritis (OA) and sarcopenia, often present in the same patient. To highlight a possible contribution of organ-specific endothelial cells (ECs), we compare ECs derived from bone and skeletal muscle of the same OA patients. OA bone ECs show a pro-inflammatory signature and higher angiogenic sprouting as compared to muscle ECs, in control conditions and stimulated with TNFα. Furthermore, growth of muscle but not bone ECs decreases with increasing patient age and systemic inflammation. Overall, our data demonstrate that inflammatory conditions in OA patients differently affect bone and muscle ECs, suggesting that inflammatory processes increase angiogenesis in subchondral bone while associated systemic low-grade inflammation impairs angiogenesis in muscle, possibly highlighting a vascular trigger linking OA and sarcopenia.
Collapse
|
11
|
Vaspin alleviates the lncRNA LEF1-AS1-induced osteogenic differentiation of vascular smooth muscle cells via the Hippo/YAP signaling pathway. Exp Cell Res 2022; 421:113407. [PMID: 36334793 DOI: 10.1016/j.yexcr.2022.113407] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/26/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022]
Abstract
Vascular calcification (VC) is closely related to higher cardiovascular mortality and morbidity, and vascular smooth muscle cell (VSMC) switching to osteogenic-like cells is crucial for VC. LncRNA LEF1-AS1 promotes atherosclerosis and dental pulp stem cells calcification, while its role in VC remains unknown. Visceral adipose tissue-derived serine protease inhibitor (vaspin) is an adipokine regulating bone metabolism. However, the relationship between vaspin and VC is still unclear. We aimed to explore the role of LEF1-AS1 on VSMC osteogenic transition, whether vaspin inhibited LEF1-AS1-mediated osteogenic differentiation of VSMCs, and the responsible mechanism. In this study, quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting analysis indicated that LEF1-AS1 overexpression significantly upregulated osteogenic marker Runt-related transcription factor-2 (RUNX2) level and downregulated VSMC contractile marker α-smooth muscle actin (α-SMA) level. Alizarin red staining, alkaline phosphatase (ALP) staining, ALP activity assay, and calcium content assay also suggested that LEF1-AS1 overexpression promoted calcium deposition in VSMCs. However, vaspin treatment abolished this phenomenon. Mechanistically, LEF1-AS1 markedly decreased phosphorylated YAP level, while vaspin reversed LEF1-AS1-induced phosphorylated YAP decline. Our results revealed that LEF1-AS1 accelerated the osteogenic differentiation of VSMCs by regulating the Hippo/YAP pathway, while vaspin eliminated the LEF1-AS1-meditated VSMCs osteogenic phenotype switch.
Collapse
|
12
|
Romejko K, Rymarz A, Szamotulska K, Bartoszewicz Z, Rozmyslowicz T, Niemczyk S. Left Ventricular Diastolic Dysfunction in Chronic Kidney Disease Patients Not Treated with Dialysis. Nutrients 2022; 14:4664. [PMID: 36364925 PMCID: PMC9655426 DOI: 10.3390/nu14214664] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 11/25/2023] Open
Abstract
BACKGROUND Left ventricular diastolic dysfunction (LVDD) is observed in the early stages of chronic kidney disease (CKD) and may lead to heart failure with preserved ejection fraction (HFpEF). The purpose of our study was to investigate the association between metabolic, nutritional and inflammatory parameters and LVDD in CKD and non-CKD patients. METHODS Two groups of patients were recruited to the study: 93 men with CKD and eGFR lower than 60 mL/min/1.73 m2 and 40 men without kidney function decrease with eGFR ≥ 60 mL/min/1.73 m2. Transthoracic echocardiography was performed to evaluate the diastolic function of the left ventricle. Bioimpedance spectroscopy (BIS) was used to measure overhydration and lean body mass. We also measured the serum concentrations of albumin, glucose, haemoglobin A1c (HgbA1c), fibrinogen, C-reactive protein (CRP), tumor necrosis factor-alpha (TNF-alpha) and osteoprotegerin (OPG). RESULTS We observed that elevated serum fibrinogen and glucose concentrations were associated with LVDD independently of CKD status. Serum fibrinogen concentrations increased with the advancement of LVDD. Low albumin concentrations in CKD were related with LVDD. In the control group, lower muscle mass presented as lean tissue index (LTI) and lean tissue mass (LTM), and overhydration were associated with LVDD. In the group of patients without kidney function decrease the OPG concentrations were significantly higher in those with LVDD, and they rose with the advancement of LVDD. CONCLUSIONS Elevated inflammatory parameters, increased serum glucose concentrations and worse nutritional status are the states that may impair the diastolic function of the left ventricle in CKD and non-CKD patients. Serum OPG levels are elevated in patients without kidney function decrease and LVDD and its concentrations rise with the advancement of LVDD.
Collapse
Affiliation(s)
- Katarzyna Romejko
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Aleksandra Rymarz
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Katarzyna Szamotulska
- Department of Epidemiology and Biostatistics, Institute of Mother and Child, Kasprzaka Street, 17a, 01-211 Warsaw, Poland
| | - Zbigniew Bartoszewicz
- Department of Internal Diseases and Endocrinology, Medical University of Warsaw, 1a Banacha Street, 02-097 Warsaw, Poland
| | - Tomasz Rozmyslowicz
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, R.217 John Morgan Building, 3620 Hamilton Walk, Philadelphia, PA 19104, USA
| | - Stanisław Niemczyk
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland
| |
Collapse
|
13
|
Zhang L, Zeng F, Jiang M, Han M, Huang B. Roles of osteoprotegerin in endocrine and metabolic disorders through receptor activator of nuclear factor kappa-B ligand/receptor activator of nuclear factor kappa-B signaling. Front Cell Dev Biol 2022; 10:1005681. [PMID: 36407115 PMCID: PMC9671468 DOI: 10.3389/fcell.2022.1005681] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 10/17/2022] [Indexed: 11/21/2023] Open
Abstract
Endocrine and metabolic diseases show increasing incidence and high treatment costs worldwide. Due to the complexity of their etiology and mechanism, therapeutic strategies are still lacking. Osteoprotegerin (OPG), a member of the tumor necrosis factor receptor superfamily, appears to be a potential candidate for the treatment of these diseases. Studies based on clinical analysis and rodent animal models reveal the roles of OPG in various endocrine and metabolic processes or disorders, such as bone remodeling, vascular calcification, and β-cell proliferation, through the receptor activator of nuclear factor kappa-B ligand (RANKL) and the receptor activator of NF-κB (RANK). Thus, in this review, we mainly focus on relevant diseases, including osteoporosis, cardiovascular disease (CVD), diabetes, and gestational diabetes mellitus (GDM), to summarize the effects of the RANKL/RANK/OPG system in endocrine and metabolic tissues and diseases, thereby providing a comprehensive insight into OPG as a potential drug for endocrine and metabolic diseases.
Collapse
Affiliation(s)
- Luodan Zhang
- Department of Nephrology, Anhui Provincial Children’s Hospital, Children’s Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Fa Zeng
- Shenzhen Longhua Maternity and Child Healthcare Hospital, Shenzhen, Guangdong, China
| | - Minmin Jiang
- Department of Occupational Health and Environmental Health, School of Public Health, Anhui Medical University, Hefei, Anhui, China
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui, China
| | - Maozhen Han
- College of Life Science, Anhui Medical University, Hefei, Anhui, China
| | - Binbin Huang
- Department of Maternal, Child and Adolescent Health, School of Public Health, Anhui Medical University, MOE Key Laboratory of Population Health Across Life Cycle, NHC Key Laboratory of Study on Abnormal Gametes and Reproductive Tract, Anhui Provincial Key Laboratory of Population Health and Aristogenics, Hefei, Anhui, China
| |
Collapse
|
14
|
Zheng G, Ma HW, Xiang GH, He GL, Cai HC, Dai ZH, Chen YL, Lin Y, Xu HZ, Ni WF, Xu C, Liu HX, Wang XY. Bone-targeting delivery of platelet lysate exosomes ameliorates glucocorticoid-induced osteoporosis by enhancing bone-vessel coupling. J Nanobiotechnology 2022; 20:220. [PMID: 36310171 PMCID: PMC9620632 DOI: 10.1186/s12951-022-01400-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 03/26/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Glucocorticoids (GCs) overuse is associated with decreased bone mass and osseous vasculature destruction, leading to severe osteoporosis. Platelet lysates (PL) as a pool of growth factors (GFs) were widely used in local bone repair by its potent pro-regeneration and pro-angiogenesis. However, it is still seldom applied for treating systemic osteopathia due to the lack of a suitable delivery strategy. The non-targeted distribution of GFs might cause tumorigenesis in other organs. RESULTS In this study, PL-derived exosomes (PL-exo) were isolated to enrich the platelet-derived GFs, followed by conjugating with alendronate (ALN) grafted PEGylated phospholipid (DSPE-PEG-ALN) to establish a bone-targeting PL-exo (PL-exo-ALN). The in vitro hydroxyapatite binding affinity and in vivo bone targeting aggregation of PL-exo were significantly enhanced after ALN modification. Besides directly modulating the osteogenic and angiogenic differentiation of bone marrow mesenchymal stem cells (BMSCs) and endothelial progenitor cells (EPCs), respectively, PL-exo-ALN also facilitate their coupling under GCs' stimulation. Additionally, intravenous injection of PL-exo-ALN could successfully rescue GCs induced osteoporosis (GIOP) in vivo. CONCLUSIONS PL-exo-ALN may be utilized as a novel nanoplatform for precise infusion of GFs to bone sites and exerts promising therapeutic potential for GIOP.
Collapse
Affiliation(s)
- Gang Zheng
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Hai-Wei Ma
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Guang-Heng Xiang
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Gao-Lu He
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Han-Chen Cai
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Zi-Han Dai
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Yan-Lin Chen
- Department of Orthopaedic Surgery, Lishui Central Hospital and Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, Zhejiang Province, China
| | - Yan Lin
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Hua-Zi Xu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Wen-Fei Ni
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Cong Xu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Hai-Xiao Liu
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| | - Xiang-Yang Wang
- Key Laboratory of Orthopaedics of Zhejiang Province, Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
- The Second School of Medicine, Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China.
| |
Collapse
|
15
|
Nasiri-Ansari N, Spilioti E, Kyrou I, Kalotychou V, Chatzigeorgiou A, Sanoudou D, Dahlman-Wright K, Randeva HS, Papavassiliou AG, Moutsatsou P, Kassi E. Estrogen Receptor Subtypes Elicit a Distinct Gene Expression Profile of Endothelial-Derived Factors Implicated in Atherosclerotic Plaque Vulnerability. Int J Mol Sci 2022; 23:ijms231810960. [PMID: 36142876 PMCID: PMC9506323 DOI: 10.3390/ijms231810960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/21/2022] Open
Abstract
In the presence of established atherosclerosis, estrogens are potentially harmful. MMP-2 and MMP-9, their inhibitors (TIMP-2 and TIMP-1), RANK, RANKL, OPG, MCP-1, lysyl oxidase (LOX), PDGF-β, and ADAMTS-4 play critical roles in plaque instability/rupture. We aimed to investigate (i) the effect of estradiol on the expression of the abovementioned molecules in endothelial cells, (ii) which type(s) of estrogen receptors mediate these effects, and (iii) the role of p21 in the estrogen-mediated regulation of the aforementioned factors. Human aortic endothelial cells (HAECs) were cultured with estradiol in the presence or absence of TNF-α. The expression of the aforementioned molecules was assessed by qRT-PCR and ELISA. Zymography was also performed. The experiments were repeated in either ERα- or ERβ-transfected HAECs and after silencing p21. HAECs expressed only the GPR-30 estrogen receptor. Estradiol, at low concentrations, decreased MMP-2 activity by 15-fold, increased LOX expression by 2-fold via GPR-30, and reduced MCP-1 expression by 3.5-fold via ERβ. The overexpression of ERα increased MCP-1 mRNA expression by 2.5-fold. In a low-grade inflammation state, lower concentrations of estradiol induced the mRNA expression of MCP-1 (3.4-fold) and MMP-9 (7.5-fold) and increased the activity of MMP-2 (1.7-fold) via GPR-30. Moreover, p21 silencing resulted in equivocal effects on the expression of the abovementioned molecules. Estradiol induced different effects regarding atherogenic plaque instability through different ERs. The balance of the expression of the various ER subtypes may play an important role in the paradoxical characterization of estrogens as both beneficial and harmful.
