1
|
Hamadi N, Beegam S, Zaaba NE, Elzaki O, Alderei A, Alfalahi M, Alhefeiti S, Alnaqbi D, Alshamsi S, Nemmar A. Protective Effects of Nerolidol on Thrombotic Events, Systemic Inflammation, Oxidative Stress, and DNA Damage Following Pulmonary Exposure to Diesel Exhaust Particles. Biomedicines 2025; 13:729. [PMID: 40149705 PMCID: PMC11940484 DOI: 10.3390/biomedicines13030729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/05/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Inhalation of environmental particulate air pollution has been reported to cause pulmonary and systemic events including coagulation disturbances, systemic inflammation, and oxidative stress. Nerolidol, a naturally occurring sesquiterpene alcohol, has effective antioxidant and anti-inflammatory effects. Hence, the aim in the present investigation was to evaluate the potential ameliorative effects of nerolidol on the coagulation and systemic actions induced by pulmonary exposure to diesel exhaust particles (DEPs). Methods: Nerolidol (100 mg/kg) was given to mice by oral gavage one hour before the intratracheal instillation of DEPs (0.5 mg/kg), and 24 h later various markers of coagulation and systemic toxicity were evaluated. Results: Nerolidol treatment significantly abrogated DEP-induced platelet aggregation in vivo and in vitro. Nerolidol has also prevented the shortening of the prothrombin time and activated plasma thromboplastin time triggered by DEP exposure. Likewise, while the concentrations of fibrinogen and plasminogen activator inhibitor-1 were increased by DEP administration, that of tissue plasminogen activator was significantly decreased. These effects were abolished in the group of mice concomitantly treated with nerolidol and DEP. Moreover, plasma markers of inflammation, oxidative stress, and endothelial dysfunction which were significantly increased in the DEP-treated group, returned to control levels in the nerolidol + DEP group. Nerolidol treatment significantly ameliorated the increase in the concentrations of hypoxia-inducible factor 1α, galectin-3, and neutrophil gelatinase-associated lipocalin induced by pulmonary exposure to DEP. The co-administration of nerolidol + DEPs significantly mitigated the increase in markers of oxidative DNA damage, 8-hydroxy-2-deoxyguanosine, and apoptosis, cleaved-caspase-3, induced by DEP. Conclusions: Collectively, our data demonstrate that nerolidol exert significant ameliorative actions against DEP-induced thrombotic events, endothelial dysfunction, systemic inflammation, oxidative stress, DNA damage, and apoptosis. Pending further pharmacological and toxicological studies, nerolidol could be a promising agent to alleviate the toxicity of inhaled DEPs and other pollutant particles.
Collapse
Affiliation(s)
- Naserddine Hamadi
- Department of Life and Environmental Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi P.O. Box 144534, United Arab Emirates;
| | - Sumaya Beegam
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Nur Elena Zaaba
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Ozaz Elzaki
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Alreem Alderei
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Maha Alfalahi
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Shamma Alhefeiti
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Dana Alnaqbi
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Salama Alshamsi
- Department of Physiology, College of Medicine and Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates; (S.B.); (N.E.Z.); (O.E.); (A.A.); (M.A.); (S.A.); (D.A.); (S.A.)
| | - Abderrahim Nemmar
- Department of Life and Environmental Sciences, College of Natural and Health Sciences, Zayed University, Abu Dhabi P.O. Box 144534, United Arab Emirates;
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain P.O. Box 15551, United Arab Emirates
| |
Collapse
|
2
|
Yan Y, Wang L, Zhong N, Wen D, Liu L. Multifaced roles of adipokines in endothelial cell function. Front Endocrinol (Lausanne) 2024; 15:1490143. [PMID: 39558976 PMCID: PMC11570283 DOI: 10.3389/fendo.2024.1490143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Obesity significantly contributes to the progression of cardiovascular diseases (CVDs) and elevates the risk of cardiovascular mortality. Atherosclerosis, the primary pathogenic process underlying CVDs, initiates with vascular endothelial dysfunction, serving as the cornerstone of vascular lesions. Adipokines, bioactive molecules secreted by adipose tissue that regulate metabolic and endocrine functions, play a pivotal role in modulating endothelial function during atherosclerosis. This review comprehensively examines the distinct roles of various adipokines in regulating endothelial function in atherosclerosis. We categorize these adipokines into two main groups: protective adipokines, including adiponectin, FGF21, CTRP9, PGRN, Omentin, and Vaspin, and detrimental adipokines such as leptin, Chemerin, Resistin, FABP4, among others. Targeting specific adipokines holds promise for novel clinical interventions in the management of atherosclerosis-related CVDs, thereby providing a theoretical foundation for cardiovascular disease treatment strategies.
Collapse
Affiliation(s)
- Yu Yan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Lihui Wang
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Ni Zhong
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Donghua Wen
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
3
|
Lee H, Liu KH, Yang YH, Liao JD, Lin BS, Wu ZZ, Chang AC, Tseng CC, Wang MC, Tsai YS. Advances in uremic toxin detection and monitoring in the management of chronic kidney disease progression to end-stage renal disease. Analyst 2024; 149:2784-2795. [PMID: 38647233 DOI: 10.1039/d4an00057a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Patients with end-stage kidney disease (ESKD) rely on dialysis to remove toxins and stay alive. However, hemodialysis alone is insufficient to completely remove all/major uremic toxins, resulting in the accumulation of specific toxins over time. The complexity of uremic toxins and their varying clearance rates across different dialysis modalities poses significant challenges, and innovative approaches such as microfluidics, biomarker discovery, and point-of-care testing are being investigated. This review explores recent advances in the qualitative and quantitative analysis of uremic toxins and highlights the use of innovative methods, particularly label-mediated and label-free surface-enhanced Raman spectroscopy, primarily for qualitative detection. The ability to analyze uremic toxins can optimize hemodialysis settings for more efficient toxin removal. Integration of multiple omics disciplines will also help identify biomarkers and understand the pathogenesis of ESKD, provide deeper understanding of uremic toxin profiling, and offer insights for improving hemodialysis programs. This review also highlights the importance of early detection and improved understanding of chronic kidney disease to improve patient outcomes.
Collapse
Affiliation(s)
- Han Lee
- Laboratory of Engineered Materials for Biomedical Applications, Department of Materials Science and Engineering, National Cheng Kung University, 1 University Road, Tainan 701, Taiwan.
| | - Kuan-Hung Liu
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City, 701, Taiwan.
| | - Yu-Hsuan Yang
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City, 701, Taiwan.
| | - Jiunn-Der Liao
- Laboratory of Engineered Materials for Biomedical Applications, Department of Materials Science and Engineering, National Cheng Kung University, 1 University Road, Tainan 701, Taiwan.
| | - Bo-Shen Lin
- Laboratory of Engineered Materials for Biomedical Applications, Department of Materials Science and Engineering, National Cheng Kung University, 1 University Road, Tainan 701, Taiwan.
| | - Zheng-Zhe Wu
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City, 701, Taiwan.
| | - Alice Chinghsuan Chang
- Center for Measurement Standards, Industrial Technology Research Institute, No. 321, Kuang Fu Road, Section 2, Hsinchu 300, Taiwan.
| | - Chin-Chung Tseng
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City, 701, Taiwan.
| | - Ming-Cheng Wang
- Division of Nephrology, Department of Internal Medicine, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City, 701, Taiwan.
| | - Yau-Sheng Tsai
- Center for Clinical Medicine Research, College of Medicine, National Cheng Kung University, No.1, University Road, Tainan City, 701, Taiwan.
| |
Collapse
|
4
|
Chan MJ, Liu KD. Acute Kidney Injury and Subsequent Cardiovascular Disease: Epidemiology, Pathophysiology, and Treatment. Semin Nephrol 2024; 44:151515. [PMID: 38849258 DOI: 10.1016/j.semnephrol.2024.151515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
Cardiovascular disease poses a significant threat to individuals with kidney disease, including those affected by acute kidney injury (AKI). In the short term, AKI has several physiological consequences that can impact the cardiovascular system. These include fluid and sodium overload, activation of the renin-angiotensin-aldosterone system and sympathetic nervous system, and inflammation along with metabolic complications of AKI (acidosis, electrolyte imbalance, buildup of uremic toxins). Recent studies highlight the role of AKI in elevating long-term risks of hypertension, thromboembolism, stroke, and major adverse cardiovascular events, though some of this increased risk may be due to the impact of AKI on the course of chronic kidney disease. Current management strategies involve avoiding nephrotoxic agents, optimizing hemodynamics and fluid balance, and considering renin-angiotensin-aldosterone system inhibition or sodium-glucose cotransporter 2 inhibitors. However, future research is imperative to advance preventive and therapeutic strategies for cardiovascular complications in AKI. This review explores the existing knowledge on the cardiovascular consequences of AKI, delving into epidemiology, pathophysiology, and treatment of various cardiovascular complications following AKI.
Collapse
Affiliation(s)
- Ming-Jen Chan
- Kidney Research Center, Department of Nephrology, Chang Gung Memorial Hospital, Taoyuan, Taiwan; Graduate Institute of Clinical Medical Science, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kathleen D Liu
- Divisions of Nephrology and Critical Care Medicine, Departments of Medicine and Anesthesia, University of California, San Francisco, CA.
| |
Collapse
|
5
|
Saenz-Pipaon G, Jover E, van der Bent ML, Orbe J, Rodriguez JA, Fernández-Celis A, Quax PHA, Paramo JA, López-Andrés N, Martín-Ventura JL, Nossent AY, Roncal C. Role of LCN2 in a murine model of hindlimb ischemia and in peripheral artery disease patients, and its potential regulation by miR-138-5P. Atherosclerosis 2023; 385:117343. [PMID: 37871404 DOI: 10.1016/j.atherosclerosis.2023.117343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 08/07/2023] [Accepted: 10/10/2023] [Indexed: 10/25/2023]
Abstract
BACKGROUND AND AIMS Peripheral arterial disease (PAD) is a leading cause of morbimortality worldwide. Lipocalin-2 (LCN2) has been associated with higher risk of amputation or mortality in PAD and might be involved in muscle regeneration. Our aim is to unravel the role of LCN2 in skeletal muscle repair and PAD. METHODS AND RESULTS WT and Lcn2-/- mice underwent hindlimb ischemia. Blood and crural muscles were analyzed at the inflammatory and regenerative phases. At day 2, Lcn2-/- male mice, but not females, showed increased blood and soleus muscle neutrophils, and elevated circulating pro-inflammatory monocytes (p < 0.05), while locally, total infiltrating macrophages were reduced (p < 0.05). Moreover, Lcn2-/- soleus displayed an elevation of Cxcl1 (p < 0.001), and Cxcr2 (p < 0.01 in males), and a decrease in Ccl5 (p < 0.05). At day 15, Lcn2 deficiency delayed muscle recovery, with higher density of regenerating myocytes (p < 0.04) and arterioles (αSMA+, p < 0.025). Reverse target prediction analysis identified miR-138-5p as a potential regulator of LCN2, showing an inverse correlation with Lcn2 mRNA in skeletal muscles (rho = -0.58, p < 0.01). In vitro, miR-138-5p mimic reduced Lcn2 expression and luciferase activity in murine macrophages (p < 0.05). Finally, in human serum miR-138-5p was inversely correlated with LCN2 (p ≤ 0.001 adjusted, n = 318), and associated with PAD (Odds ratio 0.634, p = 0.02, adjusted, PAD n = 264, control n = 54). CONCLUSIONS This study suggests a possible dual role of LCN2 in acute and chronic conditions, with a probable role in restraining inflammation early after skeletal muscle ischemia, while being associated with vascular damage in PAD, and identifies miR-138-5p as one potential post-transcriptional regulator of LCN2.
Collapse
Affiliation(s)
- Goren Saenz-Pipaon
- Laboratory of Atherothrombosis, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain
| | - Eva Jover
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - M Leontien van der Bent
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Josune Orbe
- Laboratory of Atherothrombosis, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; RICORS-ICTUS, ISCIII, Madrid, Spain
| | - Jose A Rodriguez
- Laboratory of Atherothrombosis, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; CIBERCV, ISCIII, Madrid, Spain
| | - Amaya Fernández-Celis
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | - Paul H A Quax
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Jose A Paramo
- Laboratory of Atherothrombosis, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; CIBERCV, ISCIII, Madrid, Spain; Hematology Service, Clínica Universidad de Navarra, Pamplona, Spain
| | - Natalia López-Andrés
- IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), Pamplona, Spain
| | | | - Anne Yaël Nossent
- Department of Vascular Surgery, Leiden University Medical Center, Leiden, the Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Carmen Roncal
- Laboratory of Atherothrombosis, Cima Universidad de Navarra, Pamplona, Spain; IdiSNA, Instituto de Investigación Sanitaria de Navarra, Pamplona, Spain; CIBERCV, ISCIII, Madrid, Spain.
| |
Collapse
|
6
|
Wu S, Zhang X, Wang Y, Zheng H, Zhu M. Lipid Metabolism Reprogramming of Immune Cells in Acne: An Update. Clin Cosmet Investig Dermatol 2023; 16:2391-2398. [PMID: 37675181 PMCID: PMC10478778 DOI: 10.2147/ccid.s424478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/15/2023] [Indexed: 09/08/2023]
Abstract
Acne vulgaris is one of the most widespread skin conditions and the main reason for visiting a dermatologist. Inflammatory response and abnormal infiltrations of immune cells are the main pathogenesis of acne. The increased lipid is the prerequisite for the acne, and the perturbation of lipid composition and content is consistent with the severity of acne. Furthermore, the increased lipid production not only contributes to the occurrence and development of acne, but also sensitizes the function of immune cells. The lipid metabolic dysfunction aggravates the severity of local tissue and provides pro-inflammatory-cytokine cues, which indicates the crucial roles of lipid metabolism on immune cells. Recent advances have demonstrated the lipid metabolism reprogramming of various immune cells in acne lesion. The abnormal lipid accumulation, lipolysis, and fatty acid oxidation lead to the activation and differentiation of immune cells, which promotes the pro-inflammatory cytokines production. Thus, this review discusses the emerging role of lipid metabolism reprogramming of immune cells in the progress of acne and aims to constitute food for others' projects involved in acne research.
