1
|
Sharma A, Mannan A, Singh S, Singh TG. A second act for spironolactone: cognitive benefits in renal dysfunction - a critical review. Metab Brain Dis 2025; 40:194. [PMID: 40299184 DOI: 10.1007/s11011-025-01623-9] [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: 03/04/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
Renal dysfunction or Chronic kidney disease (CKD) are increasingly associated with cognitive deficit and memory impairment, suggesting a crucial kidney-brain axis. This review examines spironolactone's emerging role as a neuroprotective agent in the context of renal dysfunction-induced cognitive impairment. As a selective mineralocorticoid receptor (MR) antagonist, spironolactone demonstrates multifaceted protective mechanisms beyond its well established renoprotective effects. Evidences also suggests that spironolactone attenuates neuroinflammation, mitigates oxidative stress in brain, preserve blood-brain barrier (BBB) integrity and regulates hormonal imbalances associated with renal dysfunction. This review focuses on the reported beneficial effects of spironolactone in various neurodegenerative diseases (NDDs). These mechanisms collectively protect against the neurodegeneration in memory impairment induced by renal dysfunction. The dual action of spironolactone on both renal and cerebral tissues presents a novel therapeutic advantage in addressing this complex pathophysiology. This study elucidates multiple beneficial mechanisms by which spironolactone addresses cognitive impairment associated with renal dysfunction. Spironolactone enhances BBB protection and restores BBB integrity which is often compromised with renal dysfunction. It promotes neuroplasticity (allowing for improved neural adaptation and cognitive function), additionally mediates cerebral blood flow (CBF) ensuring adequate oxygen and nutrient delivery to brain. Spironolactone's anti-inflammatory effects by inhibiting the nuclear factor-kappa B (NF-κB) pathway and modulation of neuregulin1 (NRG1)/v-erb-b2 avian erythroblastic leukemia viral oncogene homolog 4 (ERBB4) signaling effectively reduce neuroinflammation that contributes to memory impairment. It also mitigates oxidative stress by targeting NADPH-oxidase (NOX), a major source of reactive oxygen species (ROS) in the central nervous system (CNS). Spironolactone also maintains hormonal balance, particularly regarding aldosterone levels, which become dysregulated in renal dysfunction and negatively impact brain function. These insights provide new possibilities for developing targeted therapies against renal dysfunction-induced memory impairment.
Collapse
Affiliation(s)
- Akhil Sharma
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Shareen Singh
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, 140401, Rajpura, Punjab, India.
| |
Collapse
|
2
|
Thompson SE, Roy A, Geberhiwot T, Gehmlich K, Steeds RP. Fabry Disease: Insights into Pathophysiology and Novel Therapeutic Strategies. Biomedicines 2025; 13:624. [PMID: 40149601 PMCID: PMC11940501 DOI: 10.3390/biomedicines13030624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 02/23/2025] [Accepted: 02/27/2025] [Indexed: 03/29/2025] Open
Abstract
Fabry disease (FD) is an X-linked lysosomal storage disorder characterized by deficiency of α-galactosidase A (α-GalA), leading to the accumulation of glycosphingolipids and multi-organ dysfunction, particularly affecting the cardiovascular and renal systems. Disease-modifying treatments such as enzyme replacement therapy (ERT) and oral chaperone therapy (OCT) have limited efficacy, particularly in advanced disease, prompting a need for innovative therapeutic approaches targeting underlying molecular mechanisms beyond glycosphingolipid storage alone. Recent insights into the pathophysiology of FD highlights chronic inflammation and mitochondrial, lysosomal, and endothelial dysfunction as key mediators of disease progression. Adjunctive therapies such as sodium-glucose cotransporter-2 (SGLT2) inhibitors, glucagon-like peptide-1 (GLP-1) agonists, and mineralocorticoid receptor antagonists (MRAs) demonstrate significant cardiovascular and renal benefits in conditions including heart failure and chronic kidney disease. These drugs also modulate pathways involved in the pathophysiology of FD, such as autophagy, oxidative stress, and pro-inflammatory cytokine signaling. While theoretical foundations support their utility, dedicated trials are necessary to confirm efficacy in the FD-specific population. This narrative review highlights the importance of expanding therapeutic strategies in FD, advocating for a multi-faceted approach involving evidence-based adjunctive treatments to improve outcomes. Tailored research focusing on diverse FD phenotypes, including females and non-classical variants of disease, will be critical to advancing care and improving outcomes in this complex disorder.