Collapse
Affiliation(s)
- Narjes Nasiri-Ansari
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eliana Spilioti
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Laboratory of Toxicological Control of Pesticides, Scientific Directorate of Pesticides’ Control and Phytopharmacy, Benaki Phytopathological Institute, 14561 Athens, Greece
| | - Ioannis Kyrou
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
- Laboratory of Dietetics and Quality of Life, Department of Food Science and Human Nutrition, School of Food and Nutritional Sciences, Agricultural University of Athens, 11855 Athens, Greece
- Centre for Sport, Exercise and Life Sciences, Research Institute for Health & Wellbeing, Coventry University, Coventry CV1 5FB, UK
| | - Vassiliki Kalotychou
- Department of Internal Medicine, Laikon General Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Despina Sanoudou
- Clinical Genomics and Pharmacogenomics Unit, 4th Department of Internal Medicine, Attikon Hospital Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece
| | - Karin Dahlman-Wright
- Department of Biosciences and Nutrition, Novum, Karolinska Institute, SE-14183 Huddinge, Sweden
| | - Harpal S. Randeva
- Warwickshire Institute for the Study of Diabetes, Endocrinology and Metabolism (WISDEM), University Hospitals Coventry and Warwickshire NHS Trust, Coventry CV2 2DX, UK
- Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK
| | - Athanasios G. Papavassiliou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Paraskevi Moutsatsou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eva Kassi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Endocrine Unit, 1st Department of Propaedeutic Internal Medicine, Laiko General Hospital, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Correspondence: ; Tel.: +30-21-0746-2699; Fax: +30-21-0746-2703
| |
Collapse
|
16
|
Critical illness and bone metabolism: where are we now and what is next? Eur J Med Res 2022; 27:177. [PMID: 36104724 PMCID: PMC9472372 DOI: 10.1186/s40001-022-00805-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 09/02/2022] [Indexed: 11/10/2022] Open
Abstract
AbstractCritical illness refers to the clinical signs of severe, variable and life-threatening critical conditions, often accompanied by insufficiency or failure of one or more organs. Bone health of critically ill patients is severely affected during and after ICU admission. Therefore, clinical work should focus on ICU-related bone loss, and early development and implementation of related prevention and treatment strategies: optimized and personalized nutritional support (high-quality protein, trace elements and intestinal prebiotics) and appropriate physiotherapy and muscle training should be implemented as early as possible after ICU admission and discharge. At the same time, the drug regulates excessive metabolism and resists osteoporosis.
Collapse
|
17
|
Mousa SO, Abd El-Hafez AH, Abu El-Ela MA, Mourad MAF, Saleh RN, Sayed SZ. RANK/RANKL/OPG axis genes relation to cognitive impairment in children with transfusion-dependent thalassemia: a cross-sectional study. BMC Pediatr 2022; 22:435. [PMID: 35858838 PMCID: PMC9297631 DOI: 10.1186/s12887-022-03479-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 07/06/2022] [Indexed: 01/19/2023] Open
Abstract
Background RANK/RANKL/OPG axis was implicated in many pathological conditions. The study aimed to assess the relationship between the studied RANK, RANKL, and OPG polymorphisms and alleles and cognitive impairment in children with transfusion-dependent thalassemia (TDT). Methods This study included 60 TDT children. Real-time PCR was done for: rs1805034, rs1245811, and rs75404003 polymorphisms for the RANK gene, rs9594782 and rs2277438 polymorphisms for the RANKL gene, and rs207318 polymorphism for the OPG gene. The intelligence quotient (IQ) was assessed using the Wechsler Intelligence Scale for Children-Third Edition. Results TDT children had a low average total IQ, verbal IQ, and borderline performance IQ. RANK rs1805034 (C > T) had a significant effect on total IQ (p = 0.03). Its TT polymorphism and the CT polymorphism of RANKL rs9494782 (C > T) had a significantly lower total IQ (p = 0.01 for both). The G allele of the RANKL rs2277438 (G > A) had a significantly lower total IQ (p = 0.02). RANK rs1805034 (C > T) and RANKL rs2277438 (G > A) significantly affected verbal IQ (p = 0.01 and 0.03). TT genotype of RANK rs1805034 (C > T) had significantly lower verbal IQ (p = 0.002). Furthermore, the GG genotype of RANKL rs2277438 (G > A) had a significantly lower verbal and performance IQ than the AA genotype (p = 0.04 and 0.01 respectively), and its G allele had a significantly lower performance IQ than the A allele (p = 0.02). Conclusion TDT children had low average total and verbal IQ while their performance IQ was borderline. The RANK/RANKL/OPG pathway affects cognition in TDT children, as some of the studied genes’ polymorphisms and alleles had significant effects on total, verbal, and performance IQ of the studied TDT children.
Collapse
Affiliation(s)
- Suzan Omar Mousa
- Department of Pediatrics, Children's University hospital, Faculty of Medicine, Minia University, El-Minya, Egypt.
| | - Asmaa Hosni Abd El-Hafez
- Department of Pediatrics, Children's University hospital, Faculty of Medicine, Minia University, El-Minya, Egypt
| | | | | | - Rasha Nady Saleh
- Department of Neuropsychiatry, Faculty of Medicine, El-Minya, Egypt
| | - Samira Zain Sayed
- Department of Pediatrics, Children's University hospital, Faculty of Medicine, Minia University, El-Minya, Egypt
| |
Collapse
|
18
|
Garnier L, Duloquin G, Meloux A, Benali K, Sagnard A, Graber M, Dogon G, Didier R, Pommier T, Vergely C, Béjot Y, Guenancia C. Multimodal Approach for the Prediction of Atrial Fibrillation Detected After Stroke: SAFAS Study. Front Cardiovasc Med 2022; 9:949213. [PMID: 35911547 PMCID: PMC9326228 DOI: 10.3389/fcvm.2022.949213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/20/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundIntensive screening for atrial fibrillation (AF) has led to a better recognition of this cause in stroke patients. However, it is currently debated whether AF Detected After Stroke (AFDAS) has the same pathophysiology and embolic risk as prior-to-stroke AF. We thus aimed to systematically approach AFDAS using a multimodal approach combining clinical, imaging, biological and electrocardiographic markers.MethodsPatients without previously known AF admitted to the Dijon University Hospital (France) stroke unit for acute ischemic stroke were prospectively enrolled. The primary endpoint was the presence of AFDAS at 6 months, diagnosed through admission ECG, continuous electrocardiographic monitoring, long-term external Holter during the hospital stay, or implantable cardiac monitor if clinically indicated after discharge.ResultsOf the 240 included patients, 77 (32%) developed AFDAS. Compared with sinus rhythm patients, those developing AFDAS were older, more often women and less often active smokers. AFDAS patients had higher blood levels of NT-proBNP, osteoprotegerin, galectin-3, GDF-15 and ST2, as well as increased left atrial indexed volume and lower left ventricular ejection fraction. After multivariable analysis, galectin-3 ≧ 9 ng/ml [OR 3.10; 95% CI (1.03–9.254), p = 0.042], NT-proBNP ≧ 290 pg/ml [OR 3.950; 95% CI (1.754–8.892, p = 0.001], OPG ≥ 887 pg/ml [OR 2.338; 95% CI (1.015–5.620), p = 0.046) and LAVI ≥ 33.5 ml/m2 [OR 2.982; 95% CI (1.342–6.625), p = 0.007] were independently associated with AFDAS.ConclusionA multimodal approach combining imaging, electrocardiography and original biological markers resulted in good predictive models for AFDAS. These results also suggest that AFDAS is probably related to an underlying atrial cardiopathy.Clinical Trial Registration[www.ClinicalTrials.gov], identifier [NCT03570060].
Collapse
Affiliation(s)
- Lucie Garnier
- Department of Neurology, University Hospital, Dijon, France
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
| | - Gauthier Duloquin
- Department of Neurology, University Hospital, Dijon, France
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
| | - Alexandre Meloux
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
| | - Karim Benali
- Department of Cardiology, University Hospital, Dijon, France
| | - Audrey Sagnard
- Department of Cardiology, University Hospital, Dijon, France
| | - Mathilde Graber
- Department of Neurology, University Hospital, Dijon, France
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
| | - Geoffrey Dogon
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
| | - Romain Didier
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
- Department of Cardiology, University Hospital, Dijon, France
| | - Thibaut Pommier
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
- Department of Cardiology, University Hospital, Dijon, France
| | - Catherine Vergely
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
| | - Yannick Béjot
- Department of Neurology, University Hospital, Dijon, France
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
| | - Charles Guenancia
- Pathophysiology and Epidemiology of Cerebro-Cardiovascular Diseases (EA 7460), Faculty of Health Sciences, Université de Bourgogne, Université de Bourgogne Franche-Comté, Dijon, France
- Department of Cardiology, University Hospital, Dijon, France
- *Correspondence: Charles Guenancia,
| |
Collapse
|
19
|
Di Maggio N, Banfi A. The osteo-angiogenic signaling crosstalk for bone regeneration: harmony out of complexity. Curr Opin Biotechnol 2022; 76:102750. [PMID: 35841865 DOI: 10.1016/j.copbio.2022.102750] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 05/31/2022] [Indexed: 11/03/2022]
Abstract
In recent years it has been increasingly appreciated that blood vessels are not simply suppliers of nutrients and oxygen, but actually play an exquisite regulatory role in bone development and repair. A specialized kind of endothelium, named type H because of its high expression of CD31 and Endomucin, constitutes anatomically defined vessels in proximity of the epiphyseal growth plate. Type H endothelium regulates the proliferation and differentiation of both osteoblasts and osteoclasts through the secretion of angiocrine signals and is a hub for the bidirectional molecular crosstalk between the different cell populations of the osteogenic microenvironment. Type H vessels are a key target for current translational approaches aiming at coupling angiogenesis and osteogenesis for bone repair. Open questions remain about their presence and features in notstereotyped tissues, like engineered osteogenic grafts, and the opportunities for their clinical stimulation by pharmacological treatments.
Collapse
Affiliation(s)
- Nunzia Di Maggio
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland.
| | - Andrea Banfi
- Cell and Gene Therapy, Department of Biomedicine, Basel University Hospital and University of Basel, Basel, Switzerland; Plastic, Reconstructive, Aesthetic and Hand Surgery, Basel University Hospital, Switzerland.
| |
Collapse
|
20
|
Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL): A Novel Biomarker for Prognostic Assessment and Risk Stratification of Acute Pulmonary Embolism. J Clin Med 2022; 11:jcm11133908. [PMID: 35807194 PMCID: PMC9267658 DOI: 10.3390/jcm11133908] [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: 04/10/2022] [Revised: 06/02/2022] [Accepted: 06/27/2022] [Indexed: 12/10/2022] Open
Abstract
Background: Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is associated with poor prognosis in cardiovascular diseases. However, the predictive value of TRAIL for the short-term outcome and risk stratification of acute pulmonary embolism (PE) remains unknown. Methods: This study prospectively included 151 normotensive patients with acute PE. The study outcome was a composite of 30-day adverse events, defined as PE-related death, shock, mechanical ventilation, cardiopulmonary resuscitation, and major bleeding. Results: Overall, nine of 151 (6.0%) patients experienced 30-day adverse composite events. Multivariable logistic regression showed that TRAIL was an independent predictor of study outcome (OR 0.19 per SD; 95% CI 0.04–0.90). An ROC curve revealed that TRAIL’s area under the curve (AUC) was 0.83 (95% CI 0.76–0.88). The optimal cut-off value for TRAIL was 18 pg/mL, with a sensitivity, specificity, negative predictive value, positive predictive value, positive likelihood ratio, and negative likelihood ratio of 89%, 69%, 99%, 15%, 2.87, and 0.16, respectively. Compared with the risk stratification algorithm outlined in the 2019 ESC guidelines, our biomarker-based risk stratification strategy (combining TRAIL and hs-cTnI) has a similar risk classification effect. Conclusion: Reduced plasma TRAIL levels predict short-term adverse events in normotensive patients with acute PE. The combination of the 2019 ESC algorithm and TRAIL aids risk stratification in normotensive patients with acute PE.