Collapse
Affiliation(s)
- Shuhui Wu
- Department of Dermatology, Key Laboratory of Vascular Biology and Translational Medicine, Education Department of Hunan Province, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Xi Zhang
- Department of Physical Education and Health Promotion, Hunan University of Technology and Business, Changsha, Hunan, People’s Republic of China
| | - Yun Wang
- Department of Dermatology, Key Laboratory of Vascular Biology and Translational Medicine, Education Department of Hunan Province, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Huie Zheng
- Department of Dermatology, Key Laboratory of Vascular Biology and Translational Medicine, Education Department of Hunan Province, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| | - Mingfang Zhu
- Department of Dermatology, Key Laboratory of Vascular Biology and Translational Medicine, Education Department of Hunan Province, The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, People’s Republic of China
| |
Collapse
|
7
|
Romejko K, Markowska M, Niemczyk S. The Review of Current Knowledge on Neutrophil Gelatinase-Associated Lipocalin (NGAL). Int J Mol Sci 2023; 24:10470. [PMID: 37445650 DOI: 10.3390/ijms241310470] [Citation(s) in RCA: 75] [Impact Index Per Article: 37.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 06/15/2023] [Accepted: 06/20/2023] [Indexed: 07/15/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a 25-kDa protein that is secreted mostly by immune cells such as neutrophils, macrophages, and dendritic cells. Its production is stimulated in response to inflammation. The concentrations of NGAL can be measured in plasma, urine, and biological fluids such as peritoneal effluent. NGAL is known mainly as a biomarker of acute kidney injury and is released after tubular damage and during renal regeneration processes. NGAL is also elevated in chronic kidney disease and dialysis patients. It may play a role as a predictor of the progression of renal function decreases with complications and mortality due to kidney failure. NGAL is also useful in the diagnostic processes of cardiovascular diseases. It is highly expressed in injured heart tissue and atherosclerostic plaque; its serum concentrations correlate with the severity of heart failure and coronary artery disease. NGAL increases inflammatory states and its levels rise in arterial hypertension, obesity, diabetes, and metabolic complications such as insulin resistance, and is also involved in carcinogenesis. In this review, we present the current knowledge on NGAL and its involvement in different pathologies, especially its role in renal and cardiovascular diseases.
Collapse
Affiliation(s)
- Katarzyna Romejko
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Magdalena Markowska
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| | - Stanisław Niemczyk
- Department of Internal Diseases, Nephrology and Dialysis, Military Institute of Medicine-National Research Institute, 128 Szaserów Street, 04-141 Warsaw, Poland
| |
Collapse
|
8
|
Martínez-Ayala P, Alanis-Sánchez GA, Álvarez-Zavala M, Sánchez-Reyes K, Ruiz-Herrera VV, Cabrera-Silva RI, González-Hernández LA, Ramos-Becerra C, Cardona-Muñoz E, Andrade-Villanueva JF. Effect of antiretroviral therapy on decreasing arterial stiffness, metabolic profile, vascular and systemic inflammatory cytokines in treatment-naïve HIV: A one-year prospective study. PLoS One 2023; 18:e0282728. [PMID: 36930649 PMCID: PMC10022802 DOI: 10.1371/journal.pone.0282728] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 02/21/2023] [Indexed: 03/18/2023] Open
Abstract
INTRODUCTION Cardiovascular disease is a major cause of death among people living with HIV (PLH). Non-treated PLH show increased levels of inflammation and biomarkers of vascular activation, and arterial stiffness as a prognostic cardiovascular disease risk factor. We investigated the effect of one year of ART on treatment-naïve HIV(+) individuals on arterial stiffness and inflammatory and vascular cytokines. METHODS We cross-sectionally compared aortic stiffness via tonometry, inflammatory, and vascular serum cytokines on treatment-naïve (n = 20) and HIV (-) (n = 9) matched by age, sex, metabolic profile, and Framingham score. We subsequently followed young, treatment-naïve individuals after 1-year of ART and compared aortic stiffness, metabolic profile, and inflammatory and vascular serum biomarkers to baseline. Inflammatory biomarkers included: hs-CRP, D-Dimer, SAA, sCD163s, MCP-1, IL-8, IL-18, MRP8/14. Vascular cytokines included: myoglobin, NGAL, MPO, Cystatin C, ICAM-1, VCAM-1, and MMP9. RESULTS Treatment-naïve individuals were 34.8 years old, mostly males (95%), and with high smoking prevalence (70%). Baseline T CD4+ was 512±324 cells/mcL. cfPWV was similar between HIV(-) and treatment-naïve (6.8 vs 7.3 m/s; p = 0.16) but significantly decreased after ART (-0.52 m/s; 95% CI -0.87 to -0.16; p0.006). Almost all the determined cytokines were significantly higher compared to controls, except for MCP-1, myoglobin, NGAL, cystatin C, and MMP-9. At follow-up, only total cholesterol and triglycerides increased and all inflammatory cytokines significantly decreased. Regarding vascular cytokines, MPO, ICAM-1, and VCAM-1 showed a reduction. D-Dimer tended to decrease (p = 0.06) and hs-CRP did not show a significant reduction (p = 0.17). CONCLUSION One year of ART had a positive effect on reducing inflammatory and vascular cytokines and arterial stiffness.
Collapse
Affiliation(s)
- Pedro Martínez-Ayala
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
| | | | - Monserrat Álvarez-Zavala
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Karina Sánchez-Reyes
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Vida Verónica Ruiz-Herrera
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
| | - Rodolfo Ismael Cabrera-Silva
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Luz Alicia González-Hernández
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
| | - Carlos Ramos-Becerra
- Department of Physiology, Arterial Stiffness Laboratory, University of Guadalajara, Guadalajara, Mexico
| | - Ernesto Cardona-Muñoz
- Department of Physiology, Arterial Stiffness Laboratory, University of Guadalajara, Guadalajara, Mexico
| | - Jaime Federico Andrade-Villanueva
- HIV Unit Department, University Hospital "Fray Antonio Alcalde", University of Guadalajara, Guadalajara, Mexico
- Clinical Medicine Department, HIV and Immunodeficiencies Research Institute, CUCS-University of Guadalajara, Guadalajara, Mexico
- * E-mail:
| |
Collapse
|
9
|
Opdebeeck B, Huysmans I, Van den Branden A, Orriss IR, D'Haese PC, Verhulst A. Deletion of the P2Y 2 receptor aggravates internal elastic lamina calcification in chronic kidney disease mice through upregulation of alkaline phosphatase and lipocalin-2. FASEB J 2023; 37:e22701. [PMID: 36520031 DOI: 10.1096/fj.202201044r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 10/29/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022]
Abstract
Calcification of the medial layer, inducing arterial stiffness, contributes significantly to cardiovascular mortality in patients with chronic kidney disease (CKD). Extracellular nucleotides block the mineralization of arteries by binding to purinergic receptors including the P2Y2 receptor. This study investigates whether deletion of the P2Y2 receptor influences the development of arterial media calcification in CKD mice. Animals were divided into: (i) wild type mice with normal renal function (control diet) (n = 8), (ii) P2Y2 R-/- mice with normal renal function (n = 8), (iii) wild type mice with CKD (n = 27), and (iv) P2Y2 R-/- mice with CKD (n = 22). To induce CKD, animals received an alternating (0.2-0.3%) adenine diet for 7 weeks. All CKD groups developed a similar degree of chronic renal failure as reflected by high serum creatinine and phosphorus levels. Also, the presence of CKD induced calcification in the heart and medial layer of the aortic wall. However, deletion of the P2Y2 receptor makes CKD mice more susceptible to the development of calcification in the heart and aorta (aortic calcium scores (median ± IQR), CKD-wild type: 0.34 ± 4.3 mg calcium/g wet tissue and CKD-P2Y2 R-/- : 4.0 ± 13.2 mg calcium/g wet tissue). As indicated by serum and aortic mRNA markers, this P2Y2 R-/- mediated increase in CKD-related arterial media calcification was associated with an elevation of calcification stimulators, including alkaline phosphatase and inflammatory molecules interleukin-6 and lipocalin 2. The P2Y2 receptor should be considered as an interesting therapeutic target for tackling CKD-related arterial media calcification.
Collapse
Affiliation(s)
- Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Ine Huysmans
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Astrid Van den Branden
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, UK
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
10
|
Jover E, Matilla L, Martín-Núñez E, Garaikoetxea M, Navarro A, Fernández-Celis A, Gainza A, Arrieta V, García-Peña A, Álvarez V, Sádaba R, Jaisser F, López-Andrés N. Sex-dependent expression of neutrophil gelatinase-associated lipocalin in aortic stenosis. Biol Sex Differ 2022; 13:71. [PMID: 36510294 PMCID: PMC9743642 DOI: 10.1186/s13293-022-00480-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/20/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Accumulating evidence suggest the existence of sex-related differences in the pathogenesis of aortic stenosis (AS) with inflammation, oxidative stress, fibrosis and calcification being over-represented in men. Neutrophil gelatinase-associated lipocalin (NGAL) is expressed in a myriad of tissues and cell types, and it is associated with acute and chronic pathological processes comprising inflammation, fibrosis or calcification. Sex-dependent signatures have been evidenced for NGAL which expression has been associated predominantly in males to metabolic and cardiovascular disorders. We aimed to analyse sex-related differences of NGAL in AS and its role in the inflammatory and fibrocalcific progression of AS. METHODS AND RESULTS 220 (60.45% men) patients with severe AS elective for surgical aortic valve (AV) replacement were recruited. Immunohistochemistry revealed higher expression of NGAL in calcific areas of AVs and that was validated by qPCR in in 65 (60% men) donors. Valve interstitial cells (VICs) were a source of NGAL in these samples. Proteome profiler analyses evidenced higher expression of NGAL in men compared to women, and that was further validated by ELISA. NGAL expression in the AV was correlated with inflammation, oxidative stress, and osteogenic markers, as well as calcium score. The expression of NGAL, both intracellular and secreted (sNGAL), was significantly deregulated only in calcifying male-derived VICs. Depletion of intracellular NGAL in calcifying male-derived VICs was associated with pro-inflammatory profiles, dysbalanced matrix remodelling and pro-osteogenic profiles. Conversely, exogenous NGAL mediated inflammatory and dysbalanced matrix remodelling in calcifying VICs, and all that was prevented by the pharmacological blockade of NGAL. CONCLUSIONS Owing to the over-expression of NGAL, the AV from men may be endowed with higher expression of inflammatory, oxidative stress, matrix remodelling and osteogenic markers supporting the progression of calcific AS phenotypes. The expression of NGAL in the VIC emerges as a potential therapeutic checkpoint, with its effects being potentially reverted by the pharmacological blockade of extracellular NGAL.
Collapse
Affiliation(s)
- Eva Jover
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Lara Matilla
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Ernesto Martín-Núñez
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Mattie Garaikoetxea
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Adela Navarro
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Amaya Fernández-Celis
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Alicia Gainza
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Vanessa Arrieta
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Amaia García-Peña
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Virginia Álvarez
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Rafael Sádaba
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| | - Frederic Jaisser
- grid.508487.60000 0004 7885 7602Centre de Recherche des Cordeliers, INSERM, UMRS 1138, Sorbonne Université, USPC, Université Paris Descartes, Université Paris Diderot, Université Paris Cité, 15 rue de l’Ecole de Médecine, 75006 Paris, France ,grid.410527.50000 0004 1765 1301Université de Lorraine, INSERM, Centre d’Investigations Cliniques-Plurithématique 1433, UMR 1116, CHRU de Nancy, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT (Cardiovascular and Renal Clinical Trialists), Nancy, France
| | - Natalia López-Andrés
- grid.411730.00000 0001 2191 685XCardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigación Sanitaria de Navarra (IdiSNA), Hospital Universitario de Navarra (HUN), Universidad Pública de Navarra (UPNA), C/Irunlarrea 3, 31008 Pamplona, Spain
| |
Collapse
|
11
|
Urinary neutrophil gelatinase-associated lipocalin (NGAL) can potentially predict vascular complications and reliably risk stratify patients with peripheral arterial disease. Sci Rep 2022; 12:8312. [PMID: 35585171 PMCID: PMC9117222 DOI: 10.1038/s41598-022-12286-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 05/09/2022] [Indexed: 01/22/2023] Open
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is expressed in atherosclerotic plaques and implicated in the development of cardiovascular diseases. Peripheral arterial disease (PAD) is an atherosclerotic disease that often results in major cardiovascular events. This study aimed to prospectively examine the potential of urine NGAL (uNGAL) in predicting worsening PAD status and major adverse limb events (MALE). Baseline urine NGAL (uNGAL) and urine creatinine (uCr) concentrations were measured in PAD (n = 121) and non-PAD (n = 77) patients. Levels of uNGAL were normalized for urine creatinine (uNGAL/uCr). Outcomes included worsening PAD status, which was defined as a drop in ankle brachial index (ABI) > 0.15, and major adverse limb events (MALE), which was defined as a need for surgical revascularization or amputations. PAD patients had 2.30-fold higher levels of uNGAL/uCr [median (IQR) 31.8 (17.0–62.5) μg/g] in comparison to non-PAD patients [median (IQR) 73.3 (37.5–154.7) μg/g] (P = 0.011). Multivariate cox analysis showed that uNGAL/uCr levels were independently associated with predicting worsening PAD status and MALE outcomes. Cumulative survival analysis, over follow up period, demonstrated a direct correlation between elevated uNGAL/uCr levels and PAD disease progression and MALE outcomes. These data demonstrate an association between elevated uNGAL/uCr levels and worsening PAD disease status and MALE outcomes, indicating its potential for risk-stratification of PAD patients.
Collapse
|
12
|
Loureiro ACC, Nocrato GF, Correia ALL, de Matos RS, Filho JCCN, Daher EDF, Pinto FHM, de Oliveira AC, Ceccatto VM, Fortunato RS, de Carvalho DP. Serum and Urinary Neutrophil Gelatinase-Associated Lipocalin Are Not Associated With Serum Redox Parameters in Amateur Athletes After an Ultramarathon. Front Physiol 2022; 13:811514. [PMID: 35370771 PMCID: PMC8970054 DOI: 10.3389/fphys.2022.811514] [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: 11/08/2021] [Accepted: 01/31/2022] [Indexed: 11/13/2022] Open
Abstract
Objective To evaluate the relationship between oxidative stress and NGAL levels in blood and urine of amateur athletes after participating in a 100 km ultramarathon. Methodology The sample was composed of seven athletes, submitted to anthropometric assessment, cardiopulmonary exercise test, collection of urine and blood, measurement of body weight. The rate of perceived exertion (RPE), competition duration, heart rate (HR), energy expenditure and oxygen consumption (V'O2") were also measured during the event. The energy consumption during the race was verified at its end. The analyses were based on the means (M) and respective standard deviations (SD), with statistical significance set at 5% (p < 0.05). Paired t-test was used for comparison between the periods before and after the competition, and Pearson's correlation coefficient was used to measure the linear correlation between quantitative variables. Results Body mass index (BMI) of the sample was 25.75 kg/m2 ± 3.20, body fat percentage 18.54% ± 4.35% and V'O2"max 48.87% ± 4.78. Glucose, cortisol, and neutrophil gelatinase-associated lipocalin (NGAL) (p < 0.01) as well as glutathione peroxidase (GPx) active were higher after the race when compared to basal values. Moreover, lactate, creatinine, microalbuminuria, and glomerular filtration rate (GFR) (p < 0.001) were also higher after the race. After the competition, there was a significant correlation only between serum NGAL and creatinine, which was classified as strong and positive (r: 0.77; p < 0.05). There was a significant reduction (p < 0.05) of body weight after the event (72.40 kg ± 9.78) compared to before it (73.98 kg ± 10.25). In addition, we found an increase of RPE (p < 0.001) after the race. The competition lasted 820.60 min (±117.00), with a 127.85 bpm (±12.02) HR, a 2209.72 kcal ± 951.97 energy consumption, 7837.16 kcal ± 195.71 energy expenditure, and 28.78 ml/kg/min-1 (±4.66) relative V'O2"max. Conclusion The lack of correlation between oxidative stress biomarkers and serum and urine NGAL suggests that NGAL is more sensitive to inflammatory processes than to ROS levels.