Collapse
Affiliation(s)
- Sophie Elizabeth Thompson
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Department of Cardiology, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Ashwin Roy
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Department of Cardiology, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| | - Tarekegn Geberhiwot
- Department of Diabetes, Endocrinology and Metabolism, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
- Institute of Metabolism and System Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
| | - Katja Gehmlich
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford OX1 2JD, UK
| | - Richard Paul Steeds
- Department of Cardiovascular Sciences, School of Medical Sciences, College of Medicine and Health, University of Birmingham, Birmingham B15 2TT, UK
- Department of Cardiology, Queen Elizabeth Hospital, University Hospital Birmingham NHS Foundation Trust, Birmingham B15 2TH, UK
| |
Collapse
|
3
|
Zhao M, Cao Y, Ma L. New insights in the treatment of DKD: recent advances and future prospects. BMC Nephrol 2025; 26:72. [PMID: 39934650 DOI: 10.1186/s12882-025-03953-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
Diabetic kidney disease (DKD) represents the predominant and severe microvascular complication associated with diabetes, frequently culminating in End-Stage Kidney Disease (ESKD). The escalating prevalence of diabetes has correspondingly led to a rise in DKD incidence, imposing significant challenges on both individuals and society. The etiology of DKD is multifaceted and remains devoid of definitive therapeutic interventions. This article examines the pharmacological actions and mechanisms of different drugs used for the prevention and treatment of DKD that are currently in clinical use or undergoing development. The goal is to offer insights for early intervention based on therapeutic combinations to potentially slow kidney disease progression.
Collapse
Affiliation(s)
- Meimei Zhao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China
| | - Yongtong Cao
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China.
| | - Liang Ma
- Department of Clinical Laboratory, China-Japan Friendship Hospital, Beijing, China.
| |
Collapse
|
4
|
Yu J, Tu X, Xu K, Tang X, Wu Y, Jiang X. Clinical significance of hyperuricaemia in biopsy-proven diabetic kidney disease ━ a single-centre retrospective study. Front Endocrinol (Lausanne) 2025; 16:1481977. [PMID: 39926344 PMCID: PMC11802813 DOI: 10.3389/fendo.2025.1481977] [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: 08/17/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Aims Hyperuricaemia is associated with the development of Diabetic kidney disease (DKD). However, the mechanism of hyperuricaemia causing the progression of DKD remain unclear. Methods This is a single-centre retrospective study. 155 biopsy-proven DKD patients were grouped into hyperuricaemia and non-hyperuricaemia groups. Kaplan-Meier analysis and landmark curves were performed to explore predictors of end-stage renal disease (ESRD), Cox regression analysis was used to screen for factors, a nomogram was constructed to predict the renal prognosis of DKD. Results Patients in hyperuricaemia group had higher serum creatinine (Scr), degree of mesangial expansion and IFTA score and lower GFR, haemoglobin. SUA level was positively correlated with IFTA scores. The Kaplan-Meier curve and landmark analysis revealed worse survival in hyperuricaemia group, especially after 12 months. 11 variables, including age, sex, haemoglobin, Scr, SUA, and pathological score were collected to make a nomogram model. In the testing and training sets, the AUCs at 1, 3, and 5 years were 0.888, 0.939, and 0.886 and 0.947, 0.867, and 0.905, respectively. Conclusion The clinicopathologic manifestation of DKD patients with hyperuricaemia was much more severe, and hyperuricaemia predicted a worse renal prognosis. A new renal prognosis prediction model including SUA was constructed for DKD with higher accuracy.
Collapse
Affiliation(s)
- Jin Yu
- Department of Nephrology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Precise Prevention and Treatment of Rheumatism Syndrome of Renal Wind Disease, Hangzhou, China
| | - Xiao Tu
- Department of Nephrology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Precise Prevention and Treatment of Rheumatism Syndrome of Renal Wind Disease, Hangzhou, China
| | - Kunyue Xu
- Department of Nephrology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Xuanli Tang
- Department of Nephrology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Precise Prevention and Treatment of Rheumatism Syndrome of Renal Wind Disease, Hangzhou, China
| | - Yufan Wu
- Department of Nephrology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Xue Jiang
- Department of Nephrology, Hangzhou Traditional Chinese Medicine (TCM) Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
- Key Laboratory of Precise Prevention and Treatment of Rheumatism Syndrome of Renal Wind Disease, Hangzhou, China
| |
Collapse
|
5
|
Zhou Q, Shao X, Xu L, Zou H, Chen W. Association between Monocyte-to-Lymphocyte Ratio and Inflammation in Chronic Kidney Disease: A Cross-Sectional Study. Kidney Blood Press Res 2024; 49:1066-1074. [PMID: 39561718 PMCID: PMC11844676 DOI: 10.1159/000542625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 11/01/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION Inflammation plays a key role in chronic kidney disease (CKD). Monocyte-to-lymphocyte ratio (MLR) is a novel inflammatory marker. The purpose of this study was to evaluate the relationship between MLR and inflammation in CKD patients. METHODS In total, 1,809 subjects were recruited from Wanzhai Town, Zhuhai City, between December 2017 and March 2018 for a cross-sectional survey. Patients were categorized based on the absence (hypersensitive C-reactive protein [hsCRP] level ≦3 mg/L) or presence (hsCRP level >3 mg/L) of inflammation. Logistic regression models and MLR quartiles were used to explore the relationship between MLR and inflammation in CKD patients. RESULTS Among 1,809 subjects, 403 (22.2%) had CKD. Significant differences in systolic blood pressure, estimated glomerular filtration rate, white blood cell (WBC), neutrophil, monocyte, MLR, and interleukin-6 (IL-6) levels were observed between noninflammatory group and inflammatory group. The highest MLR quartile had higher Scr, WBC, neutrophil, monocyte, IL-6, and hsCRP values and lower eGFR and lymphocyte values. Comparing the lowest quartile of MLR, the OR (95% CI) of inflammation risk in the highest quartile was 2.30 (1.24-4.27) after adjustment for confounding factors. The area under the curve of MLR for predicting inflammation was 0.631. The cutoff point for the MLR was 0.153. CONCLUSION A high MLR was significantly and independently associated with inflammation in patients with CKD, making MLR a potential marker for inflammation in this demographic. MLR may also predict the severity of CKD.