Collapse
|
21
|
Mandel A, Schwarting A, Cavagna L, Triantafyllias K. Novel Surrogate Markers of Cardiovascular Risk in the Setting of Autoimmune Rheumatic Diseases: Current Data and Implications for the Future. Front Med (Lausanne) 2022; 9:820263. [PMID: 35847825 PMCID: PMC9279857 DOI: 10.3389/fmed.2022.820263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 05/30/2022] [Indexed: 11/25/2022] Open
Abstract
Patients suffering from rheumatologic diseases are known to have an increased risk for cardiovascular disease (CVD). Although the pathological mechanisms behind this excess risk have been increasingly better understood, there still seems to be a general lack of consensus in early detection and treatment of endothelial dysfunction and CVD risk in patients suffering from rheumatologic diseases and in particular in those who haven't yet shown symptoms of CVD. Traditional CVD prediction scores, such as Systematic Coronary Risk Evaluation (SCORE), Framingham, or PROCAM Score have been proposed as valid assessment tools of CVD risk in the general population. However, these risk calculators developed for the general population do not factor in the effect of the inflammatory burden, as well as other factors that can increase CVD risk in patients with rheumatic diseases, such as glucocorticoid therapy, abnormal lipoprotein function, endothelial dysfunction or accelerated atherosclerosis. Thus, their sole use could lead to underestimation of CVD risk in patients with rheumatic diseases. Therefore, there is a need for new biomarkers which will allow a valid and early assessment of CVD risk. In recent years, different research groups, including ours, have examined the value of different CVD risk factors such as carotid sonography, carotid-femoral pulse wave velocity, flow-mediated arterial dilation and others in the assessment of CVD risk. Moreover, various novel CVD laboratory markers have been examined in the setting of autoimmune diseases, such as Paraoxonase activity, Endocan and Osteoprotegerin. Dyslipidemia in rheumatoid arthritis (RA) is for instance better quantified by lipoproteins and apolipoproteins than by cholesterol levels; screening as well as pre-emptive carotid sonography hold promise to identify patients earlier, when prophylaxis is more likely to be effective. The early detection of subtle changes indicating CVD in asymptomatic patients has been facilitated through improved imaging methods; the inclusion of artificial intelligence (AI) shows promising results in more recent studies. Even though the pathophysiology of coronary artery disease in patients with autoimmune rheumatic diseases has been examined in multiple studies, as we continuously gain an increased understanding of this comorbidity, particularly in subclinical cases we still seem to fail in the stratification of who really is at risk—and who is not. A the time being, a multipronged and personalized approach of screening patients for traditional CVD risk factors, integrating modern imaging and further CV diagnostic tools and optimizing treatment seems to be a solid approach. There is promising research on novel biomarkers, likewise, methods using artificial intelligence in imaging provide encouraging data indicating possibilities of risk stratification that might become gold standard in the near future. The present review concentrates on showcasing the newest findings concerning CVD risk in patients with rheumatologic diseases and aims to evaluate screening methods in order to optimize CVD risk evaluation and thus avoiding underdiagnosis and undertreatment, as well as highlighting which patient groups are most at risk.
Collapse
Affiliation(s)
- Anna Mandel
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Andreas Schwarting
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Rheumatology, Rheumatology Center RL-P, Bad Kreuznach, Germany
| | - Lorenzo Cavagna
- Division of Rheumatology, University and IRCCS Policlinico S. Matteo Foundation, Pavia, Italy
| | - Konstantinos Triantafyllias
- Department of Internal Medicine I, Division of Rheumatology and Clinical Immunology, Johannes Gutenberg University Medical Center, Mainz, Germany
- Department of Rheumatology, Rheumatology Center RL-P, Bad Kreuznach, Germany
- *Correspondence: Konstantinos Triantafyllias
| |
Collapse
|
22
|
Arida A, Nezos A, Papadaki I, Sfikakis PP, Mavragani CP. Osteoprotegerin and MTHFR gene variations in rheumatoid arthritis: association with disease susceptibility and markers of subclinical atherosclerosis. Sci Rep 2022; 12:9534. [PMID: 35680906 PMCID: PMC9184606 DOI: 10.1038/s41598-022-13265-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 05/23/2022] [Indexed: 02/07/2023] Open
Abstract
We aimed to explore whether the rs2073618 variant (G1181C) of the osteoprotegerin (OPG) gene and the methylenetetrahydrofolate reductase (MTHFR) rs1801131 (A1298AC) and rs1801133 (C677T) gene polymorphisms contribute to rheumatoid arthritis (RA) susceptibility and RA related subclinical atherosclerosis. Overall 283 RA patients and 595 healthy controls (HC) were genotyped for common variants of the OPG and MTHFR genes using PCR based assays. Clinical and laboratory parameters were recorded following thorough chart review. Surrogate markers of subclinical atherosclerosis (Carotid/Femoral intima media thickness/plaque formation) along with traditional risk factors for atherosclerosis were assessed in all RA patients and 280HC. Increased prevalence of the CC genotype of the rs2073618 variant was detected in RA patients vs HC (42.4% vs. 33%, p-value: 0.04). RA patients with high serum titers of rheumatoid factor (RF) or anti-cyclic citrullinated peptide (CCP) antibodies displayed increased prevalence of the CC genotype of the rs2073618 variant of the OPG gene compared to HC (48.6% and 47.5 vs 33.3%, p-values: 0.0029and 0.0077 respectively). Of interest, this genotype turned to be associated with higher carotid IMT scores (0.872 ± 0.264 vs 0.816 ± 0.284, p-value: 0.01) and marginally with higher rates of carotid plaque formation (66% vs 54.1%, p = 0.06). The MTHFR 1298CC genotype was more prevalent only in the anti-CCP positive group compared to HC, with no associations detected with markers of subclinical atherosclerosis, following adjustment for traditional cardiovascular (CVD) risk factors. Reduced rates of carotid/femoral plaque formation were detected among RA patients harboring the MTHFR TT genotype (52.4 vs 72.7, p-value: 0.009, respectively). This association remained significant following adjustment for classical CVD risk factors (OR [95% CI 0.364 [0.173-0.765], p-value: 0.008). Genetic variations of the osteoprotegerin and MTHFR genes seem to increase susceptibility for seropositive RA and potentially contribute to subclinical atherosclerosis linked to RA. Larger studies are needed to confirm these findings.
Collapse
Affiliation(s)
- Aikaterini Arida
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Adrianos Nezos
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Ioanna Papadaki
- Department of Rheumatology, General Hospital of Athens "G. Gennimatas", Athens, Greece
| | - Petros P Sfikakis
- First Department of Propaedeutic Internal Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Clio P Mavragani
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens Medical School, Athens, Greece.
- Joint Academic Rheumatology Program, National and Kapodistrian University of Athens Medical School, Athens, Greece.
- Rheumatology and Clinical Immunology Unit, Fourth Department of Internal Medicine, School of Medicine, University Hospital Attikon, NKUA, 12462, Haidari, Greece.
| |
Collapse
|
23
|
Shui X, Dong R, Wu Z, Chen Z, Wen Z, Tang L, Xie X, Chen L. Association of serum sclerostin and osteoprotegerin levels with the presence, severity and prognosis in patients with acute myocardial infarction. BMC Cardiovasc Disord 2022; 22:213. [PMID: 35546224 PMCID: PMC9092859 DOI: 10.1186/s12872-022-02654-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Accepted: 05/03/2022] [Indexed: 11/18/2022] Open
Abstract
Background Bone-related proteins (such as sclerostin and osteoprotegerin [OPG]) are involved in the development of atherosclerosis. However, the relationship between bone-related proteins and acute myocardial infarction (AMI) has not been extensively evaluated. The purpose of this study was to assess the association of serum sclerostin and OPG with the presence, severity and prognosis in patients with AMI. Methods This study prospectively enrolled 152 patients attacked by acute chest pain. Serum sclerostin and OPG were detected within the first 24 h after AMI diagnosis by ELISA kits. The AMI predictive efficacy of sclerostin and OPG were analyzed by receiver operating characteristics (ROC) curve. Univariable and multivariable linear regression analyses were performed to determine the association between bone-related proteins and scores indicating the severity of coronary artery occlusion. Moreover, prognostic values were assessed by Kaplan–Meier curves and Cox regression analysis. Results There were 92 patients in AMI group, 60 in non-AMI group. Serum levels of sclerostin and OPG were significantly higher in AMI group than in non-AMI group (all p < 0.001), which showed predictive value for the presence of AMI (all p < 0.001). The area under the ROC curve values of sclerostin and OPG were 0.744 and 0.897, respectively. A multivariable linear regression analysis demonstrated that Ln-transformed sclerostin (β = 0.288, p = 0.009) and Ln-transformed OPG (Ln-OPG: β = 0.295, p = 0.019) levels were associated with GENISINI score, independently of conventional clinical parameters. In addition, Ln-OPG levels were still positively associated with GRACE score after adjustments (β = 0.320, p = 0.001). During a 1-year follow-up, patients above the median of sclerostin levels had higher incidence of major adverse cardiac events (MACE) than those below the median (p = 0.028). It was also observed that the MACE rates were higher in patients above the median of OPG levels, though no statistic importance (p = 0.060). After adjusting conventional risk factors by multivariate Cox regression, Ln-OPG was associated with incident MACE (hazard ratio = 2.188 [95% confidence intervals 1.102–4.344], p = 0.025). Conclusions Bone-related proteins could exert a potential role in early risk stratification and prognosis assessment in patients with AMI.
Collapse
Affiliation(s)
- Xing Shui
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, SunYat-Sen University, No. 600, Tianhe Road, Guangzhou, 510630, China
| | - Ruimin Dong
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, SunYat-Sen University, No. 600, Tianhe Road, Guangzhou, 510630, China
| | - Zhen Wu
- Department of Cardiac Care Unit, The Third Affiliated Hospital, SunYat-Sen University, Guangzhou, China
| | - Zefeng Chen
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, SunYat-Sen University, No. 600, Tianhe Road, Guangzhou, 510630, China
| | - Zheqi Wen
- Department of Cardiac Care Unit, The Third Affiliated Hospital, SunYat-Sen University, Guangzhou, China
| | - Leile Tang
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, SunYat-Sen University, No. 600, Tianhe Road, Guangzhou, 510630, China
| | - Xujing Xie
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, SunYat-Sen University, No. 600, Tianhe Road, Guangzhou, 510630, China.
| | - Lin Chen
- Department of Cardiovascular Medicine, The Third Affiliated Hospital, SunYat-Sen University, No. 600, Tianhe Road, Guangzhou, 510630, China.
| |
Collapse
|
24
|
The roles of osteoprotegerin in cancer, far beyond a bone player. Cell Death Dis 2022; 8:252. [PMID: 35523775 PMCID: PMC9076607 DOI: 10.1038/s41420-022-01042-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 11/08/2022]
Abstract
Osteoprotegerin (OPG), also known as tumor necrosis factor receptor superfamily member 11B (TNFRSF11B), is a member of the tumor necrosis factor (TNF) receptor superfamily. Characterized by its ability to bind to receptor activator of nuclear factor kappa B ligand (RANKL), OPG is critically involved in bone remodeling. Emerging evidence implies that OPG is far beyond a bone-specific modulator, and is involved in multiple physiological and pathological processes, such as immunoregulation, vascular function, and fibrosis. Notably, numerous preclinical and clinical studies have been conducted to assess the participation of OPG in tumorigenesis and cancer development. Mechanistic studies have demonstrated that OPG is involved in multiple hallmarks of cancer, including tumor survival, epithelial to mesenchymal transition (EMT), neo-angiogenesis, invasion, and metastasis. In this review, we systematically summarize the basis and advances of OPG from its molecular structure to translational applications. In addition to its role in bone homeostasis, the physiological and pathological impacts of OPG on human health and its function in cancer progression are reviewed, providing a comprehensive understanding of OPG. We aim to draw more attention to OPG in the field of cancer, and to propose it as a promising diagnostic or prognostic biomarker as well as potential therapeutic target for cancer.