Collapse
Affiliation(s)
| | | | | | - Robson Salviano de Matos
- Department of Clinical Medicine at the Federal University of Ceará, Ceará Federal University, Fortaleza, Brazil
| | | | | | | | | | | | - Rodrigo Soares Fortunato
- Carlos Chagas Filho Biophysics Institute, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| | - Denise Pires de Carvalho
- Carlos Chagas Filho Biophysics Institute, Rio de Janeiro Federal University, Rio de Janeiro, Brazil
| |
Collapse
|
13
|
Utkusavas A, Gurel Gurevin E, Yilmazer N, Uvez A, Oztay F, Bulut H, Ustunova S, Esener OBB, Sonmez K, Erol Kutucu D, Meral I, Dimas K, Armutak EI. Effects of combined administration of doxorubicin and chloroquine on lung pathology in mice with solid Ehrlich ascites carcinoma. Biotech Histochem 2022; 97:555-566. [PMID: 35240890 DOI: 10.1080/10520295.2022.2036369] [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] [Indexed: 12/24/2022] Open
Abstract
Combined use of a chemotherapeutic agent and an autophagy inhibitor is a novel cancer treatment strategy. We investigated the effects of chloroquine (CQ) on lung pathology caused by both solid Ehrlich ascites carcinoma (EAC) and doxorubicin (DXR). A control group and eight experimental groups of adult female mice were inoculated subcutaneously with 2.5 × 106 EAC cells. DXR (1.5 mg/kg and 3 mg/kg) and CQ (25 mg/kg and 50 mg/kg) alone or in combination were injected intraperitoneally on days 2, 7 and 12 following inoculation with EAC cells. Lung tissue samples were examined using immunohistochemistry (IHC) for endothelial (eNOS), inducible nitric oxide synthase (iNOS) and neutrophil gelatinase-associated lipocalin (NGAL). Serum catalase (CAT), glutathione peroxidase (GPx), superoxide dismutase (SOD) and malondialdehyde (MDA) levels were measured using ELISA. We found decreased levels of iNOS and eNOS in the groups that received 1.5 mg/kg DXR alone and in combination with 25 mg/kg and 50 mg/kg CQ. Combined administration of DXR and CQ partially prevented disruption of alveolar structure. Levels of antioxidant enzymes and MDA were lower in all treated groups; the greatest reduction was observed in mice that received the combination of 25 mg/kg CQ + 1.5 mg/kg DXR. Levels of NGAL were elevated in all treated groups. We found that CQ ameliorated both EAC and DOX induced lung pathology in female mice with solid EAC by reducing oxidative stress.
Collapse
Affiliation(s)
- Ayfer Utkusavas
- Department of Pulmonology, Istanbul Mehmet Akif Ersoy Thoracic and Cardiovascular Surgery Training and Research Hospital, Istanbul, Turkey
| | - Ebru Gurel Gurevin
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Nadim Yilmazer
- Department of Biology, Faculty of Arts and Sciences, Tekirdag Namik Kemal University, Tekirdag, Turkey
| | - Ayca Uvez
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Fusun Oztay
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Huri Bulut
- Department of Medical Biochemistry, School of Medicine, Istinye University, Istanbul, Turkey
| | - Savas Ustunova
- Department of Physiology, School of Medicine, Bezmialem Vakıf University, Istanbul, Turkey
| | - Osman B Burak Esener
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Kivilcim Sonmez
- Department of Pathology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Deniz Erol Kutucu
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey.,Department of Biology, Institute of Graduate Studies in Science and Engineering, Istanbul University, Istanbul, Turkey
| | - Ismail Meral
- Department of Physiology, School of Medicine, Bezmialem Vakıf University, Istanbul, Turkey
| | - Konstantinos Dimas
- Department of Pharmacology, School of Medicine, Thessaly University, Larissa, Greece
| | - Elif Ilkay Armutak
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
14
|
Saenz-Pipaon G, Ravassa S, Larsen KL, Martinez-Aguilar E, Orbe J, Rodriguez JA, Fernandez-Alonso L, Gonzalez A, Martín-Ventura JL, Paramo JA, Lindholt JS, Roncal C. Lipocalin-2 and Calprotectin Potential Prognosis Biomarkers in Peripheral Arterial Disease. Eur J Vasc Endovasc Surg 2022; 63:648-656. [DOI: 10.1016/j.ejvs.2022.01.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2021] [Revised: 01/07/2022] [Accepted: 01/16/2022] [Indexed: 11/03/2022]
|
15
|
Ren K, Xia Y. Lipocalin 2 Participates in the Epidermal Differentiation and Inflammatory Processes of Psoriasis. J Inflamm Res 2022; 15:2157-2166. [PMID: 35386225 PMCID: PMC8979418 DOI: 10.2147/jir.s358492] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/22/2022] [Indexed: 12/16/2022] Open
Abstract
As a multifunctional cytokine, lipocalin 2 is weakly expressed in skin and serum under normal conditions. However, it is over-expressed by neutrophils and keratinocytes in the skin lesions and sera in several skin diseases. Recent studies demonstrated that lipocalin 2 participates in the pathogenesis of psoriasis by exerting versatile effects on skin resident cells and infiltrating immune cells. Lipocalin 2 inhibits the synthesis of keratin, involucrin, and loricrin in keratinocytes, leading to epidermal parakeratosis via the Tcf7l1-lipocalin 2 signaling axis. It also recruits inflammatory cells such as T cells and neutrophils into skin lesions via the IL-23/IL17, p38-MAPK, and ERK-1/2 signaling pathways. Additionally, lipocalin 2 and other cytokines such as IL-17 have the synergetic effects on skin cells. The neutralization of lipocalin 2 or relevant cytokines can alleviate psoriasis, verifying that lipocalin 2 is an effective interfering target for psoriasis. In this review, we summarize the roles of lipocalin 2 in the processes of psoriatic inflammation and the promising therapeutic strategies based on lipocalin 2-related molecules.
Collapse
Affiliation(s)
- Kaixuan Ren
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
| | - Yumin Xia
- Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, 710004, People’s Republic of China
- Correspondence: Yumin Xia, Department of Dermatology, The Second Affiliated Hospital of Xi’an Jiaotong University, 157 Xiwu Road, Xi’an, 710004, People’s Republic of China, Tel/Fax +86-29-87679969, Email
| |
Collapse
|
16
|
Magnetic Resonance Imaging of Contrast-Induced Acute Renal Injury and Related Pathological Alterations In Vivo. Anal Cell Pathol (Amst) 2022; 2022:6984200. [PMID: 35256925 PMCID: PMC8898140 DOI: 10.1155/2022/6984200] [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: 11/13/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 11/18/2022] Open
Abstract
Background. The definitive mechanisms of CI-AKI include contrast medium (CM) nephrotoxicity and CM disturbances in renal blood flow, but how the immune system responds to CM has rarely been mentioned in previous studies, and different cell death pathways have not been clearly distinguished. Aim. To confirm whether MRI detect early CI-AKI and to investigate whether immunity-related responses, pyroptosis, and mitophagy participate in contrast-induced acute renal injury (CI-AKI). Methods. C57BL/6 mice with CI-AKI were established by tail vein injection of iodixanol 320. Magnetic resonance imaging of 9.4 T scanner and microscopic appearance of renal H&E staining were tools to test the occurrence of CI-AKI at different times. Immunohistochemistry and NGAL were used to examine the immune responses in the kidneys with CI-AKI. Transmission electron microscopy and western blot methods were used to distinguish various cell death pathways in CI-AKI. Key Results. The densitometry of T2WI, DTI, and BOLD presents CI-AKI in a regular way. The microscopic appearance presents the strongest renal damage in CI-AKI mice that existed between 12 h (
) and 24 h (
) after contrast medium (CM) injection. Strong correlation may exist between MRI densitometry (T2WI, DTI, and BOLD) and pathology. Neutrophil and macrophage chemotaxis occurred in CI-AKI, and we observed that Ly6G was the strongest at 48 h (
). Pyroptosis (Nlrp3/caspase-1,
), mitophagy (BNIP/Nix,
), and apoptosis (Bax,
) occurred in CI-AKI. Conclusions. fMRI can detect early CI-AKI immediately after CM injection. NLRP3 inflammasomes are involved in CI-AKI, and mitophagy may play a role in mitigating kidney injury. The mitochondrion is one of the key organelles in the tubular epithelium implicated in CI-AKI.
Collapse
|
17
|
Lipocalin 2 Mediates Skin Inflammation in Psoriasis via the SREBP2-NLRC4 Axis. J Invest Dermatol 2022; 142:2194-2204.e11. [PMID: 35120997 DOI: 10.1016/j.jid.2022.01.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 12/24/2021] [Accepted: 01/17/2022] [Indexed: 01/26/2023]
Abstract
Lipocalins are a family of secreted adipokines that regulate cell lipid metabolism and immune responses. Although we have previously revealed that lipocalin 2 (LCN2) modulates neutrophil activation in psoriasis, the other roles of LCN2 in psoriatic local inflammation have remained elusive. Here, we found that 24p3R, the well-known specific receptor of LCN2, was highly expressed in the lesional epidermis of psoriasis patients. Silencing 24p3R alleviated hyperkeratosis, inflammatory cell infiltration, and overexpression of inflammatory mediators in an imiquimod (IMQ)-induced psoriasis-like mouse model. In vitro, LCN2 enhanced the expression of pro-inflammatory factors in primary keratinocytes, such as interleukin (IL)-1β, IL-23, C-X-C motif chemokine ligand (CXCL)1, and CXCL10, which was paralleled by enforced cholesterol biosynthetic signaling. Importantly, taking in vivo and in vitro approaches, we discovered the sterol response element binding factor 2 (SREBP2), a vital transcriptional factor in cholesterol synthesis pathway, as the critical mediator of LCN2-induced keratinocyte activation, which bond to the promoter region of NLR-family CARD-containing protein 4 (NLRC4). Suppressing SREBP2 in mice attenuated NLRC4 signaling and psoriasis-like dermatitis. Taken together, this study identifies the critical role of LCN2-SREBP2-NLRC4 axis in the pathogenesis of psoriasis, and proposes 24p3R or SREBP2 as potential therapeutic target for psoriasis.
Collapse
|
18
|
Gan J, Zheng Y, Yu Q, Zhang Y, Xie W, Shi Y, Yu N, Yan Y, Lin Z, Yang H. Serum Lipocalin-2 Levels Are Increased and Independently Associated With Early-Stage Renal Damage and Carotid Atherosclerotic Plaque in Patients With T2DM. Front Endocrinol (Lausanne) 2022; 13:855616. [PMID: 35547005 PMCID: PMC9081837 DOI: 10.3389/fendo.2022.855616] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 03/15/2022] [Indexed: 11/24/2022] Open
Abstract
OBJECTIVES Diabetic nephropathy (DN), one of the major complications of diabetes mellitus, is the major cause of end-stage renal failure that finally increases the risk of cardiovascular disease and mortality. The aim of this study is to explore the relationship between serum lipocalin-2 (LCN-2) levels and DN and carotid atherosclerotic plaque (CAP) in patients with type 2 diabetes mellitus (T2DM). METHODS We have performed a prospective study of 749 T2DM patients with or without DN. Blood samples were collected and used to test serum LCN-2 levels, renal function, as well as biochemical parameters. CAP in these subjects was determined by ultrasonography. RESULTS In these 749 subjects with T2DM, an increased morbidity of CAP was observed in T2DM patients with DN as compared with those without this complication (P < 0.05). Interestingly, serum LCN-2 levels were significantly increased in T2DM patients with DN or CAP compared with T2DM alone [97.71 (71.49-130.13) vs. 77.29 (58.83-115.05) ng/ml, P < 0.001]. In addition, serum LCN-2 levels in T2DM patients with DN and CAP were significantly higher than that of T2DM patients with DN or CAP [131.37 (101.43-182.04) vs. 97.71(71.49-130.13) ng/ml, P < 0.001]. Furthermore, serum LCN-2 levels were positively correlated with hemoglobin A1c, systolic blood pressure, hypertension, CAP, and DN, as well as renal function factors including uric acid, creatinine, the estimated glomerular filtration rate, and urinary albumin-to-creatinine ratio, respectively (P < 0.05), but negatively correlated with HDL-c (P < 0.05). The multinomial logistic regression analysis showed that serum LCN-2 was independently associated with DN and CAP in patients with T2DM after the adjustment for risk factors (P < 0.001). CONCLUSIONS Early-stage renal damage is a risk factor associated with the incidence of CAP in patients with T2DM. Serum LCN-2 is significantly increased and associated with early-stage renal damage and the incidence of CAP in patients with T2DM.
Collapse
Affiliation(s)
- Jing Gan
- The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yu Zheng
- The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiongli Yu
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Yingchao Zhang
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Wei Xie
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Yaru Shi
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Ning Yu
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
| | - Yu Yan
- The 2nd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhuofeng Lin
- School of Pharmaceutical College, Wenzhou Medical University, Wenzhou, China
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- Laboratory Animal Center of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhuofeng Lin, ; Hong Yang,
| | - Hong Yang
- The 3rd Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Zhuofeng Lin, ; Hong Yang,
| |
Collapse
|
19
|
Baran R, Marchal S, Garcia Campos S, Rehnberg E, Tabury K, Baselet B, Wehland M, Grimm D, Baatout S. The Cardiovascular System in Space: Focus on In Vivo and In Vitro Studies. Biomedicines 2021; 10:59. [PMID: 35052739 PMCID: PMC8773383 DOI: 10.3390/biomedicines10010059] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 12/24/2021] [Accepted: 12/25/2021] [Indexed: 12/13/2022] Open
Abstract
On Earth, humans are subjected to a gravitational force that has been an important determinant in human evolution and function. During spaceflight, astronauts are subjected to several hazards including a prolonged state of microgravity that induces a myriad of physiological adaptations leading to orthostatic intolerance. This review summarises all known cardiovascular diseases related to human spaceflight and focusses on the cardiovascular changes related to human spaceflight (in vivo) as well as cellular and molecular changes (in vitro). Upon entering microgravity, cephalad fluid shift occurs and increases the stroke volume (35-46%) and cardiac output (18-41%). Despite this increase, astronauts enter a state of hypovolemia (10-15% decrease in blood volume). The absence of orthostatic pressure and a decrease in arterial pressures reduces the workload of the heart and is believed to be the underlying mechanism for the development of cardiac atrophy in space. Cellular and molecular changes include altered cell shape and endothelial dysfunction through suppressed cellular proliferation as well as increased cell apoptosis and oxidative stress. Human spaceflight is associated with several cardiovascular risk factors. Through the use of microgravity platforms, multiple physiological changes can be studied and stimulate the development of appropriate tools and countermeasures for future human spaceflight missions in low Earth orbit and beyond.