Collapse
Affiliation(s)
- Qin Zhou
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Shao
- Department of Nephrology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Li Xu
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hequn Zou
- Department of Nephrology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, China
| | - Wenli Chen
- Department of Nephrology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
6
|
Ho QV, Young MJ. Mineralocorticoid receptors, macrophages and new mechanisms for cardiovascular disease. Mol Cell Endocrinol 2024; 593:112340. [PMID: 39134137 DOI: 10.1016/j.mce.2024.112340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/16/2024]
Affiliation(s)
- Quoc Viet Ho
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia
| | - Morag J Young
- Cardiovascular Endocrinology Laboratory, Baker Heart and Diabetes Institute, Australia; Department of Cardiometabolic Health, University of Melbourne, Victoria, Australia.
| |
Collapse
|
7
|
Wang F, Huang X, Wang S, Wu D, Zhang M, Wei W. The main molecular mechanisms of ferroptosis and its role in chronic kidney disease. Cell Signal 2024; 121:111256. [PMID: 38878804 DOI: 10.1016/j.cellsig.2024.111256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/25/2024] [Accepted: 06/07/2024] [Indexed: 06/21/2024]
Abstract
The term ferroptosis, coined in 2012, has been widely applied in various disease research fields. Ferroptosis is a newly regulated form of cell death distinct from apoptosis, necrosis, and autophagy, the mechanisms of which have been extensively studied. Chronic kidney disease, characterized by renal dysfunction, is a common disease severely affecting human health, with its occurrence and development influenced by multiple factors and leading to dysfunction in multiple systems. It often lacks obvious clinical symptoms in the early stages, and thus, diagnosis is typically made in the later stages, complicating treatment. While research on ferroptosis and acute kidney injury has made continuous progress, studies on the association between ferroptosis and chronic kidney disease remain limited. This review aims to summarize chronic kidney disease, investigate the mechanism and regulation of ferroptosis, and attempt to elucidate the role of ferroptosis in the occurrence and development of chronic kidney disease.
Collapse
Affiliation(s)
- Fulin Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Xuesong Huang
- Department of Urology, Jilin People's Hospital, Jilin, China
| | - Shaokun Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Dawei Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | | | - Wei Wei
- Department of Urology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
8
|
Song S, Cai X, Hu J, Zhu Q, Shen D, Ma H, Zhang Y, Ma R, Zhou P, Yang W, Hong J, Zhang D, Li N. Plasma aldosterone concentrations elevation in hypertensive patients: the dual impact on hyperuricemia and gout. Front Endocrinol (Lausanne) 2024; 15:1424207. [PMID: 39140032 PMCID: PMC11319118 DOI: 10.3389/fendo.2024.1424207] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 07/03/2024] [Indexed: 08/15/2024] Open
Abstract
Background Prior research has highlighted the association between uric acid (UA) and the activation of the renin-angiotensin-aldosterone system (RAAS). However, the specific relationship between aldosterone, the RAAS's end product, and UA-related diseases remains poorly understood. This study aims to clarify the impact of aldosterone on the development and progression of hyperuricemia and gout in hypertensive patients. Methods Our study involved 34534 hypertensive participants, assessing plasma aldosterone concentration (PAC)'s role in UA-related diseases, mainly hyperuricemia and gout. We applied multiple logistic regression to investigate the impact of PAC and used restricted cubic splines (RCS) for examining the dose-response relationship between PAC and these diseases. To gain deeper insights, we conducted threshold analyses, further clarifying the nature of this relationship. Finally, we undertook subgroup analyses to evaluate PAC's effects across diverse conditions and among different subgroups. Results Multivariate logistic regression analysis revealed a significant correlation between the occurrence of hyperuricemia and gout and the elevation of PAC levels. Compared to the first quartile (Q1) group, groups Q2, Q3, and Q4 all exhibited a significantly increased risk of occurrence. Moreover, the conducted RCS analysis demonstrated a significant nonlinear dose-response relationship, especially when PAC was greater than 14 ng/dL, with a further increased risk of hyperuricemia and gout. Finally, comprehensive subgroup analyses consistently reinforced these findings. Conclusion This study demonstrates a close association between elevated PAC levels and the development of UA-related diseases, namely hyperuricemia and gout, in hypertensive patients. Further prospective studies are warranted to confirm and validate this relationship.