Collapse
|
25
|
Peyronnel C, Totoson P, Petitcolin V, Bonnefoy F, Guillot X, Saas P, Verhoeven F, Martin H, Demougeot C. Effects of local cryotherapy on systemic endothelial activation, dysfunction, and vascular inflammation in adjuvant-induced arthritis (AIA) rats. Arthritis Res Ther 2022; 24:97. [PMID: 35488311 PMCID: PMC9052534 DOI: 10.1186/s13075-022-02774-1] [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: 10/05/2021] [Accepted: 04/11/2022] [Indexed: 11/10/2022] Open
Abstract
Aim This study explored the systemic vascular effects of local cryotherapy with a focus on endothelial changes and arterial inflammation in the model of rat adjuvant-induced arthritis (AIA). Methods Cryotherapy was applied twice a day on hind paws of AIA rats from the onset of arthritis to the acute inflammatory phase. Endothelial activation was studied in the aorta by measuring the mRNA levels of chemokines (CXCL-1, MCP-1 (CCL-2), MIP-1α (CCL-3)) and adhesion molecules (ICAM-1, VCAM-1) by qRT-PCR. Endothelial dysfunction was measured in isolated aortic and mesenteric rings. Aortic inflammation was evaluated via the mRNA expression of pro-inflammatory cytokines (TNF-α, IL-6) by qRT-PCR and leucocyte infiltration analysis (flow cytometry). Plasma levels of TNF-α, IL-6, IL-1β, IL-17A, and osteoprotegerin (OPG) were measured using Multiplex/ELISA. Results AIA was associated with an increased aortic expression of CXCL-1 and ICAM-1 as well as an infiltration of leucocytes and increased mRNA expression of IL-6, IL-1β, and TNF-α. Local cryotherapy, which decreased arthritis score and structural damages, reduced aortic mRNA expression of CXCL-1, IL-6, IL-1β, and TNF-α, as well as aortic infiltration of leucocytes (T lymphocytes, monocytes/macrophages, neutrophils) and improved acetylcholine-induced vasorelaxation in the aorta and mesenteric arteries. Plasma levels of IL-17A and OPG were significantly reduced by cryotherapy, while the number of circulating leucocytes was not. IL-17A levels positively correlated with endothelial activation and dysfunction. Conclusion In the AIA model, local cryotherapy reduced systemic endothelial activation, immune cell infiltration, and endothelial dysfunction. Mechanistically, the reduction of circulating levels of IL-17A appears as the possible link between joint cooling and the remote vascular effects. Supplementary Information The online version contains supplementary material available at 10.1186/s13075-022-02774-1.
Collapse
Affiliation(s)
- C Peyronnel
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - P Totoson
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - V Petitcolin
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - F Bonnefoy
- INSERM UMR 1098 RIGHT, EFS BFC, Univ. Bourgogne Franche-Comté, LabEX LipSTIC, F-25000, Besançon, France
| | - X Guillot
- Service de Rhumatologie, CHU Felix Guyon, Ile de la Réunion, Saint-Denis, France
| | - P Saas
- INSERM UMR 1098 RIGHT, EFS BFC, Univ. Bourgogne Franche-Comté, LabEX LipSTIC, F-25000, Besançon, France
| | - F Verhoeven
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France.,Service de Rhumatologie, CHRU Besançon, F-25000, Besançon, France
| | - H Martin
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France
| | - C Demougeot
- PEPITE EA4267, FHU INCREASE, Univ. Bourgogne Franche-Comté, F-25000, Besançon, France.
| |
Collapse
|
26
|
Osteoprotegerin Is a Better Predictor for Cardiovascular and All-Cause Mortality than Vascular Calcifications in a Multicenter Cohort of Patients on Peritoneal Dialysis. Biomolecules 2022; 12:biom12040551. [PMID: 35454141 PMCID: PMC9025174 DOI: 10.3390/biom12040551] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 03/29/2022] [Accepted: 04/04/2022] [Indexed: 11/19/2022] Open
Abstract
The purpose of this study was to compare vascular calcification (VC), serum osteoprotegerin (OPG) levels, and other biochemical markers to determine their value as available predictors of all-cause and cardiovascular (CV) mortality in patients on peritoneal dialysis (PD). A total of 197 patients were recruited from seven dialysis centers in Mexico City. VC was assessed with multi-slice computed tomography, measured using the calcification score (CaSc). OPG, albumin, calcium, hsC-reactive protein, phosphorous, osteocalcin, total alkaline phosphatase, and intact parathormone were also analyzed. Follow-up and mortality analyses were assessed using the Cox regression model. The mean age was 43.9 ± 12.9 years, 64% were males, and 53% were diabetics. The median OPG was 11.28 (IQR: 7.6−17.4 pmol/L), and 42% of cases had cardiovascular calcifications. The median VC was 424 (IQR:101−886). During follow-up (23 ± 7 months), there were 34 deaths, and 44% were cardiovascular in origin. In multivariable analysis, OPG was a significant predictor for all-cause (HR 1.08; p < 0.002) and CV mortality (HR 1.09; p < 0.013), and performed better than VC (HR 1.00; p < 0.62 for all-cause mortality and HR 1.00; p < 0.16 for CV mortality). For each mg/dL of albumin-corrected calcium, there was an increased risk for CV mortality, and each g/dL of albumin decreased the risk factor for all-cause mortality. OPG levels above 14.37 and 13.57 pmol/L showed the highest predictive value for all-cause and CV mortality in incident PD patients and performed better than VC.
Collapse
|
27
|
Pro-Calcific Environment Impairs Ischaemia-Driven Angiogenesis. Int J Mol Sci 2022; 23:ijms23063363. [PMID: 35328786 PMCID: PMC8954938 DOI: 10.3390/ijms23063363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/14/2022] [Accepted: 03/17/2022] [Indexed: 02/04/2023] Open
Abstract
Peripheral arterial disease (PAD) is characterised by accelerated arterial calcification and impairment in angiogenesis. Studies implicate vascular calcification as a contributor to PAD, but the mechanisms remain unclear. We aimed to determine the effect of calcification on ischaemia-driven angiogenesis. Human coronary artery endothelial cells (ECs) were treated with calcification medium (CM: CaCl2 2.7 mM, Na2PO4 2.0 mM) for 24 h and exposed to normoxia (5% CO2) or hypoxia (1.2% O2; 5% CO2 balanced with N2). In normoxia, CM significantly inhibited tubule formation and migration and upregulated calcification markers of ALP, BMP2, and Runx2. CM elevated levels of calcification-protective gene OPG, demonstrating a compensatory mechanism by ECs. CM failed to induce pro-angiogenic regulators VEGFA and HIF-1α in hypoxia and further suppressed the phosphorylation of endothelial nitric oxide synthase (eNOS) that is essential for vascular function. In vivo, osteoprotegerin-deficient mice (OPG−/−), a calcification model, were subjected to hind-limb ischaemia (HLI) surgery. OPG−/− mice displayed elevated serum alkaline phosphatase (ALP) activity compared to wild-type controls. OPG−/− mice experienced striking reductions in blood-flow reperfusion in both 8-week-old and 6-month-old mice post-HLI. This coincided with significant impairment in tissue ischaemia and reduced limb function as assessed by clinical scoring (Tarlov). This study demonstrated for the first time that a pro-calcific environment is detrimental to ischaemia-driven angiogenesis. The degree of calcification in patients with PAD can often be a limiting factor with the use of standard therapies. These highly novel findings require further studies for full elucidation of the mechanisms involved and have implications for the development of therapies to suppress calcification in PAD.
Collapse
|
28
|
Rochette L. Emerging New Biomarkers for Cardiovascular Disease. Int J Mol Sci 2022; 23:ijms23063274. [PMID: 35328695 PMCID: PMC8953083 DOI: 10.3390/ijms23063274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 03/15/2022] [Indexed: 01/27/2023] Open
Affiliation(s)
- Luc Rochette
- Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé, Université de Bourgogne-Franche Comté, 7 Bd Jeanne d'Arc, 21000 Dijon, France
| |
Collapse
|
29
|
Inverse Regulation of Serum Osteoprotegerin and B-Type Natriuretic Peptide Concentrations by Free Fatty Acids Elevation in Young Healthy Humans. Nutrients 2022; 14:nu14040837. [PMID: 35215487 PMCID: PMC8879157 DOI: 10.3390/nu14040837] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 02/06/2022] [Accepted: 02/15/2022] [Indexed: 02/05/2023] Open
Abstract
Osteoprotegerin (OPG) and B-type natriuretic peptide (BNP) are cardiovascular risk factors, interrelated with each other, with possible associations with insulin sensitivity and glucose homeostasis. The aim of this study was to assess association between OPG and BNP concentrations in a young healthy population, their relation to insulin sensitivity and obesity and their regulation by hyperinsulinemia and serum free fatty acids (FFA) elevation. The study group consisted of 59 male volunteers, 30 of whom were of a normal weight (BMI < 25 kg/m2), and 29 were overweight/obese (BMI > 25 kg/m2). Insulin sensitivity was assessed with the 2-h hyperinsulinemic-euglycemic clamp (HEC). In the subgroup of 20 subjects, the clamp was prolonged to 6 h. After one week, another 6-h clamp, with concurrent Intralipid/heparin infusion, was performed. Serum OPG was positively associated with insulin sensitivity (p = 0.002) and negatively with BMI (p = 0.019) and serum BNP (p = 0.025). In response to 6-h hyperinsulinemia, circulating BNP decreased (p < 0.001). In response to HEC with Intralipid/heparin infusion, OPG decreased (p < 0.001) and BNP increased (p < 0.001). Our data show that OPG and BNP are differentially regulated by FFA, which suggests their association with lipid-induced insulin resistance. The assessment of these cardiovascular risk factors should take into account both long-term and short-term effects associated with insulin resistance.
Collapse
|
30
|
Rochette L, Rigal E, Dogon G, Malka G, Zeller M, Vergely C, Cottin Y. Mitochondrial-derived peptides: New markers for cardiometabolic dysfunction. Arch Cardiovasc Dis 2022; 115:48-56. [PMID: 34972639 DOI: 10.1016/j.acvd.2021.10.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 02/07/2023]
Abstract
Great attention is being paid to the evaluation of new markers in blood circulation for the estimation of tissue metabolism disturbance. This endogenous disturbance may contribute to the onset and progression of cardiometabolic disease. In addition to their role in energy production and metabolism, mitochondria play a main function in cellular mechanisms, including apoptosis, oxidative stress and calcium homeostasis. Mitochondria produce mitochondrial-derived peptides that mediate the transcriptional stress response by translocating into the nucleus and interacting with deoxyribonucleic acid. This class of peptides includes humanin, mitochondrial open reading frame of the 12S ribosomal ribonucleic acid type c (MOTS-c) and small humanin-like peptides. Mitochondrial-derived peptides are regulators of metabolism, exerting cytoprotective effects through antioxidative stress, anti-inflammatory responses and antiapoptosis; they are emerging biomarkers reflecting mitochondrial function, and the circulating concentration of these proteins can be used to diagnose cardiometabolic dysfunction. The aims of this review are: (1) to describe the emerging role for mitochondrial-derived peptides as biomarkers; and (2) to discuss the therapeutic application of these peptides.