Collapse
Affiliation(s)
- Ronni Baran
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus, Denmark; (R.B.); (D.G.)
| | - Shannon Marchal
- Department of Astronomy, Catholic University of Leuven, 3000 Leuven, Belgium;
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.R.); (K.T.); (B.B.)
| | - Sebastian Garcia Campos
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (S.G.C.); (M.W.)
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Emil Rehnberg
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.R.); (K.T.); (B.B.)
- Department of Molecular Biotechnology, Ghent University, 9000 Ghent, Belgium
| | - Kevin Tabury
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.R.); (K.T.); (B.B.)
- Department of Biomedical Engineering, University of South Carolina, Columbia, SC 29208, USA
| | - Bjorn Baselet
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.R.); (K.T.); (B.B.)
| | - Markus Wehland
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (S.G.C.); (M.W.)
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Daniela Grimm
- Department of Biomedicine, Aarhus University, Ole Worms Allé 4, 8000 Aarhus, Denmark; (R.B.); (D.G.)
- Department of Microgravity and Translational Regenerative Medicine, Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany; (S.G.C.); (M.W.)
- Research Group ‘Magdeburger Arbeitsgemeinschaft für Forschung unter Raumfahrt- und Schwerelosigkeitsbedingungen’ (MARS), Otto von Guericke University, Universitätsplatz 2, 39106 Magdeburg, Germany
| | - Sarah Baatout
- Department of Astronomy, Catholic University of Leuven, 3000 Leuven, Belgium;
- Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Boeretang 200, 2400 Mol, Belgium; (E.R.); (K.T.); (B.B.)
- Department of Molecular Biotechnology, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
20
|
Bonnard B, Ibarrola J, Lima-Posada I, Fernández-Celis A, Durand M, Genty M, Lopez-Andreés N, Jaisser F. Neutrophil Gelatinase-Associated Lipocalin From Macrophages Plays a Critical Role in Renal Fibrosis Via the CCL5 (Chemokine Ligand 5)-Th2 Cells-IL4 (Interleukin 4) Pathway. Hypertension 2021; 79:352-364. [PMID: 34794340 DOI: 10.1161/hypertensionaha.121.17712] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
NGAL (neutrophil gelatinase-associated lipocalin; or lipocalin 2, Lcn2) is a novel mineralocorticoid target in the cardiovascular system. We showed that Lcn2 gene invalidation protects against proteinuria and renal injury upon mineralocorticoid excess and we hypothesized that NGAL produced from macrophages promotes the expression of chemoattractant molecules involved these renal lesions. The role of NGAL was analyzed using myeloid-specific (MΦ KO NGAL) Lcn2 knockout mice challenged with uni-nephrectomy, aldosterone, and salt (NAS) for 6 weeks. The role of the CCL5 (chemokine ligand 5) and IL4 (interleukin 4) in kidney fibrosis was studied by administration of the CCL5 receptor antagonist maraviroc or by injections of an anti-IL4 neutralizing antibody. In CTL mice, NAS increased the renal expression of extracellular matrix proteins, such as collagen I, αSMA, and fibronectin associated with interstitial fibrosis which were blunted in MΦ KO NGAL mice. The expression of CCL5 was blunted in sorted macrophages from MΦ KO NGAL mice challenged by NAS and in macrophages obtained from KO NGAL mice and challenged ex vivo with aldosterone and salt. The pharmacological blockade of the CCL5 receptor reduced renal fibrosis and the CD4+ Th cell infiltration induced by NAS. Neutralization of IL4 in NAS mice blunted kidney fibrosis and the overexpression of profibrotic proteins, such as collagen I, αSMA, and fibronectin. In conclusion, NGAL produced by macrophages plays a critical role in renal fibrosis and modulates the CCL5/IL4 pathway in mice exposed to mineralocorticoid excess.
Collapse
Affiliation(s)
- Benjamin Bonnard
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Jaime Ibarrola
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Ixchel Lima-Posada
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Amaya Fernández-Celis
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Manon Durand
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Marie Genty
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.)
| | - Natalia Lopez-Andreés
- Cardiovascular Translational Research, Navarrabiomed (Miguel Servet Foundation), Instituto de Investigacioón Sanitaria de Navarra (IdiSNA), Pamplona, Spain (J.I., A.F.-C., N.L.-A.)
| | - Frédéric Jaisser
- Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, France (B.B., I.L.-P., M.D., M.G., F.J.).,INSERM, Clinical Investigation Centre 1433, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, Nancy, France (F.J.)
| |
Collapse
|
21
|
Gu Y, Sun W, Xu ZH, Wang J, Hu X, Lu ZZ, Zhang XW. Neutrophil Gelatinase-Associated Lipocalin 2 Accelerates Hypoxia-Induced Endothelial Cell Injury via eNOS/NRF2 Signalling. CELL JOURNAL 2021; 23:435-444. [PMID: 34455719 PMCID: PMC8405076 DOI: 10.22074/cellj.2021.7167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Accepted: 03/17/2020] [Indexed: 11/18/2022]
Abstract
Objective Neutrophil gelatinase-associated lipocalin (NGAL), a lipocalin, is implicated in many cardiovascular diseases
(CVD). The effect of NGAL on endothelial cells (ECs), particularly on ECs injured because of hypoxia, is unclear. In this
study, we aim to explore the effect of NGAL in an EC injury in response to hypoxia. Materials and Methods In this experimental study, we isolated and cultured mouse heart ECs (MHECs). The EC
injury model was established by exposure of the ECs to hypoxia for 24 hours. The ECs were treated with NGAL (30,
60, 120, 250 and 500 ng/ml). Cell inflammation and oxidative stress were detected by corresponding assays. Apoptotic
cells were stained by the terminal deoxynucleotidyl transferase dUTP nick end labelling (TUNEL) assay.
Results NGAL increased the inflammatory response at the baseline level and further augmented the hypoxia-induced
inflammation response. Reactive oxygen species (ROS) levels increased upon NGAL treatment, which caused
antioxidase/oxidase imbalance. NGAL also exaggerated hypoxia-induced oxidative stress. The cell apoptosis rate also
increased in both the NGAL-treated normoxic and hypoxic conditions. NGAL also reduced endothelial nitric oxide
synthase (eNOS)-nitric oxide (NO) signalling, thus decreasing the expression and nuclear translocation of nuclear
factor erythroid-2-related factor 2 (NRF2), which was confirmed by overexpression of NRF2.
Conclusion NGAL exaggerates EC injury in both normoxic and hypoxic conditions by inhibiting the eNOS-NRF2 pathway.
Collapse
Affiliation(s)
- Yang Gu
- Department of Cardiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Z Huo Xu
- Department of Cardiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jing Wang
- Department of Cardiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xiao Hu
- Department of Cardiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Zhou-Zhou Lu
- Department of Cardiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Xi-Wen Zhang
- Department of Cardiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China.
| |
Collapse
|
22
|
Efentakis P, Molitor M, Kossmann S, Bochenek ML, Wild J, Lagrange J, Finger S, Jung R, Karbach S, Schäfer K, Schulz A, Wild P, Münzel T, Wenzel P. Tubulin-folding cofactor E deficiency promotes vascular dysfunction by increased endoplasmic reticulum stress. Eur Heart J 2021; 43:488-500. [PMID: 34132336 PMCID: PMC8830526 DOI: 10.1093/eurheartj/ehab222] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 10/29/2020] [Accepted: 03/26/2021] [Indexed: 12/14/2022] Open
Abstract
AIMS Assessment of endothelial function in humans by measuring flow-mediated dilation (FMD) risk-stratifies individuals with established cardiovascular disease, whereas its predictive value is limited in primary prevention. We therefore aimed to establish and evaluate novel markers of FMD at the population level. METHODS AND RESULTS In order to identify novel targets that were negatively correlated with FMD and investigate their contribution to vascular function, we performed a genome-wide association study (GWAS) of 4175 participants of the population based Gutenberg Health Study. Subsequently, conditional knockout mouse models deleting the gene of interest were generated and characterized. GWAS analysis revealed that single-nucleotide polymorphisms (SNPs) in the tubulin-folding cofactor E (TBCE) gene were negatively correlated with endothelial function and TBCE expression. Vascular smooth muscle cell (VSMC)-targeted TBCE deficiency was associated with endothelial dysfunction, aortic wall hypertrophy, and endoplasmic reticulum (ER) stress-mediated VSMC hyperproliferation in mice, paralleled by calnexin up-regulation and exacerbated by the blood pressure hormone angiotensin II. Treating SMMHC-ERT2-Cre+/-TBCEfl/fl mice with the ER stress modulator tauroursodeoxycholic acid amplified Raptor/Beclin-1-dependent autophagy and reversed vascular dysfunction. CONCLUSION TBCE and tubulin homeostasis seem to be novel predictors of vascular function and offer a new drug target to ameliorate ER stress-dependent vascular dysfunction.
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Michael Molitor
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Sabine Kossmann
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Magdalena L Bochenek
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Johannes Wild
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Jeremy Lagrange
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Stefanie Finger
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Rebecca Jung
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Susanne Karbach
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Katrin Schäfer
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Andreas Schulz
- Department of Cardiology-Preventive Cardiology and Medical Prevention, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Philipp Wild
- Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Department of Cardiology-Preventive Cardiology and Medical Prevention, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Philip Wenzel
- Department of Cardiology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,Center for Thrombosis and Hemostasis, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany.,German Center for Cardiovascular Research (DZHK)-Partner site Rhine-Main, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
23
|
Morales-Primo AU, Becker I, Zamora-Chimal J. Neutrophil extracellular trap-associated molecules: a review on their immunophysiological and inflammatory roles. Int Rev Immunol 2021; 41:253-274. [PMID: 34036897 DOI: 10.1080/08830185.2021.1921174] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Neutrophil extracellular traps (NETs) are a defense mechanism against pathogens. They are composed of DNA and various proteins and have the ability to hinder microbial spreading and survival. However, NETs are not only related to infections but also participate in sterile inflammatory events. In addition to DNA, NETs contain histones, serine proteases, cytoskeletal proteins and antimicrobial peptides, all of which have immunomodulatory properties that can augment or decrease the inflammatory response. Extracellular localization of these molecules alerts the immune system of cellular damage, which is triggered by recognition of damage-associated molecular patterns (DAMPs) through specific pattern recognition receptors. However, not all of these molecules are DAMPs and may have other immunophysiological properties in the extracellular space. The release of NETs can lead to production of pro-inflammatory cytokines (due to TLR2/4/9 and inflammasome activation), the destruction of the extracellular matrix, activation of serine proteases and of matrix metallopeptidases (MMPs), modulation of cellular proliferation, induction of cellular migration and adhesion, promotion of thrombogenesis and angiogenesis and disruption of epithelial and endothelial permeability. Understanding the dynamics of NET-associated molecules, either individually or synergically, will help to unravel their role in inflammatory events and open novel perspectives for potential therapeutic targets. We here review molecules contained within NETS and their immunophysiological roles.
Collapse
Affiliation(s)
- Abraham U Morales-Primo
- Laboratory of Immunoparasitology, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City, Mexico
| | - Ingeborg Becker
- Laboratory of Immunoparasitology, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City, Mexico
| | - Jaime Zamora-Chimal
- Laboratory of Immunoparasitology, Unidad de Investigación en Medicina Experimental, Facultad de Medicina, Universidad Nacional Autónoma de México, Hospital General de México, Mexico City, Mexico
| |
Collapse
|
24
|
Park Y, Ban TH, Kim HD, Ko EJ, Lee J, Kim SC, Park CW, Yang CW, Kim YS, Chung BH. Mortality prediction of serum neutrophil gelatinase-associated lipocalin in patients requiring continuous renal replacement therapy. Korean J Intern Med 2021; 36:392-400. [PMID: 32623875 PMCID: PMC7969065 DOI: 10.3904/kjim.2019.446] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 02/04/2020] [Accepted: 02/05/2020] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND/AIMS We investigated whether serum neutrophil gelatinase-associated lipocalin (NGAL) can predict mortality in patients with acute kidney injury (AKI) requiring continuous renal replacement therapy (CRRT). METHODS This study enrolled 169 patients who underwent serum NGAL testing at CRRT initiation from June 2017 to January 2019. The predictive power of serum NGAL level for 28-day mortality was compared to the Acute Physiology and Chronic Health Evaluation-II (APACHE-II) score and Sequential Organ Failure Assessment (SOFA) score via area under the receiver operating characteristic curve (AuROC) value. RESULTS There were 55 survivors and 114 non-survivors at 28 days post-CRRT initiation. Median serum NGAL level was significantly higher in the non-survivor group than in the survivor group (743.0 ng/mL vs. 504.0 ng/mL, p = 0.003). The AuROC value of serum NGAL level was 0.640, which was lower than APACHEII score and SOFA score values (0.767 and 0.715, respectively). However, in the low APACHE-II score group (< 27.5), AuROC value of serum NGAL was significantly increased (0.698), and it was an independent risk factor for 28 day-mortality (hazard ratio, 2.405; 95% confidence interval, 1.209 to 4.783; p = 0.012). CONCLUSION In patients with AKI requiring CRRT, serum NGAL levels may be useful for predicting short-term mortality in those with low APACHE-II scores.
Collapse
Affiliation(s)
- Yohan Park
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Tae Hyun Ban
- Division of Nephrology, Department of Internal Medicine, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyung Duk Kim
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Eun Jeong Ko
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jongmin Lee
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seok Chan Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Cheol Whee Park
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Chul Woo Yang
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Yong-Soo Kim
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Byung Ha Chung
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
25
|
Plasma lipocalin-2/NGAL is stable over 12 weeks and is not modulated by exercise or dieting. Sci Rep 2021; 11:4056. [PMID: 33603000 PMCID: PMC7893047 DOI: 10.1038/s41598-021-83472-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Accepted: 12/29/2020] [Indexed: 02/07/2023] Open
Abstract
Amongst other immune cells, neutrophils play a key role in systemic inflammation leading to cardiovascular disease and can release inflammatory factors, including lipocalin-2 (LCN2). LCN2 drives cardiac hypertrophy and plays a role in maladaptive remodelling of the heart and has been associated with renal injury. While lifestyle factors such as diet and exercise are known to attenuate low-grade inflammation, their ability to modulate plasma LCN2 levels is unknown. Forty-eight endurance athletes and 52 controls (18–55 years) underwent measurement for various cardiovascular health indicators, along with plasma LCN2 concentration. No significant difference in LCN2 concentration was seen between the two groups. LCN2 was a very weak predictor or absent from models describing blood pressures or predicting athlete status. In another cohort, 57 non-diabetic overweight or obese men and post-menopausal women who fulfilled Adult Treatment Panel III metabolic syndrome criteria were randomly allocated into either a control, modified Dietary Approaches to Stop Hypertension (DASH) diet, or DASH and exercise group. Pre- and post-intervention demographic, cardiovascular health indicators, and plasma LCN2 expression were measured in each individual. While BMI fell in intervention groups, LCN2 levels remained unchanged within and between all groups, as illustrated by strong correlations between LCN2 concentrations pre- and 12 weeks post-intervention (r = 0.743, P < 0.0001). This suggests that circulating LCN2 expression are stable over a period of at least 12 weeks and is not modifiable by diet and exercise.