Collapse
Affiliation(s)
- Shuaiwei Song
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Xintian Cai
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Junli Hu
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Qing Zhu
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Di Shen
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Huimin Ma
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Yingying Zhang
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Rui Ma
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Pan Zhou
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Wenbo Yang
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Jing Hong
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Delian Zhang
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| | - Nanfang Li
- Key Laboratory of Xinjiang Uygur Autonomous Region “Hypertension Research Laboratory”, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
- NHC Key Laboratory of Hypertension Clinical Research, Hypertension Center of People’s Hospital of Xinjiang Uygur Autonomous Region, Xinjiang Hypertension Institute, Xinjiang Clinical Medical Research Center for Hypertension (Cardio-Cerebrovascular) Diseases, Urumqi, China
| |
Collapse
|
9
|
Finsen SH, Hansen MR, Hansen PBL, Mortensen SP. Eight weeks of treatment with mineralocorticoid receptor blockade does not alter vascular function in individuals with and without type 2 diabetes. Physiol Rep 2024; 12:e16010. [PMID: 38610066 PMCID: PMC11014871 DOI: 10.14814/phy2.16010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 03/27/2024] [Accepted: 03/31/2024] [Indexed: 04/14/2024] Open
Abstract
Aldosterone has been suggested to be involved in the microvascular complications observed in type 2 diabetes. We aimed to investigate the effect of mineralocorticoid receptor (MR) blockade on endothelial function in individuals with type 2 diabetes compared to healthy controls. We included 12 participants with type 2 diabetes and 14 controls. We measured leg hemodynamics at baseline and during femoral arterial infusion of acetylcholine and sodium nitroprusside before and 8 weeks into treatment with MR blockade (eplerenone). Acetylcholine infusion was repeated with concomitant n-acetylcysteine (antioxidant) infusion. No difference in leg blood flow or vascular conductance was detected before or after the treatment with MR blockade in both groups and there was no difference between groups. Infusion of n-acetylcysteine increased baseline blood flow and vascular conductance, but did not change the vascular response to acetylcholine before or after treatment with MR blockade. Skeletal muscle eNOS content was unaltered by MR blockade and no difference between groups was detected. In conclusion, we found no effect of MR blockade endothelial function in individuals with and without type 2 diabetes. As the individuals with type 2 diabetes did not have vascular dysfunction, these results might not apply to individuals with vascular dysfunction.
Collapse
Affiliation(s)
- Stine H. Finsen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of NephrologyOdense University HospitalOdenseDenmark
| | - Mie R. Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Pernille B. L. Hansen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Stefan P. Mortensen
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
10
|
Costa RM, Cerqueira DM, Bruder-Nascimento A, Alves JV, Awata WMC, Singh S, Kufner A, Prado DS, Johny E, Cifuentes-Pagano E, Hawse WF, Dutta P, Pagano PJ, Ho J, Bruder-Nascimento T. Role of the CCL5 and Its Receptor, CCR5, in the Genesis of Aldosterone-Induced Hypertension, Vascular Dysfunction, and End-Organ Damage. Hypertension 2024; 81:776-786. [PMID: 38240165 PMCID: PMC10954408 DOI: 10.1161/hypertensionaha.123.21888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 01/03/2024] [Indexed: 01/30/2024]
Abstract
BACKGROUND Aldosterone has been described to initiate cardiovascular diseases by triggering exacerbated sterile vascular inflammation. The functions of CCL5 (C-C motif chemokine ligand 5) and its receptor CCR5 (C-C motif chemokine receptor 5) are well known in infectious diseases, their contributions to aldosterone-induced vascular injury and hypertension remain unknown. METHODS We analyzed the vascular profile, blood pressure, and renal damage in wild-type (CCR5+/+) and CCR5 knockout (CCR5-/-) mice treated with aldosterone (600 µg/kg per day for 14 days) while receiving 1% saline to drink. Vascular function was analyzed in aorta and mesenteric arteries, blood pressure was measured by telemetry and renal injury and inflammation were analyzed via histology and flow cytometry. Endothelial cells were used to study the molecular signaling whereby CCL5 induces endothelial dysfunction. RESULTS Aldosterone treatment resulted in exaggerated CCL5 circulating levels and vascular CCR5 expression in CCR5+/+ mice accompanied by endothelial dysfunction, hypertension, and renal inflammation and damage. CCR5-/- mice were protected from these aldosterone-induced effects. Mechanistically, we demonstrated that CCL5 increased NOX1 (NADPH oxidase 1) expression, reactive oxygen species formation, NFκB (nuclear factor kappa B) activation, and inflammation and reduced NO production in isolated endothelial cells. These effects were abolished by antagonizing CCR5 with Maraviroc. Finally, aorta incubated with CCL5 displayed severe endothelial dysfunction, which is prevented by blocking NOX1, NFκB, or CCR5. CONCLUSIONS Our data demonstrate that CCL5/CCR5, through activation of NFκB and NOX1, is critically involved in aldosterone-induced vascular and renal damage and hypertension placing CCL5 and CCR5 as potential therapeutic targets for conditions characterized by aldosterone excess.
Collapse
Affiliation(s)
- Rafael M Costa
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Center for Pediatrics Research in Obesity and Metabolism at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Department of Medicine, Division of Cardiology (R.M.C., P.D.), University of Pittsburgh, PA
- Academic Unit of Health Sciences, Federal University of Jatai, GO, Brazil (R.M.C.)