Collapse
Affiliation(s)
- Luc Rochette
- Équipe d'Accueil (EA 7460), physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche Comté, 21000 Dijon, France.
| | - Eve Rigal
- Équipe d'Accueil (EA 7460), physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche Comté, 21000 Dijon, France
| | - Geoffrey Dogon
- Équipe d'Accueil (EA 7460), physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche Comté, 21000 Dijon, France
| | - Gabriel Malka
- Centre interface applications médicales (CIAM), université Mohammed VI Polytechnique, 43150 Ben Guerir, Morocco
| | - Marianne Zeller
- Équipe d'Accueil (EA 7460), physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche Comté, 21000 Dijon, France
| | - Catherine Vergely
- Équipe d'Accueil (EA 7460), physiopathologie et épidémiologie cérébro-cardiovasculaires (PEC2), faculté des sciences de santé, université de Bourgogne-Franche Comté, 21000 Dijon, France
| | - Yves Cottin
- Cardiology Unit, CHU de Dijon-Bourgogne, 21000 Dijon, France
| |
Collapse
|
31
|
Dawson A, Li Y, Li Y, Ren P, Vasquez HG, Zhang C, Rebello KR, Ageedi W, Azares AR, Mattar AB, Sheppard MB, Lu HS, Coselli JS, Cassis LA, Daugherty A, Shen YH, LeMaire SA. Single-Cell Analysis of Aneurysmal Aortic Tissue in Patients with Marfan Syndrome Reveals Dysfunctional TGF-β Signaling. Genes (Basel) 2021; 13:95. [PMID: 35052435 PMCID: PMC8774900 DOI: 10.3390/genes13010095] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/23/2021] [Accepted: 12/23/2021] [Indexed: 02/08/2023] Open
Abstract
The molecular and cellular processes leading to aortic aneurysm development in Marfan syndrome (MFS) remain poorly understood. In this study, we examined the changes of aortic cell populations and gene expression in MFS by performing single-cell RNA sequencing (scRNA seq) on ascending aortic aneurysm tissues from patients with MFS (n = 3) and age-matched non-aneurysmal control tissues from cardiac donors and recipients (n = 4). The expression of key molecules was confirmed by immunostaining. We detected diverse populations of smooth muscle cells (SMCs), fibroblasts, and endothelial cells (ECs) in the aortic wall. Aortic tissues from MFS showed alterations of cell populations with increased de-differentiated proliferative SMCs compared to controls. Furthermore, there was a downregulation of MYOCD and MYH11 in SMCs, and an upregulation of COL1A1/2 in fibroblasts in MFS samples compared to controls. We also examined TGF-β signaling, an important pathway in aortic homeostasis. We found that TGFB1 was significantly upregulated in two fibroblast clusters in MFS tissues. However, TGF-β receptor genes (predominantly TGFBR2) and SMAD genes were downregulated in SMCs, fibroblasts, and ECs in MFS, indicating impairment in TGF-β signaling. In conclusion, despite upregulation of TGFB1, the rest of the canonical TGF-β pathway and mature SMCs were consistently downregulated in MFS, indicating a potential compromise of TGF-β signaling and lack of stimulus for SMC differentiation.
Collapse
Affiliation(s)
- Ashley Dawson
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Yanming Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Yang Li
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Pingping Ren
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Hernan G. Vasquez
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Chen Zhang
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Kimberly R. Rebello
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Waleed Ageedi
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
| | - Alon R. Azares
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX 77030, USA;
| | - Aladdein Burchett Mattar
- Division of Cardiothoracic Transplantation and Circulatory Support, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Mary Burchett Sheppard
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (M.B.S.); (H.S.L.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Hong S. Lu
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (M.B.S.); (H.S.L.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Joseph S. Coselli
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX 77030, USA;
| | - Lisa A. Cassis
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, KY 40536, USA;
| | - Alan Daugherty
- Saha Cardiovascular Research Center, University of Kentucky, Lexington, KY 40536, USA; (M.B.S.); (H.S.L.); (A.D.)
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Ying H. Shen
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX 77030, USA;
| | - Scott A. LeMaire
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (A.D.); (Y.L.); (Y.L.); (P.R.); (H.G.V.); (C.Z.); (K.R.R.); (W.A.); (J.S.C.); (Y.H.S.)
- Department of Cardiovascular Surgery, Texas Heart Institute, Houston, TX 77030, USA;
| |
Collapse
|
32
|
Zhou Y, Deng Y, Liu Z, Yin M, Hou M, Zhao Z, Zhou X, Yin L. Cytokine-scavenging nanodecoys reconstruct osteoclast/osteoblast balance toward the treatment of postmenopausal osteoporosis. SCIENCE ADVANCES 2021; 7:eabl6432. [PMID: 34818042 PMCID: PMC8612675 DOI: 10.1126/sciadv.abl6432] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Imbalance between osteoblasts and osteoclasts accounts for the incidence and deterioration of postmenopausal osteoporosis. Abnormally elevated RANKL and TNF-α levels after menopause promote osteoclast formation and inhibit osteoblast differentiation, respectively. Here, nanodecoys capable of scavenging RANKL and TNF-α were developed from preosteoclast (RAW 264.7 cell) membrane–coated poly(lactic-co-glycolic acid) (PLGA) nanoparticles, which inhibited osteoporosis and maintained bone integrity. The nanodecoys effectively escaped from macrophage capture and enabled prolonged blood circulation after systemic administration. The abundant RANK and TNF-α receptor (TNF-αR) on the cell membranes effectively neutralized RANKL and TNF-α to prevent osteoclastogenesis and promote osteoblastogenesis, respectively, thus reversing the progression of osteoporosis in the ovariectomized (OVX) mouse model. These biomimetic nanodecoys provide an effective strategy for reconstructing the osteoclast/osteoblast balance and hold great potentials for the clinical management of postmenopausal osteoporosis.
Collapse
Affiliation(s)
- Yang Zhou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Yekun Deng
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Zhongmin Liu
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Mengyuan Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Mengying Hou
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Ziyin Zhao
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
| | - Xiaozhong Zhou
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou 215004, China
| | - Lichen Yin
- Institute of Functional Nano and Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials and Devices, Soochow University, Suzhou 215123, China
- Corresponding author.
| |
Collapse
|
33
|
Jin TC, Lu JF, Luo S, Wang LC, Lu XJ, Chen J. Characterization of large yellow croaker (Larimichthys crocea) osteoprotegerin and its role in the innate immune response against to Vibrio alginolyticus. Comp Biochem Physiol B Biochem Mol Biol 2021; 258:110680. [PMID: 34688907 DOI: 10.1016/j.cbpb.2021.110680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 12/25/2022]
Abstract
Osteoprotegerin (OPG) is a member of the tumor necrosis factor receptor superfamily, contributing to inflammation, apoptosis, and differentiation. However, the function of OPG in the host immune system of teleosts remains unclear. Here, we cloned the cDNA of the LcOPG gene from large yellow croaker. LcOPG mRNA was expressed in all analyzed tissues and was upregulated by Vibrio alginolyticus infection in immune tissues and monocytes/macrophages (MO/MФ). Subsequently, the LcOPG protein was expressed and purified using a prokaryotic expression system. Recombinant LcOPG protein (rLcOPG) treatment suppressed V. alginolyticus-induced pro-inflammatory cytokine and enhanced V. alginolyticus-induced anti-inflammatory cytokine mRNA expression. Furthermore, rLcOPG decreased V. alginolyticus-induced MO/MФ apoptosis. Therefore, the results indicate that LcOPG might play a role in the immune response of V. alginolyticus-infected large yellow croaker.
Collapse
Affiliation(s)
- Tian-Cheng Jin
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Jian-Fei Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Sheng Luo
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Li-Cong Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Xin-Jiang Lu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China
| | - Jiong Chen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products, Ningbo University, Ningbo 315211, China; Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo 315211, China.
| |
Collapse
|
34
|
Liu H, Ru NY, Cai Y, Lyu Q, Guo CH, Zhou Y, Li SH, Cheng JH, Chang JR, Ma J, Su XL. The OPG/RANKL/RANK system modulates calcification of common carotid artery in simulated microgravity rats by regulating NF-κB pathway. Can J Physiol Pharmacol 2021; 100:324-333. [PMID: 34670103 DOI: 10.1139/cjpp-2021-0329] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Functional and structural adaptation of common carotid artery could be one of the important causes of postflight orthostatic intolerance after microgravity exposure, the mechanisms of which remain unclear. Recent evidence indicates that long-term spaceflight increases carotid artery stiffness, which might present a high risk to astronaut health and postflight working ability. Studies have suggested that vascular calcification is a common pathological change in cardiovascular diseases that is mainly manifested as an increase in vascular stiffness. Therefore, this study aimed to investigate whether simulated microgravity induces calcification of common carotid artery and to elucidate the underlying mechanisms. Four-week hindlimb-unweighted (HU) rats were used to simulate the deconditioning effects of microgravity on cardiovascular system. We found that simulated microgravity induced vascular smooth muscle cell (VSMC) osteogenic differentiation and medial calcification, increased receptor activator of nuclear factor κB ligand (RANKL) and RANK expression, and enhanced NF-κB activation in rat common carotid artery. In vitro activation of the RANK pathway with exogenous RANKL, a RANK ligand, increased RANK and osteoprotegerin (OPG) expression in HU rats. Moreover, the expression of osteogenic markers and activation of NF-κB in HU rats were further enhanced by exogenous RANKL but suppressed by the RANK inhibitor OPG-Fc. These results indicated that the OPG/RANKL/RANK system modulates VSMC osteogenic differentiation and medial calcification of common carotid artery in simulated microgravity rats by regulating NF-kB pathway.
Collapse
Affiliation(s)
- Huan Liu
- Xi'an Medical University, Department of Basic Medicine, Xi'an, Shaanxi, China.,Fourth Military Medical University, Department of Aerospace Physiology, Xi'an, China;
| | - Ning-Yu Ru
- Xi'an Medical University, Department of Basic Medicine, Xi'an, China;
| | - Yue Cai
- Fourth Military Medical University, Department of Cardiology, Xijing Hospital, Xi'an, China;
| | - Qiang Lyu
- Fourth Military Medical University, Department of Aerospace Physiology, Xi'an, Shaanxi, China;
| | - Chi-Hua Guo
- Xi'an Medical University, Department of Basic Medicine, Xi'an, China;
| | - Ying Zhou
- Xi'an Medical University, Department of Basic Medicine, Xi'an, China;
| | - Shao-Hua Li
- Fourth Military Medical University, 12644, Department of Aerospace Physiology, Xi'an, China;
| | - Jiu-Hua Cheng
- Fourth Military Medical University, Department of Aerospace Physiology, Xi'an, Shaanxi, China;
| | - Jin-Rui Chang
- Xi'an Medical University, Department of Basic Medicine, Xi'an, China;
| | - Jin Ma
- Fourth Military Medical University, Department of Aerospace Physiology, Xi'an, China;
| | - Xing-Li Su
- Xi'an Medical University, Department of Basic Medicine, Xi'an, China;
| |
Collapse
|
35
|
Rymarz A, Romejko K, Matyjek A, Bartoszewicz Z, Niemczyk S. Serum Osteoprotegerin Is an Independent Marker of Metabolic Complications in Non-DialysisDependent Chronic Kidney Disease Patients. Nutrients 2021; 13:3609. [PMID: 34684610 PMCID: PMC8538217 DOI: 10.3390/nu13103609] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Osteoprotegerin (OPG) belongs to the tumour necrosis factor superfamily and is known to accelerate endothelial dysfunction and vascular calcification. OPG concentrations are elevated in patients with chronic kidney disease. The aim of this study was to investigate the association between OPG levels and frequent complications of chronic kidney disease (CKD) such as anaemia, protein energy wasting (PEW), inflammation, overhydration, hyperglycaemia and hypertension. METHODS One hundred non-dialysis-dependent men with CKD stage 3-5 were included in the study. Bioimpedance spectroscopy (BIS) was used to measure overhydration, fat amount and lean body mass. We also measured the serum concentrations of haemoglobin, albumin, total cholesterol, C-reactive protein (CRP), fibrinogen and glycated haemoglobin (HgbA1c), as well as blood pressure. RESULTS We observed a significant, positive correlation between OPG and age, serum creatinine, CRP, fibrinogen, HgbA1c concentrations, systolic blood pressure and overhydration. Negative correlations were observed between OPG and glomerular filtration rate (eGFR), serum albumin concentrations and serum haemoglobin level. Logistic regression models revealed that OPG is an independent marker of metabolic complications such as anaemia, PEW, inflammation and poor renal prognosis (including overhydration, uncontrolled diabetes and hypertension) in the studied population. CONCLUSION Our results suggest that OPG can be an independent marker of PEW, inflammation and vascular metabolic disturbances in patients with chronic kidney disease.