Collapse
|
26
|
Gu Z, Huang Y, Yang F, Tang S, Sun J, Chen T, Liu F, Wu H, Wang J. The application of neutrophil gelatin-related lipid delivery protein in evaluation of renal function, nutrition, anemia and inflammation in patients with CKD. Nephrol Ther 2021; 17:35-41. [PMID: 33468440 DOI: 10.1016/j.nephro.2020.10.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 09/15/2020] [Accepted: 10/11/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To investigate the role of neutrophil gelatinase-associated lipocalin in the evaluation of renal function, nutrition, anemia and inflammation in patients with chronic kidney diseases. MATERIALS AND METHODS A total of 302 patients with chronic kidney diseases were selected, and their clinical data, blood neutrophil gelatinase-associated lipocalin levels, renal function, nutrition, anemia, inflammation and calcium, and phosphorus metabolism were analyzed. RESULT Serum neutrophil gelatinase-associated lipocalin level increased with the progression of chronic kidney diseases. Higher neutrophil gelatinase-associated lipocalin levels were observed in patients with chronic kidney diseases stage 3b compared with healthy individuals (P<0.05), while the patients with chronic kidney diseases stage 5 showed higher levels compared with other chronic kidney diseases stages (P<0.01). Moreover, the ROC curve showed that neutrophil gelatinase-associated lipocalin had a better diagnostic performance from the chronic kidney diseases stage 3b to 5 (P<0.05). In addition, the serum neutrophil gelatinase-associated lipocalin levels in patient with chronic kidney diseases were negatively correlated with body mass index, number of red blood cells, hemoglobin, transferrin, the estimatedglomerular filtration rate (eGFR), serum calcium (P<0.01); and were positively correlated with mean arterial blood pressure, blood BUN, SCr and alpha 1 microglobulin, beta 2 microglobulin, urinary inhibition C, homocysteine, PTH levels, neutrophils ratio, free serum ferritin and c-reactive protein (P<0.01); while no significant correlation was found with gender, and age (P>0.05). CONCLUSION Serum neutrophil gelatinase-associated lipocalin levels are closely related to renal function injury, inflammatory response and anemia-related indicators in patients with chronic kidney diseases, and thus could be used as a diagnostic biomarker for evaluating the degree of renal injury and related complications in patients with chronic kidney diseases.
Collapse
Affiliation(s)
- Zhoupeng Gu
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Yan Huang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Fengjie Yang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Shiqi Tang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Juan Sun
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Tianru Chen
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Fang Liu
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Hong Wu
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China; Department of anesthesiology, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China
| | - Jianwen Wang
- Department of Nephropathy, Third Xiangya Hospital of Central South University, 410013 Changsha, Hunan Province, China.
| |
Collapse
|
27
|
Al-Absi B, AL-Habori M, Saif-Ali R. Plasma Lipocalin-2 and Adiponectin are Affected by Obesity Rather Than Type 2 Diabetes Mellitus per se. Diabetes Metab Syndr Obes 2021; 14:4547-4556. [PMID: 34815681 PMCID: PMC8605802 DOI: 10.2147/dmso.s338254] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 11/03/2021] [Indexed: 04/20/2023] Open
Abstract
PURPOSE Changes in plasma adipocytokines and inflammatory markers in type 2 DM remain controversial as to whether they are due to obesity or directly associated with the diabetic state. Our objective was to study the effect of obesity and diabetes on plasma lipocalin-2 (LCN2), adiponectin, and interleukin-1β (IL-1β) by comparing their levels in non-diabetic obese subjects and non-obese type 2 DM patients, as well as determining the association of these adipocytokines with metabolic syndrome factors and diabetic parameters. PATIENTS AND METHODS In this study, 85 Yemeni male volunteers aged 30-60 years old were enrolled, 25 of whom were healthy subjects with BMI < 25 kg/m2 served as control; 30 non-diabetic obese subjects (BMI ≥ 30 kg/m2 and FBG < 6.1 mmol/l); and 30 non-obese type 2 DM patients (BMI < 25 kg/m2 and FBG > 7 mmol/l). RESULTS Lipocalin-2 and adiponectin were significantly (p = 0.043 and p = 0.034) lower in non-diabetic obese subjects by 16.2% and 29.7% with respect to control group, with no effect in the non-obese type 2 DM patients. Moreover, LCN2 was significantly (p = 0.04) lower in the non-diabetic obese subjects by 15.8% as compared with the non-obese type 2 DM patients, with no significant difference in adiponectin levels. In contrast, serum IL-1β was significantly (p = 0.001 and p = 0.003) higher in both non-diabetic obese subjects and the non-obese type 2 DM patients by 76.5% and 67.7% as compared to control group. The significant decrease in both LCN2 and adiponectin and the significant increase in IL-1β in the non-diabetic obese subjects disappeared upon adjustment for waist circumference (WC). In contrast, the significant increase in IL-1β in the non-obese Type 2 DM patients was not affected upon adjustment for WC. CONCLUSION Plasma LCN2 and adiponectin were not affected by diabetes per se, suggesting that the observed changes in LCN2 and adiponectin in type 2 DM may be due to obesity rather than the diabetic state, whereas IL-1β levels were affected by both obesity and diabetes.
Collapse
Affiliation(s)
- Boshra Al-Absi
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
| | - Molham AL-Habori
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
- Correspondence: Molham AL-Habori Email
| | - Riyadh Saif-Ali
- Department of Biochemistry and Molecular Biology, Faculty of Medicine and Health Sciences, University of Sana`a, Sana`a, Yemen
| |
Collapse
|
28
|
Willey JS, Britten RA, Blaber E, Tahimic CG, Chancellor J, Mortreux M, Sanford LD, Kubik AJ, Delp MD, Mao XW. The individual and combined effects of spaceflight radiation and microgravity on biologic systems and functional outcomes. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, TOXICOLOGY AND CARCINOGENESIS 2021; 39:129-179. [PMID: 33902391 PMCID: PMC8274610 DOI: 10.1080/26896583.2021.1885283] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Both microgravity and radiation exposure in the spaceflight environment have been identified as hazards to astronaut health and performance. Substantial study has been focused on understanding the biology and risks associated with prolonged exposure to microgravity, and the hazards presented by radiation from galactic cosmic rays (GCR) and solar particle events (SPEs) outside of low earth orbit (LEO). To date, the majority of the ground-based analogues (e.g., rodent or cell culture studies) that investigate the biology of and risks associated with spaceflight hazards will focus on an individual hazard in isolation. However, astronauts will face these challenges simultaneously Combined hazard studies are necessary for understanding the risks astronauts face as they travel outside of LEO, and are also critical for countermeasure development. The focus of this review is to describe biologic and functional outcomes from ground-based analogue models for microgravity and radiation, specifically highlighting the combined effects of radiation and reduced weight-bearing from rodent ground-based tail suspension via hind limb unloading (HLU) and partial weight-bearing (PWB) models, although in vitro and spaceflight results are discussed as appropriate. The review focuses on the skeletal, ocular, central nervous system (CNS), cardiovascular, and stem cells responses.
Collapse
Affiliation(s)
| | | | - Elizabeth Blaber
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute
| | | | | | - Marie Mortreux
- Department of Neurology, Harvard Medical School, Beth Israel Deaconess Medical Center
| | - Larry D. Sanford
- Department of Radiation Oncology, Eastern Virginia Medical School
| | - Angela J. Kubik
- Department of Biomedical Engineering, Rensselaer Polytechnic Institute
| | - Michael D. Delp
- Department of Nutrition, Food and Exercise Sciences, Florida State University
| | - Xiao Wen Mao
- Division of Biomedical Engineering Sciences (BMES), Department of Basic Sciences, Loma Linda University
| |
Collapse
|
29
|
Antimicrobial peptides: bridging innate and adaptive immunity in the pathogenesis of psoriasis. Chin Med J (Engl) 2020; 133:2966-2975. [PMID: 33237697 PMCID: PMC7752697 DOI: 10.1097/cm9.0000000000001240] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Antimicrobial peptides (AMPs) are small molecules produced by a myriad of cells and play important roles not only in protecting against infections and sustaining skin barrier homeostasis but also in contributing to immune dysregulation under pathological conditions. Recently, increasing evidence has indicated that AMPs, including cathelicidin (LL-37), human β-defensins, S100 proteins, lipocalin 2, and RNase 7, are highly expressed in psoriatic skin lesions. These peptides broadly regulate immunity by interacting with various immune cells and linking innate and adaptive immune responses during the progression of psoriasis. In this review, we summarize the recent findings regarding AMPs in the pathogenesis of psoriasis with a main focus on their immunomodulatory abilities.
Collapse
|
30
|
Shi Z, Yin Y, Li C, Ding H, Mu N, Wang Y, Jin S, Ma H, Liu M, Zhou J. Lipocalin-2-induced proliferative endoplasmic reticulum stress participates in Kawasaki disease-related pulmonary arterial abnormalities. SCIENCE CHINA-LIFE SCIENCES 2020; 64:1000-1012. [PMID: 32915407 DOI: 10.1007/s11427-019-1772-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 06/30/2020] [Indexed: 11/30/2022]
Abstract
Clinical cases have reported pulmonary arterial structural and functional abnormalities in patients with Kawasaki disease (KD); however, the underlying mechanisms are unclear. In this study, a KD rat model was established via the intraperitoneal injection of Lactobacillus casei cell wall extract (LCWE). The results showed that pulmonary arterial functional and structural abnormalities were observed in KD rats. Furthermore, proliferative endoplasmic reticulum stress (ER stress) was observed in the pulmonary arteries of KD rats. Notably, the level of lipocalin-2 (Lcn 2), a trigger factor of inflammation, was remarkably elevated in the plasma and lung tissues of KD rats; increased Lcn 2 levels following LCWE stimulation may result from polymorphonuclear neutrophils (PMNs). Correspondingly, in cultured pulmonary artery smooth muscle cells (PASMCs), Lcn 2 markedly augmented the cleavage and nuclear localization of activating transcription factor-6 (ATF6), upregulated the transcription of glucose regulated protein 78 (GRP78) and neurite outgrowth inhibitor (NOGO), and promoted PASMCs proliferation. However, proapoptotic C/EBP homologous protein (CHOP) and caspase 12 levels were not elevated. Treatment with 4-phenyl butyric acid (4-PBA, a specific inhibitor of ER stress) inhibited PASMCs proliferation induced by Lcn 2 and attenuated pulmonary arterial abnormalities and right ventricular hypertrophy and reduced right ventricular systolic pressure in KD rats. In conclusion, Lcn 2 remarkably facilitates proliferative ER stress in PASMCs, which probably accounts for KD-related pulmonary arterial abnormalities.
Collapse
Affiliation(s)
- Zhaoling Shi
- Department of Pediatrics, Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China.,Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Yue Yin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Chen Li
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Ding
- Department of Pediatrics, Second Affiliated Hospital of Shaanxi University of Chinese Medicine, Xianyang, 712000, China
| | - Nan Mu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Yishi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Shanshan Jin
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China
| | - Heng Ma
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China.
| | - Manling Liu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jie Zhou
- Department of Endocrinology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
31
|
Schreinlechner M, Noflatscher M, Lener D, Bauer A, Kirchmair R, Marschang P, Theurl M. NGAL Correlates with Femoral and Carotid Plaque Volume Assessed by Sonographic 3D Plaque Volumetry. J Clin Med 2020; 9:jcm9092811. [PMID: 32878068 PMCID: PMC7565934 DOI: 10.3390/jcm9092811] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 08/27/2020] [Indexed: 12/20/2022] Open
Abstract
Background/Objectives: Inflammation represents a cornerstone in the development of atherosclerosis and early detection is essential to avoid cardiovascular events. Biomarkers like interleukin-1 beta, interleukin-6, or high sensitivity CRP (hs-CRP) have been investigated intensively in this field. Since they have several limitations, additional biomarkers are needed for cardiovascular risk stratification. The acute phase protein, neutrophil gelatinase-associated lipocalin (NGAL), modulates inflammation and is elevated in cardiovascular disease (CVD). Moreover, it contributes to plaque destabilization. Methods: In this prospective, single-center study, we included 323 asymptomatic patients with at least one cardiovascular risk factor or established CVD. NGAL levels were measured in plasma samples using a commercially available ELISA. Carotid, femoral, and total atherosclerotic plaque volumes (PV) were measured using a 3D ultrasound system (Philips iU22). Patients were separated into a low (n = 243) and high (n = 80) total PV group. Results: NGAL was significantly higher in patients with high total PV versus patients with low total PV. The NGAL amplitude for the prediction of high total PV was significantly higher when compared with hs-CRP. A high predictive value could also be observed for patients without established CVD. Conclusion: NGAL seems to be a promising biomarker for the identification of asymptomatic patients with atherosclerotic disease.
Collapse
Affiliation(s)
- Michael Schreinlechner
- Medical University of Innsbruck, University Hospital of Internal Medicine, Cardiology and Angiology, Anichstrasse 35, A-6020 Innsbruck, Austria; (M.S.); (M.N.); (D.L.); (A.B.); (R.K.); (P.M.)
| | - Maria Noflatscher
- Medical University of Innsbruck, University Hospital of Internal Medicine, Cardiology and Angiology, Anichstrasse 35, A-6020 Innsbruck, Austria; (M.S.); (M.N.); (D.L.); (A.B.); (R.K.); (P.M.)
| | - Daniela Lener
- Medical University of Innsbruck, University Hospital of Internal Medicine, Cardiology and Angiology, Anichstrasse 35, A-6020 Innsbruck, Austria; (M.S.); (M.N.); (D.L.); (A.B.); (R.K.); (P.M.)
| | - Axel Bauer
- Medical University of Innsbruck, University Hospital of Internal Medicine, Cardiology and Angiology, Anichstrasse 35, A-6020 Innsbruck, Austria; (M.S.); (M.N.); (D.L.); (A.B.); (R.K.); (P.M.)
| | - Rudolf Kirchmair
- Medical University of Innsbruck, University Hospital of Internal Medicine, Cardiology and Angiology, Anichstrasse 35, A-6020 Innsbruck, Austria; (M.S.); (M.N.); (D.L.); (A.B.); (R.K.); (P.M.)
| | - Peter Marschang
- Medical University of Innsbruck, University Hospital of Internal Medicine, Cardiology and Angiology, Anichstrasse 35, A-6020 Innsbruck, Austria; (M.S.); (M.N.); (D.L.); (A.B.); (R.K.); (P.M.)
- Central Hospital of Bolzano, Department of Internal Medicine, Via Lorenz Boehler 5, I-39100 Bolzano, Italy
| | - Markus Theurl
- Medical University of Innsbruck, University Hospital of Internal Medicine, Cardiology and Angiology, Anichstrasse 35, A-6020 Innsbruck, Austria; (M.S.); (M.N.); (D.L.); (A.B.); (R.K.); (P.M.)