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, Brazil (R.M.C.)
| | - Débora M Cerqueira
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Nephrology Division at UPMC Children's Hospital of Pittsburgh (D.M.C., J.H.), University of Pittsburgh, PA
| | - Ariane Bruder-Nascimento
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Center for Pediatrics Research in Obesity and Metabolism at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
| | - Juliano V Alves
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Center for Pediatrics Research in Obesity and Metabolism at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
| | - Wanessa M C Awata
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Center for Pediatrics Research in Obesity and Metabolism at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
| | - Shubhnita Singh
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Center for Pediatrics Research in Obesity and Metabolism at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
| | - Alexander Kufner
- Vascular Medicine Institute (A.K., E.J., E.C.-P., P.D., P.J.P., T.B.-N.), University of Pittsburgh, PA
- Department of Pharmacology and Chemical Biology (A.K., E.C.-P., P.J.P.), University of Pittsburgh, PA
| | - Douglas S Prado
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA (D.S.P., W.F.H., P.D.), University of Pittsburgh, PA
| | - Ebin Johny
- Vascular Medicine Institute (A.K., E.J., E.C.-P., P.D., P.J.P., T.B.-N.), University of Pittsburgh, PA
| | - Eugenia Cifuentes-Pagano
- Vascular Medicine Institute (A.K., E.J., E.C.-P., P.D., P.J.P., T.B.-N.), University of Pittsburgh, PA
- Department of Pharmacology and Chemical Biology (A.K., E.C.-P., P.J.P.), University of Pittsburgh, PA
| | - William F Hawse
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA (D.S.P., W.F.H., P.D.), University of Pittsburgh, PA
| | - Partha Dutta
- Vascular Medicine Institute (A.K., E.J., E.C.-P., P.D., P.J.P., T.B.-N.), University of Pittsburgh, PA
- Department of Medicine, Division of Cardiology (R.M.C., P.D.), University of Pittsburgh, PA
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA (D.S.P., W.F.H., P.D.), University of Pittsburgh, PA
| | - Patrick J Pagano
- Vascular Medicine Institute (A.K., E.J., E.C.-P., P.D., P.J.P., T.B.-N.), University of Pittsburgh, PA
- Department of Pharmacology and Chemical Biology (A.K., E.C.-P., P.J.P.), University of Pittsburgh, PA
| | - Jacqueline Ho
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Nephrology Division at UPMC Children's Hospital of Pittsburgh (D.M.C., J.H.), University of Pittsburgh, PA
| | - Thiago Bruder-Nascimento
- Department of Pediatrics at University of Pittsburgh Medical Center (UPMC) Children's Hospital of Pittsburgh, (R.M.C., D.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., J.H., T.B.-N.), University of Pittsburgh, PA
- Center for Pediatrics Research in Obesity and Metabolism at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh (R.M.C., A.B.-N., J.V.A., W.M.C.A., S.S., T.B.-N.), University of Pittsburgh, PA
- Vascular Medicine Institute (A.K., E.J., E.C.-P., P.D., P.J.P., T.B.-N.), University of Pittsburgh, PA
| |
Collapse
|
11
|
Cheng Y, Chen Y, Zhao M, Wang M, Liu M, Zhao L. Metabolomic profiling reveals the mechanisms underlying the nephrotoxicity of methotrexate in children with acute lymphoblastic leukemia. Pediatr Blood Cancer 2023; 70:e30578. [PMID: 37449940 DOI: 10.1002/pbc.30578] [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: 04/06/2023] [Revised: 06/20/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Methotrexate is widely recommended as a first-line treatment for the intensive systemic and consolidation phases of childhood acute lymphoblastic leukemia. However, methotrexate-induced nephrotoxicity is a severe adverse reaction, of which the mechanisms remain unclear. METHODS An untargeted metabolomics analysis of serum from childhood acute lymphoblastic leukemia patients with delayed methotrexate excretion, with or without acute kidney injury, was performed to identify altered metabolites and metabolic pathways. An independent external validation cohort and in vitro HK-2 cell assays further verified the candidate metabolites, and explored the mechanisms underlying the nephrotoxicity of methotrexate. RESULTS Four metabolites showed significant differences between normal excretion and delayed excretion, seven metabolites reflected the differences between groups with or without acute kidney injury, and six pathways were finally enriched. In particular, oxidized glutathione was confirmed as a candidate metabolite involved in the toxicity of methotrexate. We further explored the role of glutathione deprivation-induced ferroptosis on methotrexate cytotoxicity, and it was found that methotrexate overload significantly reduced cell viability, triggered reactive oxygen species and intracellular Fe2+ accumulation, and altered the expression of ferroptosis-related proteins in HK-2 cells. These methotrexate-induced changes were alleviated or reversed by the administration of a ferroptosis inhibitor, further suggesting that ferroptosis promoted methotrexate-induced cytotoxicity in HK-2 cells. CONCLUSIONS Our findings revealed complex metabolomic profiles and provided novel insights into the mechanism by which ferroptosis contributes to the nephrotoxic effects of methotrexate.