Collapse
Affiliation(s)
- Aleksandra Rymarz
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland; (K.R.); (A.M.); (S.N.)
| | - Katarzyna Romejko
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland; (K.R.); (A.M.); (S.N.)
| | - Anna Matyjek
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland; (K.R.); (A.M.); (S.N.)
| | - Zbigniew Bartoszewicz
- Department of Internal Diseases and Endocrinology, Medical University of Warsaw, 1a Banacha St., 02-097 Warsaw, Poland;
| | - Stanisław Niemczyk
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine, 128 Szaserów Street, 04-141 Warsaw, Poland; (K.R.); (A.M.); (S.N.)
| |
Collapse
|
36
|
Cottin Y, Issa R, Benalia M, Mouhat B, Meloux A, Tribouillard L, Bichat F, Rochette L, Vergely C, Zeller M. Association between Serum Osteoprotegerin Levels and Severity of Coronary Artery Disease in Patients with Acute Myocardial Infarction. J Clin Med 2021; 10:4326. [PMID: 34640343 PMCID: PMC8509596 DOI: 10.3390/jcm10194326] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Osteoprotegerin (OPG), a glycoprotein of the tumour necrosis factor (TNF) superfamily, is one of the main biomarkers for vascular calcification. AIM We aimed to evaluate the association between serum OPG levels and extent of coronary lesions in patients with acute myocardial infarction (MI). METHODS Consecutive patients hospitalized for an acute MI who underwent coronary angiography were included. SYNTAX score was calculated to assess the severity of coronary artery disease. The population was analysed in low (5 (3-6)), medium (11 (9-13)) and high (20 (18-23)) tertiles of SYNTAX score. RESULTS Among the 378 patients included, there was a gradual increase in age, rate of diabetes, anterior wall location, and a reduction in left ventricular ejection fraction across the SYNTAX tertiles. OPG levels significantly increased across the tertiles (962 (782-1497), 1240 (870-1707), and 1464 (1011-2129) pg/mL, respectively (p < 0.001)). In multivariate analysis, OPG [OR(CI95%): 2.10 (1.29-3.49) 0.003], were associated with the high SYNTAX group, beyond hypercholesterolemia, CV history and reduced glomerular filtration rate. CONCLUSION We found an association between OPG levels and coronary lesions complexity patients with acute MI.
Collapse
Affiliation(s)
- Yves Cottin
- Cardiology Department, CHU Dijon Bourgogne, 21000 Dijon, France; (Y.C.); (R.I.); (M.B.); (B.M.); (L.T.); (F.B.)
| | - Rany Issa
- Cardiology Department, CHU Dijon Bourgogne, 21000 Dijon, France; (Y.C.); (R.I.); (M.B.); (B.M.); (L.T.); (F.B.)
| | - Mourad Benalia
- Cardiology Department, CHU Dijon Bourgogne, 21000 Dijon, France; (Y.C.); (R.I.); (M.B.); (B.M.); (L.T.); (F.B.)
| | - Basile Mouhat
- Cardiology Department, CHU Dijon Bourgogne, 21000 Dijon, France; (Y.C.); (R.I.); (M.B.); (B.M.); (L.T.); (F.B.)
| | - Alexandre Meloux
- Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (A.M.); (L.R.); (C.V.)
| | - Laura Tribouillard
- Cardiology Department, CHU Dijon Bourgogne, 21000 Dijon, France; (Y.C.); (R.I.); (M.B.); (B.M.); (L.T.); (F.B.)
| | - Florence Bichat
- Cardiology Department, CHU Dijon Bourgogne, 21000 Dijon, France; (Y.C.); (R.I.); (M.B.); (B.M.); (L.T.); (F.B.)
| | - Luc Rochette
- Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (A.M.); (L.R.); (C.V.)
| | - Catherine Vergely
- Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (A.M.); (L.R.); (C.V.)
| | - Marianne Zeller
- Equipe d’Accueil (EA 7460), Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (PEC2), Faculté des Sciences de Santé Université de Bourgogne—Franche Comté, 7 Bd Jeanne d’Arc, 21000 Dijon, France; (A.M.); (L.R.); (C.V.)
| |
Collapse
|
37
|
Circulating calcification inhibitors are associated with arterial damage in pediatric patients with primary hypertension. Pediatr Nephrol 2021; 36:2371-2382. [PMID: 33604725 PMCID: PMC8260424 DOI: 10.1007/s00467-021-04957-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 12/26/2020] [Accepted: 01/19/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Circulating calcification inhibitors: fetuin A (FA) and osteoprotegerin (OPG) together with soluble ligand of receptor activator of nuclear factor kappa-B (sRANKL) have been linked to vascular calcifications and arterial damage. This study aimed to evaluate relationships between FA, OPG, sRANKL, and arterial damage in children with primary hypertension (PH). METHODS In this cross-sectional single-center study, calcification inhibitors (FA, OPG, sRANKL) levels were measured in blood samples of 60 children with PH (median age 15.8, IQR: [14.5-16.8] years) and 20 age-matched healthy volunteers. In each participant, peripheral and central blood pressure evaluation (BP) and ambulatory BP monitoring (ABPM) were performed. Arterial damage was measured using common carotid artery intima media thickness (cIMT), pulse wave velocity (PWV), augmentation index (AIx75HR), and local arterial stiffness (ECHO-tracking-ET) analysis. RESULTS Children with PH had significantly higher peripheral and central BP, BP in ABPM, thicker cIMT, higher PWV, and AIx75HR. FA was significantly lower in patients with PH compared to healthy peers without differences in OPG, sRANKL, and OPG/sRANKL and OPG/FA ratios. In children with PH, FA level correlated negatively with cIMT Z-score and ET AIx; sRANKL level correlated negatively with ABPM systolic blood pressure (SBP), SBP load, diastolic BP load, and AIx75HR; OPG/sRANKL ratio correlated positively with SBP load, while OPG/FA ratio correlated positively with ET AIx. In multivariate analysis, FA was a significant determinant of cIMT (mm) and cIMT Z-score. CONCLUSIONS This study reveals that in children with primary hypertension, arterial damage is related to lower fetuin A concentrations.
Collapse
|
38
|
Lu CW, Wang CH, Hsu BG, Tsai JP. Serum Osteoprotegerin Level Is Negatively Associated with Bone Mineral Density in Patients Undergoing Maintenance Hemodialysis. MEDICINA (KAUNAS, LITHUANIA) 2021; 57:762. [PMID: 34440966 PMCID: PMC8399781 DOI: 10.3390/medicina57080762] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 07/13/2021] [Accepted: 07/24/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND AND OBJECTIVES Osteoprotegerin (OPG), a potent osteoclast activation inhibitor, decreases bone resorption and plays a role in mediating bone mineral density (BMD). Our aim was to evaluate the relationship between BMD and serum OPG in maintenance hemodialysis (MHD) patients. MATERIALS AND METHODS Fasting blood samples were obtained from 75 MHD patients. BMD was measured by dual-energy X-ray absorptiometry in lumbar vertebrae (L2-L4). The WHO classification criteria were applied to define osteopenia and osteoporosis. A commercial enzyme-linked immunosorbent assay was used to measure serum OPG values. RESULTS Among all MHD patients, seven (9.3%) and 20 patients (26.7%) were defined as osteoporosis and osteopenia, respectively. Female patients had lower lumbar BMD than males (p = 0.002). Older age (p = 0.023), increased serum OPG (p < 0.001) urea reduction rate (p = 0.021), Kt/V (p = 0.027), and decreased body mass index (p = 0.006) and triglycerides (p = 0.020) were significantly different between the normal, osteopenia, and osteoporosis groups. Lumbar spine BMD was positively correlated with body mass index (BMI) (p < 0.001) but negatively correlated with OPG (p < 0.001) and age (p = 0.003). After grouping patients into T scores < -1 and < -2.5, female sex and OPG (adjusted odds ratio [aOR] 1.022, 95% confidence interval [C.I.] 1.011-1.034, p < 0.001) were predictors of T scores < -1, whereas only OPG was predictive of T scores < -2.5 (aOR 1.015, 95% C.I. 1.005-1.026, p = 0.004) by multivariate stepwise logistic regression analysis. The areas under the curve for predicting T scores < -1 or < -2.5 were 0.920 (95% C.I. 0.834-0.970, p < 0.001) and 0.958 (95% C.I. 0.885-0.991, p < 0.001), respectively. CONCLUSIONS Increased serum OPG negatively correlated with lumbar BMD and could be a potential biomarker predictive of osteoporosis in MHD patients.
Collapse
Affiliation(s)
- Chia-Wen Lu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan; (C.-W.L.); (C.-H.W.)
| | - Chih-Hsien Wang
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan; (C.-W.L.); (C.-H.W.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien 97004, Taiwan; (C.-W.L.); (C.-H.W.)
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien 97004, Taiwan
- Department of Internal Medicine, Division of Nephrology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi 62247, Taiwan
| |
Collapse
|
39
|
The role of osteoprotegerin (OPG) in fibrosis: its potential as a biomarker and/or biological target for the treatment of fibrotic diseases. Pharmacol Ther 2021; 228:107941. [PMID: 34171336 DOI: 10.1016/j.pharmthera.2021.107941] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/28/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined by excessive formation and accumulation of extracellular matrix proteins, produced by myofibroblasts, that supersedes normal wound healing responses to injury and results in progressive architectural remodelling. Fibrosis is often detected in advanced disease stages when an organ is already severely damaged and can no longer function properly. Therefore, there is an urgent need for reliable and easily detectable markers to identify and monitor fibrosis onset and progression as early as possible; this will greatly facilitate the development of novel therapeutic strategies. Osteoprotegerin (OPG), a well-known regulator of bone extracellular matrix and most studied for its role in regulating bone mass, is expressed in various organs and functions as a decoy for receptor activator of nuclear factor kappa-B ligand (RANKL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Recently, OPG has been linked to fibrosis and fibrogenesis, and has been included in a panel of markers to diagnose liver fibrosis. Multiple studies now suggest that OPG may be a general biomarker suitable for detection of fibrosis and/or monitoring the impact of fibrosis treatment. This review summarizes our current understanding of the role of OPG in fibrosis and will discuss its potential as a biomarker and/or novel therapeutic target for fibrosis.
Collapse
|
40
|
Shroff GR, Bangalore S, Bhave NM, Chang TI, Garcia S, Mathew RO, Rangaswami J, Ternacle J, Thourani VH, Pibarot P. Evaluation and Management of Aortic Stenosis in Chronic Kidney Disease: A Scientific Statement From the American Heart Association. Circulation 2021; 143:e1088-e1114. [PMID: 33980041 DOI: 10.1161/cir.0000000000000979] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aortic stenosis with concomitant chronic kidney disease (CKD) represents a clinical challenge. Aortic stenosis is more prevalent and progresses more rapidly and unpredictably in CKD, and the presence of CKD is associated with worse short-term and long-term outcomes after aortic valve replacement. Because patients with advanced CKD and end-stage kidney disease have been excluded from randomized trials, clinicians need to make complex management decisions in this population that are based on retrospective and observational evidence. This statement summarizes the epidemiological and pathophysiological characteristics of aortic stenosis in the context of CKD, evaluates the nuances and prognostic information provided by noninvasive cardiovascular imaging with echocardiography and advanced imaging techniques, and outlines the special risks in this population. Furthermore, this statement provides a critical review of the existing literature pertaining to clinical outcomes of surgical versus transcatheter aortic valve replacement in this high-risk population to help guide clinical decision making in the choice of aortic valve replacement and specific prosthesis. Finally, this statement provides an approach to the perioperative management of these patients, with special attention to a multidisciplinary heart-kidney collaborative team-based approach.