- Correspondence:
| |
Collapse
|
32
|
Tidbury N, Browning N, Shaw M, Morgan M, Kemp I, Matata B. Neutrophil Gelatinase-associated Lipocalin as a Marker of Postoperative Acute Kidney Injury Following Cardiac Surgery in Patients with Preoperative Kidney Impairment. Cardiovasc Hematol Disord Drug Targets 2020; 19:239-248. [PMID: 30987577 DOI: 10.2174/1871529x19666190415115106] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2018] [Revised: 03/08/2019] [Accepted: 03/22/2019] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Acute kidney injury (AKI) is a serious complication of cardiac surgery. The current 'gold standard' for determining AKI is change in serum creatinine and urine output, however, this change occurs relatively late after the actual injury occurs. Identification of new biomarkers that detect early AKI is required. Recently, new biomarkers, such as the NephroCheck® Test and AKIRisk have also been tested and found to be good indicators of AKI. Neutrophil gelatinase-associated lipocalin (NGAL) has shown promise in paediatric patients but has displayed varied results in adult populations, particularly post cardiac surgery. The aim of this study was to assess the value of urinary NGAL as a biomarker of AKI in patients with pre-existing renal impairment (eGFR >15ml/min to eGFR<60ml/min). METHODS A post-hoc analysis of urinary NGAL concentrations from 125 patients with pre-existing kidney impairment, who participated in a randomised trial of haemofiltration during cardiac surgery, was undertaken. Urinary NGAL was measured using ELISA at baseline, post-operatively and 24 and 48 hours after surgery, and serum creatinine was measured pre and postoperatively and then at 24, 48, 72 and 96 hours as routine patient care. NGAL concentrations were compared in patients with and without AKI determined by changes in serum creatinine concentrations. A Kaplan-Meier plot compared survival for patients with or without AKI and a Cox proportional hazards analysis was performed to identify factors with the greatest influence on survival. RESULTS Following surgery, 43% of patients developed AKI (based on KDIGO definition). Baseline urinary NGAL was not found to be significantly different between patients that did and did not develop AKI. Urinary NGAL concentration was increased in all patients following surgery, regardless of whether they developed AKI and was also significant between groups at 24 (p=0.003) and 48 hours (p<0.0001). Urinary NGAL concentrations at 48 hours correlated with serum creatinine concentrations at 48 hours (r=0.477, p<0.0001), 72 hours (r=0.488, p<0.0001) and 96 hours (r=0.463, p<0.0001). Urinary NGAL at 48 hours after surgery strongly predicted AKI (AUC=0.76; P=0.0001). A Kaplan- Meier plot showed that patients with postoperative AKI had a significantly lower 7-year survival compared with those without AKI. Postoperative urinary NGAL at 48 hours >156ng/mL also strongly predicted 7-year survival. However, additive EuroSCORE, age, current smoking and post-operative antibiotics usage were distinctly significantly more predictive of 7-year survival as compared with postoperative urinary NGAL at 48 hours >156ng/mL. CONCLUSIONS Our study demonstrated that postoperative urinary NGAL levels at 48 hours postsurgery strongly predicts the onset or severity of postoperative AKI based on KDIGO classification in patients with preoperative kidney impairment and were also strongly related to 7-year survival.
Collapse
Affiliation(s)
- N Tidbury
- Liverpool Heart & Chest Hospital NHS Foundation Trust, University of Liverpool, Liverpool L14 3PE, United Kingdom
| | - N Browning
- Liverpool Heart & Chest Hospital NHS Foundation Trust, University of Liverpool, Liverpool L14 3PE, United Kingdom
| | - M Shaw
- Liverpool Heart & Chest Hospital NHS Foundation Trust, University of Liverpool, Liverpool L14 3PE, United Kingdom
| | - M Morgan
- Liverpool Heart & Chest Hospital NHS Foundation Trust, University of Liverpool, Liverpool L14 3PE, United Kingdom
| | - I Kemp
- Liverpool Heart & Chest Hospital NHS Foundation Trust, University of Liverpool, Liverpool L14 3PE, United Kingdom
| | - B Matata
- Liverpool Heart & Chest Hospital NHS Foundation Trust, University of Liverpool, Liverpool L14 3PE, United Kingdom.,Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool L14 3PE, United Kingdom
| |
Collapse
|
33
|
Dendritic cells are crucial for cardiovascular remodeling and modulate neutrophil gelatinase-associated lipocalin expression upon mineralocorticoid receptor activation. J Hypertens 2020; 37:1482-1492. [PMID: 31033725 DOI: 10.1097/hjh.0000000000002067] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
BACKGROUND Adaptive immunity is crucial in cardiovascular and renal inflammation/fibrosis upon hyperactivation of mineralocorticoid receptor. We have previously demonstrated that dendritic cells can respond to mineralocorticoid receptor activation, and the neutrophil gelatinase-associated lipocalin (NGAL) in dendritic cells is highly increased during aldosterone (Aldo)/mineralocorticoid receptor-dependent cardiovascular damage. However, the interrelationship among dendritic cells, target organs inflammation/fibrosis induced by mineralocorticoid receptor, and NGAL-dependence remains unknown. OBJECTIVE We studied the role of dendritic cells in mineralocorticoid receptor-dependent tissue remodeling and whether NGAL can modulate the inflammatory response of dendritic cells after mineralocorticoid receptor activation. METHODS Cardiovascular and renal remodeling induced by Aldo and high-salt diet [nephrectomy-Aldo-salt (NAS) model] were analyzed in CD11c.DOG mice, a model which allows dendritic cells ablation by using diphtheria toxin. In addition, in-vitro studies in NGAL-knock out dendritic cells were performed to determine the immunomodulatory role of NGAL upon Aldo treatment. RESULTS The ablation of dendritic cells prevented the development of cardiac hypertrophy, perivascular fibrosis, and the overexpression of NGAL, brain natriuretic peptide, and two profibrotic factors induced by NAS: collagen 1A1 and connective tissue growth factor. We determined that dendritic cells were not required to prevent renal hypertrophy/fibrosis induced by NAS. Between different immune cells analyzed, we observed that NGAL abundance was higher in antigen-presenting cells, while in-vitro studies showed that mineralocorticoid receptor stimulation in dendritic cells favored NGAL and IL-23 expression (p19 and p40 subunits), which are involved in the development of fibrosis and the Th17-driven response, respectively. CONCLUSION NGAL produced by dendritic cells may play a pivotal role in the activation of adaptive immunity that leads to cardiovascular fibrosis during mineralocorticoids excess.
Collapse
|
34
|
Lin X, Onda DA, Yang CH, Lewis JR, Levinger I, Loh K. Roles of bone-derived hormones in type 2 diabetes and cardiovascular pathophysiology. Mol Metab 2020; 40:101040. [PMID: 32544571 PMCID: PMC7348059 DOI: 10.1016/j.molmet.2020.101040] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/28/2020] [Accepted: 06/11/2020] [Indexed: 02/07/2023] Open
Abstract
Background Emerging evidence demonstrates that bone is an endocrine organ capable of influencing multiple physiological and pathological processes through the secretion of hormones. Recent research suggests complex crosstalk between the bone and other metabolic and cardiovascular tissues. It was uncovered that three of these bone-derived hormones—osteocalcin, lipocalin 2, and sclerostin—are involved in the endocrine regulations of cardiometabolic health and play vital roles in the pathophysiological process of developing cardiometabolic syndromes such as type 2 diabetes and cardiovascular disease. Chronic low-grade inflammation is one of the hallmarks of cardiometabolic diseases and a major contributor to disease progression. Novel evidence also implicates important roles of bone-derived hormones in the regulation of chronic inflammation. Scope of review In this review, we provide a detailed overview of the physiological and pathological roles of osteocalcin, lipocalin 2, and sclerostin in cardiometabolic health regulation and disease development, with a focus on the modulation of chronic inflammation. Major conclusions Evidence supports that osteocalcin has a protective role in cardiometabolic health, and an increase of lipocalin 2 contributes to the development of cardiometabolic diseases partly via pro-inflammatory effects. The roles of sclerostin appear to be complicated: It exerts pro-adiposity and pro-insulin resistance effects in type 2 diabetes and has an anti-calcification effect during cardiovascular disease. A better understanding of the actions of these bone-derived hormones in the pathophysiology of cardiometabolic diseases will provide crucial insights to help further research develop new therapeutic strategies to treat these diseases.
Collapse
Affiliation(s)
- Xuzhu Lin
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia.
| | - Danise-Ann Onda
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Chieh-Hsin Yang
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia
| | - Joshua R Lewis
- School of Medical and Health Sciences, Edith Cowan University, Perth, Australia; Medical School, University of Western Australia, Perth, Australia
| | - Itamar Levinger
- Institute for Health and Sport (IHES), Victoria University, Footscray, VIC, Australia; Australian Institute for Musculoskeletal Science (AIMSS), University of Melbourne and Western Health, St Albans, VIC, Australia
| | - Kim Loh
- St. Vincent's Institute of Medical Research, Fitzroy, VIC, Australia; Department of Medicine, University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
35
|
Gao X, Wang WZ, Xiao QM, Qi HN, Zhu BY, Li BY, Wang P, Han YY. Correlation between neutrophil gelatinase-associated lipocalin and soluble CD14 subtype on the prognosis evaluation of acute paraquat poisoning patients. Hum Exp Toxicol 2020; 39:402-410. [PMID: 31957486 DOI: 10.1177/0960327119897111] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
OBJECTIVE The objective of this article is to study the correlation between neutrophil gelatinase-associated lipocalin (NGAL) and soluble CD14 subtype (presepsin) on the severity and prognosis evaluation of acute paraquat poisoning (APP) patients. MATERIALS AND METHODS We studied 120 APP patients who were divided into three groups: light (28 cases), moderate (52 cases), and heavy poisoning (40 cases) groups. Twenty healthy volunteers were enrolled as controls. RESULTS Acute kidney injury (AKI) occurred in 86 APP patients (71.7%, 86 of 120). In AKI group, urine NGAL was elevated 3 h after treatment, serum NGAL was elevated 24 h after treatment, and serum creatine (SCr) was elevated 2 days after treatment, which were all significantly higher than non-AKI group. Compared with control group, there were significant differences in presepsin and acute physiology and chronic health status (APACHE) II score of different poisoning groups. There were significant differences in detection indices 24 h, 3 days, and 7 days after treatment among different poisoning groups. There was a positive correlation between urine NGAL and serum paraquat concentration, urine NGAL, and AKI morbidity (r 1 = 0.974, r 2 = 0.766, p < 0.001), suggesting higher urine NGAL level indicated higher AKI morbidity. Receiver operating characteristic curves analysis suggested serum presepsin level and urine NGAL level had higher sensitivity and specificity than APACHE II score when predicting 28-day mortality of APP patients. CONCLUSION Serum and urine NGAL level is elevated earlier than SCr, which is important for the early diagnosis of APP. Serum presepsin and urine NGAL levels can be used as markers to diagnose the severity of AKI and predict the mortality of APP patients.
Collapse
Affiliation(s)
- X Gao
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China.,Both the authors contributed equally to this work
| | - W-Z Wang
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China.,Both the authors contributed equally to this work
| | - Q-M Xiao
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| | - H-N Qi
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| | - B-Y Zhu
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| | - B-Y Li
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| | - P Wang
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| | - Y-Y Han
- Department of Emergency, Harrison International Peace Hospital Affiliated to Hebei Medical University, Hebei, China
| |
Collapse
|
36
|
Hodrea J, Balogh DB, Hosszu A, Lenart L, Besztercei B, Koszegi S, Sparding N, Genovese F, Wagner LJ, Szabo AJ, Fekete A. Reduced O-GlcNAcylation and tubular hypoxia contribute to the antifibrotic effect of SGLT2 inhibitor dapagliflozin in the diabetic kidney. Am J Physiol Renal Physiol 2020; 318:F1017-F1029. [PMID: 32116017 PMCID: PMC7242633 DOI: 10.1152/ajprenal.00021.2020] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Diabetic kidney disease is a worldwide epidemic, and therapies are incomplete. Clinical data suggest that improved renal outcomes by Na+-glucose cotransporter 2 inhibitor (SGLT2i) are partly beyond their antihyperglycemic effects; however, the mechanisms are still elusive. Here, we investigated the effect of the SGLT2i dapagliflozin (DAPA) in the prevention of elevated O-GlcNAcylation and tubular hypoxia as contributors of renal fibrosis. Type 1 diabetes was induced by streptozotocin in adult male Wistar rats. After the onset of diabetes, rats were treated for 6 wk with DAPA or DAPA combined with losartan (LOS). The effect of hyperglycemia was tested in HK-2 cells kept under normal or high glucose conditions. To test the effect of hypoxia, cells were kept in 1% O2 for 2 h. Cells were treated with DAPA or DAPA combined with LOS. DAPA slowed the loss of renal function, mitigated renal tubular injury markers (kidney injury molecule-1 and neutrophil gelatinase-associated lipocalin), and reduced tubulointerstitial fibrosis. DAPA diminished high glucose-induced protein O-GlcNAcylation and moderated the tubular response to hypoxia through the hypoxia-inducible factor pathway. DAPA alone was as effective as combined treatment with LOS in all outcome parameters. These data highlight the role of ameliorated O-GlcNAcylation and diminished tubular hypoxia as important benefits of SGLT2i treatment. Our results support the link between glucose toxicity, tubular hypoxia, and fibrosis, a vicious trio that could be targeted by SGLT2i in kidney diseases of other origins as well.
Collapse
Affiliation(s)
- Judit Hodrea
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Dora B Balogh
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Adam Hosszu
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| | - Lilla Lenart
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Balazs Besztercei
- Institute of Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Sandor Koszegi
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences, Budapest, Hungary
| | - Nadja Sparding
- Nordic Bioscience, Biomarkers & Research, Herlev, Denmark.,Biomedical Sciences, Faculty of Health and Medical Science, University of Copenhagen, Copenhagen N, Denmark
| | | | - Laszlo J Wagner
- Department of Transplantation and Surgery, Semmelweis University, Budapest, Hungary
| | - Attila J Szabo
- 1st Department of Pediatrics, Semmelweis University, Budapest, Hungary.,MTA-SE Paediatrics and Nephrology Research Group, Hungarian Academy of Sciences and Semmelweis University, Budapest, Hungary
| | - Andrea Fekete
- MTA-SE "Lendület" Diabetes Research Group, Hungarian Academy of Sciences, Budapest, Hungary.,1st Department of Pediatrics, Semmelweis University, Budapest, Hungary
| |
Collapse
|
37
|
Chong JJH, Prince RL, Thompson PL, Thavapalachandran S, Ooi E, Devine A, Lim EEM, Byrnes E, Wong G, Lim WH, Lewis JR. Association Between Plasma Neutrophil Gelatinase-Associated Lipocalin and Cardiac Disease Hospitalizations and Deaths in Older Women. J Am Heart Assoc 2020; 8:e011028. [PMID: 30595080 PMCID: PMC6405726 DOI: 10.1161/jaha.118.011028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Background Neutrophil gelatinase‐associated lipocalin (NGAL) or lipocalin 2 may promote atherosclerosis and plaque instability leading to increased risk of cardiac events. We investigated the relationships between plasma NGAL, cardiovascular disease biomarkers, and long‐term cardiac events. Methods and Results The study population consisted of 1131 ambulant older white women (mean age 75 years) without clinical coronary heart disease (CHD) and measures of plasma NGAL in the Perth Longitudinal Study of Ageing Women with 14.5‐year CHD and heart failure hospitalizations or death (events) captured using linked records. Over 14.5 years, 256 women had CHD events, while 118 had heart failure events. Per SD increase in log‐transformed NGAL there was a 35% to 37% increase in relative hazards for CHD and heart failure events in unadjusted analyses, which remained significant after adjustment for conventional risk factors for CHD events (hazard ratio 1.29, 95% CI 1.13–1.48, P<0.001) but not heart failure (P>0.05). Women in the highest 2 quartiles of NGAL had higher relative hazards for CHD events compared with women in the lowest quartile hazard ratio 1.61, 95% CI 1.08–2.39, P=0.019 and hazard ratio 1.97, 95% CI 1.33–3.93, P=0.001, respectively. These associations were independent of high‐sensitivity cardiac troponin I, homocysteine, and estimated renal function. NGAL correctly reclassified 1 in 4 women who sustained a CHD event up in risk and 1 in 10 women without CHD events down in risk. Conclusions NGAL was associated with increased risk of long‐term CHD events, independent of conventional risk factors and biomarkers. These findings provide mechanistic insight into the role of NGAL with cardiac events.