Collapse
Affiliation(s)
- Yu Cheng
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Yanan Chen
- Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research & the Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Mingming Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Minglu Wang
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, Fujian, China
| | - Limei Zhao
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
12
|
Costa RM, Cerqueira DM, Bruder-Nascimento A, Alves JV, Awata WAC, Singh S, Kufner A, Cifuentes-Pagano E, Pagano PJ, Ho J, Bruder-Nascimento T. Role Of The C-C Motif Chemokine Ligand 5 (CCL5) And Its Receptor, C-C Motif Chemokine Receptor 5 (CCR5) In The Genesis Of Aldosterone-induced Hypertension, Vascular Dysfunction, And End-organ Damage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.558020. [PMID: 37790434 PMCID: PMC10542153 DOI: 10.1101/2023.09.22.558020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Background Aldosterone, a mineralocorticoid steroid hormone, has been described to initiate cardiovascular diseases by triggering exacerbated sterile vascular inflammation. The functions of C-C Motif Chemokine Ligand 5 (CCL5) and its receptor, C-C Motif Chemokine Receptor 5 (CCR5), are well known in infectious diseases, but their roles in the genesis of aldosterone-induced vascular injury and hypertension are unknown. Methods We analyzed the vascular profile, blood pressure, and renal damage in wild-type (CCR5+/+) and CCR5 knockout (CCR5-/-) mice treated with aldosterone (600 μg/kg/day for 14 days) while receiving 1% saline to drink. Results Here, we show that CCR5 plays a central role in aldosterone-induced vascular injury, hypertension, and renal damage. Long-term infusion of aldosterone in CCR5+/+ mice resulted in exaggerated CCL5 circulating levels and vascular CCR5 expression. Aldosterone treatment also triggered vascular injury, characterized by endothelial dysfunction and inflammation, hypertension, and renal damage. Mice lacking CCR5 were protected from aldosterone-induced vascular damage, hypertension, and renal injury. Mechanistically, we demonstrated that CCL5 increased NADPH oxidase 1 (Nox1) expression, reactive oxygen species (ROS) formation, NFκB activation, and inflammation and reduced nitric oxide production in isolated endothelial cells. These effects were abolished by antagonizing CCR5 with Maraviroc. Finally, aortae incubated with CCL5 displayed severe endothelial dysfunction, which is prevented by blocking Nox1, NFκB, or with Maraviroc treatment. Conclusions Our data demonstrate that CCL5/CCR5, through activation of NFkB and Nox1, is critically involved in aldosterone-induced vascular and renal damage and hypertension. Our data place CCL5 and CCR5 as potential targets for therapeutic interventions in conditions with aldosterone excess.
Collapse
Affiliation(s)
- Rafael M Costa
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pediatrics Research in Obesity and Metabolism (CPROM) at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Academic Unit of Health Sciences, Federal University of Jatai, Jatai, GO, BR
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Ribeirao Preto, SP, BR
| | - Débora M Cerqueira
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Nephrology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ariane Bruder-Nascimento
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pediatrics Research in Obesity and Metabolism (CPROM) at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Juliano V Alves
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pediatrics Research in Obesity and Metabolism (CPROM) at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wanessa A C Awata
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pediatrics Research in Obesity and Metabolism (CPROM) at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Shubhnita Singh
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pediatrics Research in Obesity and Metabolism (CPROM) at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Alexander Kufner
- Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eugenia Cifuentes-Pagano
- Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Patrick J Pagano
- Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jacqueline Ho
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Nephrology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
| | - Thiago Bruder-Nascimento
- Department of Pediatrics at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Pediatrics Research in Obesity and Metabolism (CPROM) at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Endocrinology Division at UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- Vascular Medicine Institute (VMI), University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
13
|
Li H, Li M, Liu C, He P, Dong A, Dong S, Zhang M. Causal effects of systemic inflammatory regulators on chronic kidney diseases and renal function: a bidirectional Mendelian randomization study. Front Immunol 2023; 14:1229636. [PMID: 37711613 PMCID: PMC10498994 DOI: 10.3389/fimmu.2023.1229636] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Background While targeted systemic inflammatory modulators show promise in preventing chronic kidney disease (CKD) progression, the causal link between specific inflammatory factors and CKD remains uncertain. Methods Using a genome-wide association study of 41 serum cytokines from 8,293 Finnish individuals, we conducted a bidirectional two-sample Mendelian randomization (MR) analysis. In addition, we genetically predicted causal associations between inflammatory factors and 5 phenotypes, including CKD, estimated glomerular filtration rate (eGFR), dialysis, rapid progression of CKD, and rapid decline in eGFR. Inverse variance weighting (IVW) served as the primary MR method, while MR-Egger, weighted median, and MR-pleiotropy residual sum and outlier (MR-PRESSO) were utilized for sensitivity analysis. Cochrane's Q test for heterogeneity. Leave-one-out method ensured stability of MR results, and Bonferroni correction assessed causal relationship strength. Results Seventeen cytokines were associated with diverse renal outcomes. Among them, after Bonferroni correction test, higher tumor necrosis factor alpha levels were associated with a rapid decrease in eGFR (OR = 1.064, 95% CI 1.028 - 1.103, P = 0.001), higher interleukin-4 levels were associated with an increase in eGFR (β = 0.003, 95% CI 0.001 - 0.005, P = 0.002), and higher growth regulated oncogene alpha (GROα) levels were associated with an increased risk of CKD (OR=1.035, 95% CI 1.012 - 1.058, P = 0.003). In contrast, genetic susceptibility to CKD was associated with an increase in GROa, and a decrease in eGFR may lead to an increase in stem cell factor. We did not find the presence of horizontal pleiotropy during the analysis. Conclusion We discovered causally related inflammatory factors that contribute to the initiation and progression of CKD at the genetic prediction level.