Collapse
|
41
|
Role of Osteoprotegerin as Novel Marker in Urinary Stone Formers: A Pilot Study. Nephrourol Mon 2021. [DOI: 10.5812/numonthly.112974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Background: Urinary tract calculus formation can be prevented by identifying molecules and metabolic disturbances that affect this process. Osteoprotegerin (OPG), a cytokine of the TNF receptor superfamily, has been demonstrated to mediate vascular calcification and intimal calcification. Endothelial injury and oxidative stress are known to play a role in urolithiasis in the form of Randall’s plaques. Objectives: The present study aimed to compare 24-h urinary and serum OPG levels of patients with and without urolithiasis. Methods: In this case-control study, 24-h urinary levels of OPG (pg/mL), serum levels of OPG (pg/mL), and creatinine (mg/dL) were measured in both groups. Urinary and serum levels of OPG were determined by enzyme-linked immunosorbent assay (ELISA) using human OPG kits. Results: Mean serum creatinine was 0.86 ± 0.21 mg/dL in the case group and 0.77± 0.16 mg/dL in controls. The difference in the mean serum OPG levels between the cases (227.13 ± 98.02 pg/mL) and controls (47.28 ± 29.61 pg/mL) was highly significant (P value < 0.0001). The difference in the mean 24-h urinary OPG levels between the cases (156.12 ± 174.31 pg/mL) and controls (9.32 ± 23.72 pg/mL) was highly significant (P value < 0.001). Conclusions: There were significantly higher levels of OPG in serum and 24-h urine samples of cases than in controls. Hence, it requires further large studies to make OPG a diagnostic and prognostic marker.
Collapse
|
42
|
Fehérvári L, Frigy A, Kocsis L, Szabó IA, Szabo TM, Urkon M, Jakó Z, Nagy EE. Serum Osteoprotegerin and Carotid Intima-Media Thickness Are Related to High Arterial Stiffness in Heart Failure with Reduced Ejection Fraction. Diagnostics (Basel) 2021; 11:diagnostics11050764. [PMID: 33923139 PMCID: PMC8145213 DOI: 10.3390/diagnostics11050764] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/15/2021] [Accepted: 04/20/2021] [Indexed: 12/11/2022] Open
Abstract
Arterial stiffness (AS) is a complex vascular phenomenon with consequences for central hemodynamics and left-ventricular performance. Circulating biomarkers have been associated with AS; however, their value in heart failure is poorly characterized. Our aim was to evaluate the clinical and biomarker correlates of AS in the setting of heart failure with reduced ejection fraction (HFrEF). In 78 hospitalized, hemodynamically stable patients (20 women, 58 men, mean age 65.8 ± 1.41 years) with HFrEF, AS was measured using aortic pulse wave velocity (PWV). Serum OPG, RANKL, sclerostin, and DKK-1 were determined, and the relationships between the clinical variables, vascular-calcification-related biomarkers, and PWV were evaluated by correlation analysis and linear and logistic regression models. OPG and the OPG/RANKL ratio were significantly higher in the group of patients (n = 37, 47.4%) with increased PWV (>10 m/s). PWV was positively correlated with age, left-ventricular ejection fraction, and carotid intima-media thickness (cIMT), and negatively correlated with the glomerular filtration rate. OPG and cIMT were significantly associated with PWV in the logistic regression models when adjusted for hypertension, EF, and the presence of atherosclerotic manifestations. Elevated serum OPG, together with cIMT, were significantly related to increased AS in the setting of HFrEF.
Collapse
Affiliation(s)
- Lajos Fehérvári
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Attila Frigy
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Lóránd Kocsis
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
| | - István Adorján Szabó
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Internal Medicine IV, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540103 Targu Mures, Romania
| | - Timea Magdolna Szabo
- Department of Cardiology, Clinical County Hospital Mures, 540103 Targu Mures, Romania; (L.F.); (A.F.); (L.K.); (I.A.S.); (T.M.S.)
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Melinda Urkon
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
| | - Zita Jakó
- Laboratory of Medical Analysis, Emergency Clinical County Hospital Targu Mures, 540136 Targu Mures, Romania;
| | - Előd Ernő Nagy
- Department of Biochemistry and Environmental Chemistry, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Targu Mures, 540142 Targu Mures, Romania;
- Laboratory of Medical Analysis, Clinical County Hospital Mures, 540394 Targu Mures, Romania
- Correspondence: ; Tel.: +40-733-956-395
| |
Collapse
|
43
|
The Dual Role of Vitamin K2 in "Bone-Vascular Crosstalk": Opposite Effects on Bone Loss and Vascular Calcification. Nutrients 2021; 13:nu13041222. [PMID: 33917175 PMCID: PMC8067793 DOI: 10.3390/nu13041222] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 03/30/2021] [Accepted: 04/04/2021] [Indexed: 12/13/2022] Open
Abstract
Osteoporosis (OP) and vascular calcification (VC) represent relevant health problems that frequently coexist in the elderly population. Traditionally, they have been considered independent processes, and mainly age-related. However, an increasing number of studies have reported their possible direct correlation, commonly defined as “bone-vascular crosstalk”. Vitamin K2 (VitK2), a family of several natural isoforms also known as menaquinones (MK), has recently received particular attention for its role in maintaining calcium homeostasis. In particular, VitK2 deficiency seems to be responsible of the so-called “calcium paradox” phenomenon, characterized by low calcium deposition in the bone and its accumulation in the vessel wall. Since these events may have important clinical consequences, and the role of VitK2 in bone-vascular crosstalk has only partially been explained, this review focuses on its effects on the bone and vascular system by providing a more recent literature update. Overall, the findings reported here propose the VitK2 family as natural bioactive molecules that could be able to play an important role in the prevention of bone loss and vascular calcification, thus encouraging further in-depth studies to achieve its use as a dietary food supplement.
Collapse
|
44
|
Carrillo-López N, Martínez-Arias L, Alonso-Montes C, Martín-Carro B, Martín-Vírgala J, Ruiz-Ortega M, Fernández-Martín JL, Dusso AS, Rodriguez-García M, Naves-Díaz M, Cannata-Andía JB, Panizo S. The receptor activator of nuclear factor κΒ ligand receptor leucine-rich repeat-containing G-protein-coupled receptor 4 contributes to parathyroid hormone-induced vascular calcification. Nephrol Dial Transplant 2021; 36:618-631. [PMID: 33367746 DOI: 10.1093/ndt/gfaa290] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In chronic kidney disease, serum phosphorus (P) elevations stimulate parathyroid hormone (PTH) production, causing severe alterations in the bone-vasculature axis. PTH is the main regulator of the receptor activator of nuclear factor κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, which is essential for bone maintenance and also plays an important role in vascular smooth muscle cell (VSMC) calcification. The discovery of a new RANKL receptor, leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), which is important for osteoblast differentiation but with an unknown role in vascular calcification (VC), led us to examine the contribution of LGR4 in high P/high PTH-driven VC. METHODS In vivo studies were conducted in subtotally nephrectomized rats fed a normal or high P diet, with and without parathyroidectomy (PTX). PTX rats were supplemented with PTH(1-34) to achieve physiological serum PTH levels. In vitro studies were performed in rat aortic VSMCs cultured in control medium, calcifying medium (CM) or CM plus 10-7 versus 10-9 M PTH. RESULTS Rats fed a high P diet had a significantly increased aortic calcium (Ca) content. Similarly, Ca deposition was higher in VSMCs exposed to CM. Both conditions were associated with increased RANKL and LGR4 and decreased OPG aorta expression and were exacerbated by high PTH. Silencing of LGR4 or parathyroid hormone receptor 1 (PTH1R) attenuated the high PTH-driven increases in Ca deposition. Furthermore, PTH1R silencing and pharmacological inhibition of protein kinase A (PKA), but not protein kinase C, prevented the increases in RANKL and LGR4 and decreased OPG. Treatment with PKA agonist corroborated that LGR4 regulation is a PTH/PKA-driven process. CONCLUSIONS High PTH increases LGR4 and RANKL and decreases OPG expression in the aorta, thereby favouring VC. The hormone's direct pro-calcifying actions involve PTH1R binding and PKA activation.
Collapse
Affiliation(s)
- Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Cristina Alonso-Montes
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Beatriz Martín-Carro
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Julia Martín-Vírgala
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Marta Ruiz-Ortega
- Vascular and Renal Laboratory Fundación Jimenez Díaz, Universidad Autónoma Madrid, REDinREN-ISCIII, Madrid, Spain
| | - José Luis Fernández-Martín
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Adriana S Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Minerva Rodriguez-García
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain.,Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain.,Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Sara Panizo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| |
Collapse
|
45
|
Chen M, Li Y, Huang X, Gu Y, Li S, Yin P, Zhang L, Tang P. Skeleton-vasculature chain reaction: a novel insight into the mystery of homeostasis. Bone Res 2021; 9:21. [PMID: 33753717 PMCID: PMC7985324 DOI: 10.1038/s41413-021-00138-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 11/18/2020] [Accepted: 12/16/2020] [Indexed: 02/01/2023] Open
Abstract
Angiogenesis and osteogenesis are coupled. However, the cellular and molecular regulation of these processes remains to be further investigated. Both tissues have recently been recognized as endocrine organs, which has stimulated research interest in the screening and functional identification of novel paracrine factors from both tissues. This review aims to elaborate on the novelty and significance of endocrine regulatory loops between bone and the vasculature. In addition, research progress related to the bone vasculature, vessel-related skeletal diseases, pathological conditions, and angiogenesis-targeted therapeutic strategies are also summarized. With respect to future perspectives, new techniques such as single-cell sequencing, which can be used to show the cellular diversity and plasticity of both tissues, are facilitating progress in this field. Moreover, extracellular vesicle-mediated nuclear acid communication deserves further investigation. In conclusion, a deeper understanding of the cellular and molecular regulation of angiogenesis and osteogenesis coupling may offer an opportunity to identify new therapeutic targets.
Collapse
Affiliation(s)
- Ming Chen
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Yi Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Xiang Huang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Ya Gu
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Shang Li
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China
| | - Pengbin Yin
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Licheng Zhang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| | - Peifu Tang
- Department of Orthopedics, Chinese PLA General Hospital, Beijing, China.
- National Clinical Research Center for Orthopedics, Sports Medicine & Rehabilitation, Beijing, China.
| |
Collapse
|
46
|
Ortega MA, Asúnsolo Á, Pekarek L, Alvarez-Mon MA, Delforge A, Sáez MA, Coca S, Sainz F, Mon MÁ, Buján J, García-Honduvilla N. Histopathological study of JNK in venous wall of patients with chronic venous insufficiency related to osteogenesis process. Int J Med Sci 2021; 18:1921-1934. [PMID: 33850461 PMCID: PMC8040408 DOI: 10.7150/ijms.54052] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 02/11/2021] [Indexed: 12/14/2022] Open
Abstract
Chronic venous insufficiency (CVI) is one of the most common vascular pathologies worldwide. One of the risk factors for the development of CVI is aging, which is why it is related to senile changes. The main trigger of the changes that occur in the venous walls in CVI is blood flow reflux, which produces increased hydrostatic pressure, leading to valve incompetence. The cellular response is one of the fundamental processes in vascular diseases, causing the activation of cell signalling pathways such as c-Jun N-terminal kinase (JNK). Metabolic changes and calcifications occur in vascular pathology as a result of pathophysiological processes. The aim of this study was to determine the expression of JNK in venous disease and its relationship with the role played by the molecules involved in the osteogenic processes in venous tissue calcification. This was a cross-sectional study that analyzed the greater saphenous vein wall in 110 patients with (R) and without venous reflux (NR), classified according to age. Histopathological techniques were used and protein expression was analysed using immunohistochemistry techniques for JNK and markers of osteogenesis (RUNX2, osteocalcin (OCN), osteopontin (OPN)). Significantly increased JNK, RUNX2, OCN, OPN and pigment epithelium-derived factor (PEDF) protein expression and the presence of osseous metaplasia and amorphous calcification were observed in younger patients (<50 years) with venous reflux. This study shows for the first time the existence of an osteogenesis process related to the expression of JNK in the venous wall.