Collapse
Affiliation(s)
- James J H Chong
- 1 Centre for Heart Research Westmead Institute for Medical Research The University of Sydney Westmead New South Wales Australia.,2 Department of Cardiology Westmead Hospital Westmead New South Wales Australia.,3 Sydney Medical School The University of Sydney Sydney New South Wales Australia
| | - Richard L Prince
- 4 Medical School University of Western Australia Perth Australia.,6 Department of Endocrinology and Diabetes Sir Charles Gairdner Hospital Perth Australia
| | - Peter L Thompson
- 7 Department of Cardiology Sir Charles Gairdner Hospital Perth Australia
| | - Sujitha Thavapalachandran
- 1 Centre for Heart Research Westmead Institute for Medical Research The University of Sydney Westmead New South Wales Australia
| | - Esther Ooi
- 4 Medical School University of Western Australia Perth Australia.,5 School of Biomedical Sciences University of Western Australia Perth Australia
| | - Amanda Devine
- 10 School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia
| | - E E M Lim
- 9 PathWest Sir Charles Gairdner Hospital Perth Australia
| | | | - Germaine Wong
- 11 Centre for Kidney Research Children's Hospital at Westmead School of Public Health Sydney Medical School The University of Sydney Sydney Australia
| | - Wai H Lim
- 8 Department of Renal Medicine Sir Charles Gairdner Hospital Perth Australia
| | - Joshua R Lewis
- 4 Medical School University of Western Australia Perth Australia.,10 School of Medical and Health Sciences Edith Cowan University Joondalup Western Australia Australia.,11 Centre for Kidney Research Children's Hospital at Westmead School of Public Health Sydney Medical School The University of Sydney Sydney Australia
| |
Collapse
|
38
|
Swaminathan G, Krishnamurthy VK, Sridhar S, Robson DC, Ning Y, Grande-Allen KJ. Hypoxia Stimulates Synthesis of Neutrophil Gelatinase-Associated Lipocalin in Aortic Valve Disease. Front Cardiovasc Med 2019; 6:156. [PMID: 31737648 PMCID: PMC6828964 DOI: 10.3389/fcvm.2019.00156] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022] Open
Abstract
Objective: Aortic valve disease is commonly found in the elderly population. It is characterized by dysregulated extracellular matrix remodeling followed by extensive microcalcification of the aortic valve and activation of valve interstitial cells. The mechanism behind these events are largely unknown. Studies have reported expression of hypoxia inducible factor-1 alpha (HIF1α) in calcific nodules in aortic valve disease, therefore we investigated the effect of hypoxia on extracellular matrix remodeling in aged aortic valves. Approach and Results: Western blotting revealed elevated expression of HIF1α and the complex of matrix metalloprotease 9 (MMP9) and neutrophil gelatinase-associated lipocalin (NGAL) in aged porcine aortic valves cultured under hypoxic conditions. Consistently, immunofluorescence staining showed co-expression of MMP9 and NGAL in the fibrosa layer of these porcine hypoxic aortic valves. Gelatinase zymography demonstrated that the activity of MMP9-NGAL complex was significantly increased in aortic valves in 13% O2 compared to 20% O2. Importantly, the presence of ectopic elastic fibers in the fibrosa of hypoxic aortic valves, also detected in human diseased aortic valves, suggests altered elastin homeostasis due to hypoxia. Conclusion: This study demonstrates that hypoxia stimulates pathological extracellular matrix remodeling via expression of NGAL and MMP9 by valve interstitial cells.
Collapse
|
39
|
Heterogenous Renal Injury Biomarker Production Reveals Human Sepsis-Associated Acute Kidney Injury Subtypes. Crit Care Explor 2019; 1:e0047. [PMID: 32166228 PMCID: PMC7063889 DOI: 10.1097/cce.0000000000000047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Supplemental Digital Content is available in the text. To identify mechanisms associated with sepsis-acute kidney injury based on the expression levels of renal injury biomarkers, neutrophil gelatinase–associated lipocalin, and kidney injury molecule-1 in renal biopsies which may allow the identification of sepsis-acute kidney injury patient subtypes.
Collapse
|
40
|
Chella Krishnan K, Sabir S, Shum M, Meng Y, Acín-Pérez R, Lang JM, Floyd RR, Vergnes L, Seldin MM, Fuqua BK, Jayasekera DW, Nand SK, Anum DC, Pan C, Stiles L, Péterfy M, Reue K, Liesa M, Lusis AJ. Sex-specific metabolic functions of adipose Lipocalin-2. Mol Metab 2019; 30:30-47. [PMID: 31767179 PMCID: PMC6812340 DOI: 10.1016/j.molmet.2019.09.009] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 12/25/2022] Open
Abstract
OBJECTIVE Lipocalin-2 (LCN2) is a secreted protein involved in innate immunity and has also been associated with several cardiometabolic traits in both mouse and human studies. However, the causal relationship of LCN2 to these traits is unclear, and most studies have examined only males. METHODS Using adeno-associated viral vectors we expressed LCN2 in either adipose or liver in a tissue specific manner on the background of a whole-body Lcn2 knockout or wildtype mice. Metabolic phenotypes including body weight, body composition, plasma and liver lipids, glucose homeostasis, insulin resistance, mitochondrial phenotyping, and metabolic cage studies were monitored. RESULTS We studied the genetics of LCN2 expression and associated clinical traits in both males and females in a panel of 100 inbred strains of mice (HMDP). The natural variation in Lcn2 expression across the HMDP exhibits high heritability, and genetic mapping suggests that it is regulated in part by Lipin1 gene variation. The correlation analyses revealed striking tissue dependent sex differences in obesity, insulin resistance, hepatic steatosis, and dyslipidemia. To understand the causal relationships, we examined the effects of expression of LCN2 selectively in liver or adipose. On a Lcn2-null background, LCN2 expression in white adipose promoted metabolic disturbances in females but not males. It acted in an autocrine/paracrine manner, resulting in mitochondrial dysfunction and an upregulation of inflammatory and fibrotic genes. On the other hand, on a null background, expression of LCN2 in liver had no discernible impact on the traits examined despite increasing the levels of circulating LCN2 more than adipose LCN2 expression. The mechanisms underlying the sex-specific action of LCN2 are unclear, but our results indicate that adipose LCN2 negatively regulates its receptor, LRP2 (or megalin), and its repressor, ERα, in a female-specific manner and that the effects of LCN2 on metabolic traits are mediated in part by LRP2. CONCLUSIONS Following up on our population-based studies, we demonstrate that LCN2 acts in a highly sex- and tissue-specific manner in mice. Our results have important implications for human studies, emphasizing the importance of sex and the tissue source of LCN2.
Collapse
Affiliation(s)
| | - Simon Sabir
- Department of Psychology, University of California, Los Angeles, CA, USA
| | - Michaël Shum
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Yonghong Meng
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Rebeca Acín-Pérez
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Jennifer M Lang
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Raquel R Floyd
- Department of Biology, University of California, Los Angeles, CA, USA
| | - Laurent Vergnes
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Marcus M Seldin
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Brie K Fuqua
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Dulshan W Jayasekera
- Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA
| | - Sereena K Nand
- Department of Biology, University of California, Los Angeles, CA, USA
| | - Diana C Anum
- Department of Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Calvin Pan
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA
| | - Linsey Stiles
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA
| | - Miklós Péterfy
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA; Department of Basic Medical Sciences, Western University of Health Sciences, Pomona, CA, USA
| | - Karen Reue
- Department of Human Genetics, University of California, Los Angeles, CA, USA
| | - Marc Liesa
- Department of Medicine/Division of Endocrinology, University of California, Los Angeles, CA, USA; Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Aldons J Lusis
- Department of Medicine/Division of Cardiology, University of California, Los Angeles, CA, USA; Department of Human Genetics, University of California, Los Angeles, CA, USA; Department of Microbiology, Immunology and Molecular Genetics, University of California, Los Angeles, CA, USA.
| |
Collapse
|
41
|
Millar SA, Anderson SI, O'Sullivan SE. Osteokines and the vasculature: a review of the in vitro effects of osteocalcin, fibroblast growth factor-23 and lipocalin-2. PeerJ 2019; 7:e7139. [PMID: 31372314 PMCID: PMC6660824 DOI: 10.7717/peerj.7139] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 05/17/2019] [Indexed: 12/16/2022] Open
Abstract
Bone-derived factors that demonstrate extra-skeletal functions, also termed osteokines, are fast becoming a highly interesting and focused area of cross-disciplinary endocrine research. Osteocalcin (OCN), fibroblast growth factor-23 (FGF23) and lipocalin-2 (LCN-2), produced in bone, comprise an important endocrine system that is finely tuned with other organs to ensure homeostatic balance and health. This review aims to evaluate in vitro evidence of the direct involvement of these proteins in vascular cells and whether any causal roles in cardiovascular disease or inflammation can be supported. PubMed, Medline, Embase and Google Scholar were searched for relevant research articles investigating the exogenous addition of OCN, FGF23 or LCN-2 to vascular smooth muscle or endothelial cells. Overall, these osteokines are directly vasoactive across a range of human and animal vascular cells. Both OCN and FGF23 have anti-apoptotic properties and increase eNOS phosphorylation and nitric oxide production through Akt signalling in human endothelial cells. OCN improves intracellular insulin signalling and demonstrates protective effects against endoplasmic reticulum stress in murine and human endothelial cells. OCN may be involved in calcification but further research is warranted, while there is no evidence for a pro-calcific effect of FGF23 in vitro. FGF23 and LCN-2 increase proliferation in some cell types and increase and decrease reactive oxygen species generation, respectively. LCN-2 also has anti-apoptotic effects but may increase endoplasmic reticulum stress as well as have pro-inflammatory and pro-angiogenic properties in human vascular endothelial and smooth muscle cells. There is no strong evidence to support a pathological role of OCN or FGF23 in the vasculature based on these findings. In contrast, they may in fact support normal endothelial functioning, vascular homeostasis and vasodilation. No studies examined whether OCN or FGF23 may have a role in vascular inflammation. Limited studies with LCN-2 indicate a pro-inflammatory and possible pathological role in the vasculature but further mechanistic data is required. Overall, these osteokines pose intriguing functions which should be investigated comprehensively to assess their relevance to cardiovascular disease and health in humans.
Collapse
Affiliation(s)
- Sophie A Millar
- Division of Graduate Entry Medicine and Medical Sciences, School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
| | - Susan I Anderson
- Division of Graduate Entry Medicine and Medical Sciences, School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
| | - Saoirse E O'Sullivan
- Division of Graduate Entry Medicine and Medical Sciences, School of Medicine, Royal Derby Hospital, University of Nottingham, Derby, United Kingdom
| |
Collapse
|
42
|
Eilenberg W, Stojkovic S, Kaider A, Piechota-Polanczyk A, Nanobachvili J, Domenig CM, Wojta J, Huk I, Demyanets S, Neumayer C. Neutrophil Gelatinase Associated Lipocalin (NGAL) for Identification of Unstable Plaques in Patients with Asymptomatic Carotid Stenosis. Eur J Vasc Endovasc Surg 2019; 57:768-777. [PMID: 31164272 DOI: 10.1016/j.ejvs.2018.12.029] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 12/26/2018] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Neutrophil gelatinase associated lipocalin (NGAL) and matrix metalloproteinase (MMP)-9/NGAL complex were investigated in asymptomatic patients with carotid artery stenosis including gender specific differences aiming at vulnerable plaques prone to embolisation. METHODS Serum NGAL and MMP-9/NGAL levels were analysed in 83 patients with asymptomatic carotid artery stenosis. Pre-operative ultrasound and post-endarterectomy histology of carotid atherosclerotic lesions were evaluated. RESULTS Patients with vulnerable plaques, as determined by ultrasound (plaques with decreased echogenicity) and histological analysis (type VI according to the classification of the American Heart Association), displayed the highest levels of NGAL and MMP-9/NGAL complex (p = .0003 and p = .0078, respectively). Grade VI plaques were primarily detected in patients with "soft" plaques (12 type VI plaques in 25 patients), but also in patients with mixed (four of 19) and calcified (three of 39) plaques according to ultrasound. Higher grade carotid artery stenosis (≥90%) was not associated with elevated NGAL levels. The receiver operating characteristic curve analysis detecting grade VI lesions yields an area under the curve (AUC) = 0.85, with respect to soft plaque on ultrasound the AUC = 0.86. There were no gender specific differences in levels of NGAL 80.9 (37.7) ng/mL in women vs. 76.7 (36.3) ng/mL in men, p = .607) nor of MMP-9/NGAL 33.0 (18.2-55.5) ng/mL in women vs. 36.7 (20.2-54.0) ng/mL in men, p = .969. Likewise, there were no gender associated differences in vulnerable plaque characteristics: either for grade VI plaques (17.9% vs. 27.3%, p = .582) or for the presence of soft plaques as evaluated by ultrasound (35.9% vs. 25%, p = .503). CONCLUSION Circulating NGAL and MMP-9/NGAL are significantly increased in asymptomatic patients with vulnerable carotid atherosclerotic plaques independent of gender. Accordingly, serum NGAL may be proposed as a valuable biomarker for the detection of unstable carotid plaques in asymptomatic patients, who can then be selected for early carotid endarterectomy or stenting.