Collapse
Affiliation(s)
- Hongdian Li
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Mingxuan Li
- Department of Cardiology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Cong Liu
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Pengfei He
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Ao Dong
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaoning Dong
- Department of Nephrology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Mianzhi Zhang
- Department of Nephrology, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China
- Department of Nephrology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| |
Collapse
|
14
|
Liu XJ, Yu XJ, Su YK, Qiao JA, Sun YJ, Bai XJ, Zhang N, Yang HY, Yin LX, Kang YM, Yang ZM. Minocycline and Pyrrolidine Dithiocarbamate Attenuate Hypertension via Suppressing Activation of Microglia in the Hypothalamic Paraventricular Nucleus. TOHOKU J EXP MED 2023; 259:163-172. [PMID: 36450479 DOI: 10.1620/tjem.2022.j102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Proinflammatory cytokines, reactive oxygen species and imbalance of neurotransmitters are involved in the pathophysiology of angiotensin II-induced hypertension. The hypothalamic paraventricular nucleus (PVN) plays a vital role in hypertension. Evidences show that microglia are activated and release proinflammatory cytokines in angiocardiopathy. We hypothesized that angiotensin II induces PVN microglial activation, and the activated PVN microglia release proinflammatory cytokines and cause oxidative stress through nuclear factor-kappa B (NF-κB) pathway, which contributes to sympathetic overactivity and hypertension. Male Sprague-Dawley rats (weight 275-300 g) were infused with angiotensin II to induce hypertension. Then, rats were treated with bilateral PVN infusion of microglial activation inhibitor minocycline, NF-κB activation inhibitor pyrrolidine dithiocarbamate or vehicle for 4 weeks. When compared to control groups, angiotensin II-induced hypertensive rats had higher mean arterial pressure, PVN proinflammatory cytokines, and imbalance of neurotransmitters, accompanied with PVN activated microglia. These rats also had more PVN gp91phox (source of reactive oxygen species production), and NF-κB p65. Bilateral PVN infusion of minocycline or pyrrolidine dithiocarbamate partly or completely ameliorated these changes. This study indicates that angiotensin II-induced hypertensive rats have more activated microglia in PVN, and activated PVN microglia release proinflammatory cytokines and result in oxidative stress, which contributes to sympathoexcitation and hypertensive response. Suppression of activated PVN microglia by minocycline or pyrrolidine dithiocarbamate attenuates inflammation and oxidative stress, and improves angiotensin II-induced hypertension, which indicates that activated microglia promote hypertension through activated NF-κB. The findings may offer hypertension new strategies.
Collapse
Affiliation(s)
- Xiao-Jing Liu
- The Second Clinical Medical College, Shanxi Medical University
| | - Xiao-Jing Yu
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China
| | - Yu-Kun Su
- Hemodialysis Center, Shanxi Second People's Hospital
| | - Jin-An Qiao
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China
- Institute of Pediatric Diseases, Xi'an Children's Hospital
| | - Yao-Jun Sun
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University
| | - Xiao-Jie Bai
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, and the Department of Physiology, Shanxi Medical University
| | - Nana Zhang
- Department of Hypertension, The First Hospital of Shanxi Medical University
| | - Hui-Yu Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University
| | - Li-Xi Yin
- Basic Medical College of Shanxi Medical University
| | - Yu-Ming Kang
- Department of Physiology and Pathophysiology, Xi'an Jiaotong University School of Basic Medical Sciences, Shaanxi Engineering and Research Center of Vaccine, Key Laboratory of Environment and Genes Related to Diseases of Education Ministry of China
| | - Zhi-Ming Yang
- Department of Cardiology, The Second Hospital of Shanxi Medical University
| |
Collapse
|
15
|
Potential Impact of Non-Steroidal Mineralocorticoid Receptor Antagonists in Cardiovascular Disease. Int J Mol Sci 2023; 24:ijms24031922. [PMID: 36768246 PMCID: PMC9915890 DOI: 10.3390/ijms24031922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/14/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Inappropriate mineralocorticoid receptor (MR) activation in different cardiovascular cell types has deleterious effects on cardiac remodeling and function. Therefore, MR inhibition is a crucial pharmacological strategy to overcome cardiovascular dysfunction. Despite efficient blockade of MR with steroidal MR antagonists (MRAs), their clinical application is unsatisfactory due to the adverse effects. Newer non-steroidal MRAs with greater potency could be suitable for clinical application, especially in patients with type 2 diabetes mellitus and chronic kidney disease. Although clinical evidence has shown the beneficial effects of non-steroidal MRAs on cardiovascular outcomes in patients with heart failure with reduced ejection fraction, clinical trials are ongoing to evaluate the efficacy of heart failure with preserved ejection fraction. Therefore, comparative pharmacological characterization of non-steroidal MRAs over classic steroidal MRAs is crucial. Here, we summarize the pre-clinical evidence of non-steroidal MRAs, which suggests an improvement in cardiac dysfunction, as well as the underlying molecular mechanisms in animal models mimicking different clinical conditions. In addition, we discuss up-to-date information from clinical trials regarding the beneficial effects of non-steroidal MRAs on meaningful cardiovascular outcomes. Both pre-clinical and clinical evidence support treatment with non-steroidal MRAs in patients with cardiovascular disease.