Collapse
Affiliation(s)
- Miguel A Ortega
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Ángel Asúnsolo
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Department of Surgery, Medical and Social Sciences, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Leonel Pekarek
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Miguel A Alvarez-Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
| | - Arnaud Delforge
- UFR of pharmacy, University of Clermont Auvergne, Clermont-Ferrand, France
| | - Miguel A Sáez
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Pathological Anatomy Service, Central University Hospital of Defence-UAH Madrid, Spain
| | - Santiago Coca
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Felipe Sainz
- Ramón y Cajal Institute of Healthcare Research (IRYCIS), Madrid, Spain
- Angiology and Vascular Surgery Service, Central University Hospital of Defence-UAH Madrid, Spain
| | - Melchor Álvarez- Mon
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
- Immune System Diseases-Rheumatology, Oncology Service and Internal Medicine, University Hospital Príncipe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Julia Buján
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| | - Natalio García-Honduvilla
- Department of Medicine and Medical Specialities, Faculty of Medicine and Health Sciences, University of Alcalá, Alcalá de Henares, Madrid, Spain
- Cancer Registry and Pathology Department, Hospital Universitario Principe de Asturias, Alcalá de Henares, Madrid, Spain
| |
Collapse
|
47
|
Pihlstrøm HK, Ueland T, Michelsen AE, Aukrust P, Gatti F, Hammarström C, Kasprzycka M, Wang J, Haraldsen G, Mjøen G, Dahle DO, Midtvedt K, Eide IA, Hartmann A, Holdaas H. Exploring the potential effect of paricalcitol on markers of inflammation in de novo renal transplant recipients. PLoS One 2020; 15:e0243759. [PMID: 33326471 PMCID: PMC7743930 DOI: 10.1371/journal.pone.0243759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 11/29/2020] [Indexed: 11/18/2022] Open
Abstract
Following a successful renal transplantation circulating markers of inflammation may remain elevated, and systemic inflammation is associated with worse clinical outcome in renal transplant recipients (RTRs). Vitamin D-receptor (VDR) activation is postulated to modulate inflammation and endothelial function. We aimed to explore if a synthetic vitamin D, paricalcitol, could influence systemic inflammation and immune activation in RTRs. Newly transplanted RTRs were included in an open-label randomized controlled trial on the effect of paricalcitol on top of standard care over the first post-transplant year. Fourteen pre-defined circulating biomarkers reflecting leukocyte activation, endothelial activation, fibrosis and general inflammatory burden were analyzed in 74 RTRs at 8 weeks (baseline) and 1 year post-engraftment. Mean changes in plasma biomarker concentrations were compared by t-test. The expression of genes coding for the same biomarkers were investigated in 1-year surveillance graft biopsies (n = 60). In patients treated with paricalcitol circulating osteoprotegerin levels increased by 0.19 ng/ml, compared with a 0.05 ng/ml increase in controls (p = 0.030). In graft tissue, a 21% higher median gene expression level of TNFRSF11B coding for osteoprotegerin was found in paricalcitol-treated patients compared with controls (p = 0.026). Paricalcitol treatment did not significantly affect the blood- or tissue levels of any other investigated inflammatory marker. In RTRs, paricalcitol treatment might increase both circulating and tissue levels of osteoprotegerin, a modulator of calcification, but potential anti-inflammatory treatment effects in RTRs are likely very modest. [NCT01694160 (2012/107D)]; [www.clinicaltrials.gov].
Collapse
Affiliation(s)
- Hege Kampen Pihlstrøm
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- * E-mail:
| | - Thor Ueland
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- K.G. Jebsen Thrombosis Research and Expertise Center, University of Tromsø, Tromsø, Norway
| | - Annika E. Michelsen
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Franscesca Gatti
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Laboratory of Immunohistochemistry and Immunopathology, University of Oslo, Oslo, Norway
| | - Clara Hammarström
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Laboratory of Immunohistochemistry and Immunopathology, University of Oslo, Oslo, Norway
| | - Monika Kasprzycka
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Laboratory of Immunohistochemistry and Immunopathology, University of Oslo, Oslo, Norway
| | - Junbai Wang
- Department of Pathology, Oslo University Hospital, Oslo, Norway
| | - Guttorm Haraldsen
- Department of Pathology, Oslo University Hospital, Oslo, Norway
- K.G. Jebsen Inflammation Research Centre, Laboratory of Immunohistochemistry and Immunopathology, University of Oslo, Oslo, Norway
| | - Geir Mjøen
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Dag Olav Dahle
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Karsten Midtvedt
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Ivar Anders Eide
- Division of Medicine, Department of Nephrology, Akershus University Hospital, Oslo, Norway
| | - Anders Hartmann
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Hallvard Holdaas
- Department of Surgery, Inflammation Medicine and Transplantation, Section of Nephrology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| |
Collapse
|
48
|
Lin WC, Tsai JP, Lai YH, Lin YL, Kuo CH, Wang CH, Hsu BG. Serum osteoprotegerin level is positively associated with peripheral artery disease in patients with peritoneal dialysis. Ren Fail 2020; 42:131-136. [PMID: 31950864 PMCID: PMC7006676 DOI: 10.1080/0886022x.2020.1714654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/30/2019] [Accepted: 01/01/2020] [Indexed: 12/25/2022] Open
Abstract
Osteoprotegerin (OPG) is a potential biomarker of cardiovascular disease complications and severity. Peripheral arterial disease (PAD) is associated with an increased risk of death in patients on peritoneal dialysis (PD). Therefore, this study aimed to evaluate the relationship between serum OPG levels and PAD by measuring the ankle-brachial index (ABI) of patients on PD. A commercial enzyme-linked immunosorbent assay kit was used to measure OPG values. Left or right ABI values of <0.9 were categorized as the low ABI group. Among 70 patients on PD, 13 (18.6%) were categorized in the low ABI group. Patients in the low ABI group had higher prevalence of diabetes mellitus (p = .044) and higher serum C-reactive protein (CRP) (p < .001) and OPG levels (p < .001) but lower creatinine (p = .013) and peritoneal Kt/V (p = .048) levels than those in the normal ABI group. Results of multivariable logistic regression analysis revealed that OPG [adjusted odds ratio (aOR) 1.027, 95% confidence interval (CI) 1.010-1.045, p = .002] and CRP (aOR 1.102, 95% CI 1.006-1.207, p = .037) levels were independent predictors of PAD in patients on PD. OPG can also be used to predict PAD development with the area under the receiver operating characteristic curve of 0.823 (95% CI: 0.714-0.904, p < .001) in patients on PD. Therefore, serum OPG and CRP levels can be considered as risk factors for PAD development in patients on PD.
Collapse
Affiliation(s)
- Wei-Chen Lin
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Yu-Hsien Lai
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yu-Li Lin
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chiu-Huang Kuo
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Hsien Wang
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
49
|
Hannappe MA, Arnould L, Méloux A, Mouhat B, Bichat F, Zeller M, Cottin Y, Binquet C, Vergely C, Creuzot-Garcher C, Guenancia C. Vascular density with optical coherence tomography angiography and systemic biomarkers in low and high cardiovascular risk patients. Sci Rep 2020; 10:16718. [PMID: 33028913 PMCID: PMC7542456 DOI: 10.1038/s41598-020-73861-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 09/03/2020] [Indexed: 12/18/2022] Open
Abstract
We aimed to compare retinal vascular density in Optical Coherence Tomography Angiography (OCT-A) between patients hospitalized for acute coronary syndrome (ACS) and control patients and to investigate correlation with angiogenesis biomarkers. Patients hospitalized for an acute coronary syndrome (ACS) in the Intensive Care Unit were included in the "high cardiovascular risk" group while patients without cardiovascular risk presenting in the Ophthalmology department were included as "control". Both groups had blood sampling and OCT-A imaging. Retina microvascularization density in the superficial capillary plexus was measured on 3 × 3 mm angiograms centered on the macula. Angiopoietin-2, TGF-β1, osteoprotegerin, GDF-15 and ST-2 were explored with ELISA or multiplex method. Overall, 62 eyes of ACS patients and 42 eyes of controls were included. ACS patients had significantly lower inner vessel length density than control patients (p = 0.004). A ROC curve found that an inner vessel length density threshold below 20.05 mm-1 was moderately associated with ACS. Significant correlation was found between serum levels of angiopoietin-2 and osteoprotegerin, and retinal microvascularization in OCT-A (R = - 0.293, p = 0.003; R = - 0.310, p = 0.001). Lower inner vessel length density measured with OCT-A was associated with ACS event and was also correlated with higher concentrations of angiopoietin-2 and osteoprotegerin.
Collapse
Affiliation(s)
- Marc-Antoine Hannappe
- Ophthalmology Department, University Hospital, 14 rue Paul Gaffarel, 21079, Dijon Cedex, France.,Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France
| | - Louis Arnould
- Ophthalmology Department, University Hospital, 14 rue Paul Gaffarel, 21079, Dijon Cedex, France. .,Centre des Sciences du Goût et de l'Alimentation, AgroSup Dijon, CNRS, INRAE, Université Bourgogne Franche-Comté, 21000, Dijon, France. .,INSERM, CIC1432, Clinical Epidemiology Unit, Dijon, France. .,Dijon University Hospital, Clinical Investigation Center, Clinical Epidemiology/Clinical Trials Unit, Dijon, France.
| | - Alexandre Méloux
- Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France.,Cardiology Department, University Hospital, Dijon, France
| | - Basile Mouhat
- Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France.,Cardiology Department, University Hospital, Dijon, France
| | - Florence Bichat
- Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France.,Cardiology Department, University Hospital, Dijon, France
| | - Marianne Zeller
- Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France.,Cardiology Department, University Hospital, Dijon, France
| | - Yves Cottin
- Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France.,Cardiology Department, University Hospital, Dijon, France
| | - Christine Binquet
- INSERM, CIC1432, Clinical Epidemiology Unit, Dijon, France.,Dijon University Hospital, Clinical Investigation Center, Clinical Epidemiology/Clinical Trials Unit, Dijon, France
| | - Catherine Vergely
- Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France
| | - Catherine Creuzot-Garcher
- Ophthalmology Department, University Hospital, 14 rue Paul Gaffarel, 21079, Dijon Cedex, France.,INSERM, CIC1432, Clinical Epidemiology Unit, Dijon, France.,Dijon University Hospital, Clinical Investigation Center, Clinical Epidemiology/Clinical Trials Unit, Dijon, France
| | - Charles Guenancia
- Laboratoire de Physiopathologie et Epidémiologie Cérébro-Cardiovasculaires (EA7460, PEC2), UFR Des Sciences de Santé, Bourgogne Franche-Comté University, Dijon, France.,Cardiology Department, University Hospital, Dijon, France
| |
Collapse
|
50
|
Early Endothelial Activation Precedes Glycocalyx Degradation and Microvascular Dysfunction in Experimentally Induced Plasmodium falciparum and Plasmodium vivax Infection. Infect Immun 2020; 88:IAI.00895-19. [PMID: 32122938 DOI: 10.1128/iai.00895-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/25/2020] [Indexed: 12/13/2022] Open
Abstract
Endothelial activation and microvascular dysfunction are key pathogenic processes in severe malaria. We evaluated the early role of these processes in experimentally induced Plasmodium falciparum and P. vivax infection. Participants were enrolled in induced blood-stage malaria clinical trials. Plasma osteoprotegerin, angiopoietin-2, and von Willebrand Factor (vWF) levels were measured as biomarkers of endothelial activation. Microvascular function was assessed using peripheral arterial tonometry and near-infrared spectroscopy, and the endothelial glycocalyx was assessed by sublingual videomicroscopy and measurement of biomarkers of degradation. Forty-five healthy, malaria-naive participants were recruited from 5 studies. Osteoprotegerin and vWF levels increased in participants following inoculation with P. vivax (n = 16) or P. falciparum (n = 15), with the angiopoietin-2 level also increasing in participants following inoculation with P. falciparum For both species, the most pronounced increase was seen in osteoprotegerin. This was particularly marked in participants inoculated with P. vivax, where the osteoprotegerin level correlated with the levels of parasitemia and the malaria clinical score. There were no changes in measures of endothelial glycocalyx or microvascular function. Plasma biomarkers of endothelial activation increased in early P. falciparum and P. vivax infection and preceded changes in the endothelial glycocalyx or microvascular function. The more pronounced increase in osteoprotegerin suggests that this biomarker may play a role in disease pathogenesis.
Collapse
|