Collapse
Affiliation(s)
- Wolf Eilenberg
- Department of Surgery, Division of Vascular Surgery, Medical University of Vienna, Austria
| | - Stefan Stojkovic
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria
| | - Alexandra Kaider
- Centre for Medical Statistics, Informatics, and Intelligent Systems, Medical University of Vienna, Austria
| | - Aleksandra Piechota-Polanczyk
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Josif Nanobachvili
- Department of Surgery, Division of Vascular Surgery, Medical University of Vienna, Austria
| | - Christoph M Domenig
- Department of Surgery, Division of Vascular Surgery, Medical University of Vienna, Austria
| | - Johann Wojta
- Department of Internal Medicine II, Division of Cardiology, Medical University of Vienna, Austria; Core Facilities, Medical University of Vienna, Vienna, Austria
| | - Ihor Huk
- Department of Surgery, Division of Vascular Surgery, Medical University of Vienna, Austria
| | - Svitlana Demyanets
- Department of Laboratory Medicine, Medical University of Vienna, Austria
| | - Christoph Neumayer
- Department of Surgery, Division of Vascular Surgery, Medical University of Vienna, Austria.
| |
Collapse
|
43
|
Stankovic M, Ljujic B, Babic S, Maravic-Stojkovic V, Mitrovic S, Arsenijevic N, Radak D, Pejnovic N, Lukic ML. IL-33/IL-33R in various types of carotid artery atherosclerotic lesions. Cytokine 2019; 120:242-250. [PMID: 31132589 DOI: 10.1016/j.cyto.2019.05.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 04/26/2019] [Accepted: 05/13/2019] [Indexed: 12/22/2022]
Abstract
OBJECTIVE Inflammation plays a crucial role in the progression of atherosclerotic plaques. The aim of the study was to investigate serum levels and expression of Interleukin-33 (IL-33) and ST2 receptor in atherosclerotic plaques and to analyze correlation with the type of the carotid plaques in patients with carotid disease. METHODS This study included 191 consecutive patients submitted for carotid endarterectomy (CEA). Preoperative serum levels of IL-33 and soluble ST2 (sST2) were measured. Atherosclerotic plaques obtained during surgery were initially histologically classified and immunohistochemical analyzes of IL-33, IL-33R, CD68 and alpha-SMA expression was performed. Ultrasound assessment of the level of carotid stenosis in each patient was performed prior to carotid surgery. Demographic and clinical data such as gender, age, smoking status, blood pressure, glycaemia, hemoglobin and creatinine levels, and comorbidities were collected and the comparisons between variables were statistically evaluated. RESULTS Serum levels of IL-33 (35.86 ± 7.93 pg/ml vs.12.29 ± 1.8 pg/ml, p < 0.05) and sST2 (183 ± 8.03 pg/ml vs. 122.31 ± 15.89 pg/ml, p < 0.05) were significantly higher in the group of CEA patients vs. healthy subjects. We demonstrated abundant tissue expression of IL-33 and ST2 in atherosclerotic carotid artery lesions. The levels of IL-33 and IL-33R expression were significantly higher in vulnerable plaques and significantly correlated with the degree of inflammatory cells infiltration in these plaques (R = 0.579, p = 0.049). Immunohistochemical analysis also revealed that cells responsible for IL-33 expression are not only mononuclear cells confined to inflammatory atherosclerotic lesions, but also smooth muscle cells which gained phenotypic characteristics of foam cells and were loaded with lipid droplets. CONCLUSION The obtained results confirm the importance of IL-33/ST2 axis in the process of atherosclerosis, and indicate its ambiguous function in immune response, whether as proinflammatory cytokine in advanced atherosclerotic lesions, or as profibrotic, in early lesions.
Collapse
Affiliation(s)
- Milos Stankovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Biljana Ljujic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Srdjan Babic
- Dedinje Cardiovascular Institute, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Vera Maravic-Stojkovic
- Dedinje Cardiovascular Institute, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Slobodanka Mitrovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Nebojsa Arsenijevic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Djordje Radak
- Dedinje Cardiovascular Institute, School of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nada Pejnovic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Miodrag L Lukic
- Center for Molecular Medicine and Stem Cell Research, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia.
| |
Collapse
|
44
|
The Potential Role of Neutrophil Gelatinase-Associated Lipocalin in the Development of Abdominal Aortic Aneurysms. Ann Vasc Surg 2019; 57:210-219. [DOI: 10.1016/j.avsg.2018.11.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/14/2018] [Accepted: 11/27/2018] [Indexed: 11/20/2022]
|
45
|
Dapagliflozin attenuates early markers of diabetic nephropathy in fructose-streptozotocin-induced diabetes in rats. Biomed Pharmacother 2018; 109:910-920. [PMID: 30551545 DOI: 10.1016/j.biopha.2018.10.100] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 10/02/2018] [Accepted: 10/20/2018] [Indexed: 12/13/2022] Open
Abstract
Early detection and clinical interference are major challenges for the prevention of diabetic nephropathy (DN) progression. This study investigated the effects of dapagliflozin, a sodium glucose co-transporter 2 inhibitor, on some early markers for DN in fructose-streptozotocin (Fr-STZ)-induced diabetes in rats. Fr-STZ rats were treated with either dapagliflozin (1 mg/kg p.o. daily), metformin (350 mg/kg p.o. daily), or their combination for 6 weeks. Fr-STZ rats displayed marked early tubular renal damage and glomerular podocyte injury as evidenced by renal KIM-1, NGAL, cystatin C, and vanin-1 mRNA, as well as urinary NAG elevation and nephrin mRNA suppression, associated with the development of marked renal interstitial fibrosis and glomerulosclerosis despite the presence of normoalbuminuria. Propagation of oxidative, inflammatory, fibrotic, and apoptotic reactions was obvious in the setting of renal glucose overload. Dapagliflozin significantly attenuated the renal tubular injury makers namely KIM-1, NGAL, vanin-1 and urinary NAG. In addition, it restored glomerular nephrin expression and reversed renal histopathological changes. Oxidative, inflammatory, and fibrotic processes were also alleviated. This study suggests that dapagliflozin exerts a renoprotective effect against early features of DN in rats presumably by inhibition of diabetes-induced renal tubular and glomerular injury thereby modulating oxidative, inflammatory, and fibrotic as well as apoptotic mechanisms elicited during hyperglycemia.
Collapse
|
46
|
Gorini S, Marzolla V, Mammi C, Armani A, Caprio M. Mineralocorticoid Receptor and Aldosterone-Related Biomarkers of End-Organ Damage in Cardiometabolic Disease. Biomolecules 2018; 8:biom8030096. [PMID: 30231508 PMCID: PMC6165349 DOI: 10.3390/biom8030096] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 09/10/2018] [Accepted: 09/12/2018] [Indexed: 12/11/2022] Open
Abstract
The mineralocorticoid receptor (MR) was first identified as a blood pressure regulator, modulating renal sodium handling in response to its principal ligand aldosterone. The mineralocorticoid receptor is also expressed in many tissues other than the kidney, such as adipose tissue, heart and vasculature. Recent studies have shown that MR plays a relevant role in the control of cardiovascular and metabolic function, as well as in adipogenesis. Dysregulation of aldosterone/MR signaling represents an important cause of disease as high plasma levels of aldosterone are associated with hypertension, obesity and increased cardiovascular risk. Aldosterone displays powerful vascular effects and acts as a potent pro-fibrotic agent in cardiovascular remodeling. Mineralocorticoid receptor activation regulates genes involved in vascular and cardiac fibrosis, calcification and inflammation. This review focuses on the role of novel potential biomarkers related to aldosterone/MR system that could help identify cardiovascular and metabolic detrimental conditions, as a result of altered MR activation. Specifically, we discuss: (1) how MR signaling regulates the number and function of different subpopulations of circulating and intra-tissue immune cells; (2) the role of aldosterone/MR system in mediating cardiometabolic diseases induced by obesity; and (3) the role of several MR downstream molecules as novel potential biomarkers of cardiometabolic diseases, end-organ damage and rehabilitation outcome.
Collapse
Affiliation(s)
- Stefania Gorini
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Vincenzo Marzolla
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Caterina Mammi
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Andrea Armani
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
| | - Massimiliano Caprio
- Laboratory of Cardiovascular Endocrinology, IRCCS San Raffaele Pisana, Via di Val Cannuta 247, 00166 Rome, Italy.
- Department of Human Sciences and Promotion of the Quality of Life, San Raffaele Roma Open University, 00166 Rome, Italy.
| |
Collapse
|
47
|
Amersfoort J, Schaftenaar FH, Douna H, van Santbrink PJ, Kröner MJ, van Puijvelde GHM, Quax PHA, Kuiper J, Bot I. Lipocalin-2 contributes to experimental atherosclerosis in a stage-dependent manner. Atherosclerosis 2018; 275:214-224. [PMID: 29960897 DOI: 10.1016/j.atherosclerosis.2018.06.015] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 05/15/2018] [Accepted: 06/08/2018] [Indexed: 12/01/2022]
Abstract
BACKGROUND AND AIMS Lipocalin-2 (Lcn2) is a glycoprotein which can be secreted by immune cells. Several studies in humans have suggested Lcn2 can be used as a biomarker for the detection of unstable atherosclerotic lesions, partly as it is known to interact with MMP-9. METHODS In this study, we generated Ldlr-/-Lcn2-/- mice to assess the functional role of Lcn2 in different stages of atherosclerosis. Atherosclerotic lesions were characterized through histological analysis and myeloid cell populations were examined using flow cytometry. RESULTS We show that Ldlr-/-Lcn2-/- mice developed larger atherosclerotic lesions during earlier stages of atherosclerosis and had increased circulating Ly6Chi inflammatory monocytes compared to Ldlr-/- mice. Advanced atherosclerotic lesions from Ldlr-/-Lcn2-/- mice had decreased necrotic core area, suggesting Lcn2 deficiency may affect lesion stability. Furthermore, MMP-9 activity was diminished in plaques from Ldlr-/-Lcn2-/- mice. CONCLUSIONS Altogether, these findings suggest that Lcn2 deficiency promotes lesion growth in earlier stages of the disease while it decreases MMP-9 activity and necrotic core size in advanced atherosclerosis.
Collapse
Affiliation(s)
- J Amersfoort
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands.
| | - F H Schaftenaar
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - H Douna
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - P J van Santbrink
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - M J Kröner
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - G H M van Puijvelde
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - P H A Quax
- Department of Surgery, Leiden University Medical Center, Albinusdreef 2, 2333ZA, Leiden, The Netherlands; Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, The Netherlands
| | - J Kuiper
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| | - I Bot
- Division of Biotherapeutics, LACDR, Leiden University, Einsteinweg 55, 2333CC, Leiden, The Netherlands
| |
Collapse
|
48
|
More than a simple biomarker: the role of NGAL in cardiovascular and renal diseases. Clin Sci (Lond) 2018; 132:909-923. [PMID: 29739822 DOI: 10.1042/cs20171592] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 03/05/2018] [Accepted: 04/04/2018] [Indexed: 12/15/2022]
Abstract
Neutrophil gelatinase-associated lipocalin (NGAL) is a small circulating protein that is highly modulated in a wide variety of pathological situations, making it a useful biomarker of various disease states. It is one of the best markers of acute kidney injury, as it is rapidly released after tubular damage. However, a growing body of evidence highlights an important role for NGAL beyond that of a biomarker of renal dysfunction. Indeed, numerous studies have demonstrated a role for NGAL in both cardiovascular and renal diseases. In the present review, we summarize current knowledge concerning the involvement of NGAL in cardiovascular and renal diseases and discuss the various mechanisms underlying its pathological implications.
Collapse
|
49
|
Neutrophil Gelatinase-Associated Lipocalin from immune cells is mandatory for aldosterone-induced cardiac remodeling and inflammation. J Mol Cell Cardiol 2017; 115:32-38. [PMID: 29289651 DOI: 10.1016/j.yjmcc.2017.12.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 12/05/2017] [Accepted: 12/27/2017] [Indexed: 12/14/2022]
Abstract
Immune system activation is involved in cardiovascular (CV) inflammation and fibrosis, following activation of the mineralocorticoid receptor (MR). We previously showed that Neutrophil Gelatinase-Associated Lipocalin (NGAL) is a novel target of MR signaling in CV tissue and plays a critical role in aldosterone/MR-dependent hypertension and fibrosis. We hypothesized that the production of NGAL by immune cells may play an important part in the mediation of these deleterious mineralocorticoid-induced effects. We analyzed the effect of aldosterone on immune cell recruitment and NGAL expression in vivo. We then studied the role of NGAL produced by immune cells in aldosterone-mediated cardiac inflammation and remodeling using mice depleted for NGAL in their immune cells by bone marrow transplantation and subjected to mineralocorticoid challenge NAS (Nephrectomy, Aldosterone 200μg/kg/day, Salt 1%). NAS treatment induced the recruitment of various immune cell populations to lymph nodes (granulocytes, B lymphocytes, activated CD8+ T lymphocytes) and the induction of NGAL expression in macrophages, dendritic cells, and PBMCs. Mice depleted for NGAL in their immune cells were protected against NAS-induced cardiac remodeling and inflammation. We conclude that NGAL produced by immune cells plays a pivotal role in cardiac damage under mineralocorticoid excess. Our data further stressed a pathogenic role of NGAL in cardiac damages, besides its relevance as a biomarker of renal injury.
Collapse
|
50
|
Eilenberg W, Stojkovic S, Kaider A, Kozakowski N, Domenig CM, Burghuber C, Nanobachvili J, Huber K, Klinger M, Neumayer C, Huk I, Wojta J, Demyanets S. NGAL and MMP-9/NGAL as biomarkers of plaque vulnerability and targets of statins in patients with carotid atherosclerosis. Clin Chem Lab Med 2017; 56:147-156. [PMID: 28672747 DOI: 10.1515/cclm-2017-0156] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/13/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Neutrophil gelatinase associated lipocalin (NGAL) is expressed in atherosclerotic lesions and was recently implicated in the pathogenesis of cardiovascular pathologies. Statins are known to exert stabilizing effects on atherosclerotic plaque. The aims of our study were (1) to investigate the association of serum NGAL and metalloproteinase (MMP)-9/NGAL complex with the vulnerability of the atherosclerotic plaque, and (2) to reveal the effects of statin treatment on circulating NGAL and MMP-9/NGAL levels in patients with carotid artery stenosis. METHODS We examined the levels of NGAL and MMP-9/NGAL in blood samples from 136 patients with carotid artery stenosis by specific enzyme-linked immunosorbent assays. RESULTS Patients with vulnerable plaques, as determined by ultrasound (plaques with decreased echogenicity) and histological analysis (type VI according to the classification of American Heart Association [AHA]), displayed the highest levels of NGAL (both p<0.0001) and MMP-9/NGAL complex (p=0.0004 and p=0.004, respectively). Moreover, patients with symptomatic carotid atherosclerosis had significantly higher NGAL levels compared to asymptomatic patients (p=0.0007). The statin-treated group (n=108) demonstrated lower NGAL (73.9 vs. 128.0 μg/L, p<0.0001) and MMP-9/NGAL (28.9 vs. 40.6 μg/L, p=0.046) as compared to the non-statin group (n=28). Furthermore, in multivariate regression analysis NGAL, but not MMP-9/NGAL levels, were independently associated with symptomatic carotid artery stenosis. In addition, statin treatment was independently associated with lower NGAL levels. CONCLUSIONS Circulating NGAL and MMP-9/NGAL are associated with plaque vulnerability in patients with carotid artery stenosis. Statin treatment could contribute to plaque stabilization by reducing circulating NGAL and MMP-9/NGAL levels.
Collapse
|