Collapse
|
16
|
Abd El-Hakam FEZ, Abo Laban G, Badr El-Din S, Abd El-Hamid H, Farouk MH. Apitherapy combination improvement of blood pressure, cardiovascular protection, and antioxidant and anti-inflammatory responses in dexamethasone model hypertensive rats. Sci Rep 2022; 12:20765. [PMID: 36456799 PMCID: PMC9714403 DOI: 10.1038/s41598-022-24727-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Accepted: 11/18/2022] [Indexed: 12/03/2022] Open
Abstract
Hypertension-induced ventricular and vascular remodeling causes myocardial infarction, heart failure, and sudden death. Most available pharmaceutical products used to treat hypertension lead to adverse effects on human health. Limited data is available on apitherapy (bee products) combinations for treatment of hypertension. This study aims to evaluate the antihypertensive effects of combinations of natural apitherapy compounds used in the medical sector to treat a variety of diseases. Rats were assigned into six groups consisting of one control group and five hypertensive groups where hypertension (blood pressure > 140/90) was induced with dexamethasone. One of these groups was used as a hypertension model, while the remaining four hypertensive groups were treated with a propolis, royal jelly, and bee venom combination (PRV) at daily oral doses of 0.5, 1.0, and 2.0 mg/kg, and with losartan 10 mg/kg. The PRV combination at all doses decreased arterial blood pressure below the suboptimal value (p < 0.001), and PRV combination treatment improved dexamethasone-induced-ECG changes. The same treatment decreased angiotensin-II, endothelin-1, and tumor growth factor β serum levels in hypertensive rats. Additionally, PRV combination improved histopathological structure, and decreased serum levels of NF-kB and oxidative stress biomarkers. We concluded that PRV combination therapy may be used as a potential treatment for a variety of cardiovascular diseases.
Collapse
Affiliation(s)
| | - Gomaa Abo Laban
- Plant Protection Department, Faculty of Agriculture, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Sahar Badr El-Din
- Pharmacology Department, Faculty of Medicine for Girls, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Hala Abd El-Hamid
- Pathology Department, Faculty of Medicine for Girls, Al-Azhar University, Nasr City, 11884, Cairo, Egypt
| | - Mohammed Hamdy Farouk
- Animal Production Department, Faculty of Agriculture, Al-Azhar University, Nasr City, 11884, Cairo, Egypt.
| |
Collapse
|
17
|
Cardiorenal benefits of mineralocorticoid antagonists in CKD and type 2 diabetes : Lessons from the FIGARO-DKD trial. Herz 2022; 47:401-409. [PMID: 36094559 DOI: 10.1007/s00059-022-05138-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/22/2022] [Indexed: 11/04/2022]
Abstract
Diabetic kidney disease (DKD) develops in almost half of all patients with diabetes and is the most common cause of chronic kidney disease (CKD) worldwide. Despite the high risk of chronic renal failure in these patients, only few therapeutic strategies are available. The use of renin-angiotensin system blockers to reduce the incidence of kidney failure in patients with DKD was established years ago and remains the hallmark of therapy. The past 2 years have seen a dramatic change in our therapeutic arsenal for CKD. Sodium-glucose co-transporter‑2 inhibitors (SGLT2s) have been successfully introduced for the treatment of CKD. A further addition is a novel compound antagonizing the activation of the mineralocorticoid receptor: finerenone. Finerenone reduces albuminuria and surrogate markers of cardiovascular disease in patients who are already on optimal therapy. In the past, treatment with other mineralocorticoid receptor antagonists was hampered by a significantly increased risk of hyperkalemia. Finerenone had a much smaller effect on hyperkalemia. Together with a reduced effect on blood pressure and no signs of gynecomastia, this therapeutic strategy had a more specific anti-inflammatory effect and a smaller effect on the volume/electrolyte axis. In the FIDELIO-DKD study comparing the actions of the non-steroidal mineralocorticoid receptor antagonist finerenone with placebo, finerenone reduced the progression of DKD and the incidence of cardiovascular events, with a relatively safe adverse event profile. In this article, we summarize the available evidence on the cardioprotective and nephroprotective effects of finerenone and analyze the molecular mechanisms involved. In addition, we discuss the potential future role of mineralocorticoid receptor inhibition in the treatment of patients with diabetic CKD.
Collapse
|
18
|
Zhang L, Wang W, Xu C, Duan H, Tian X, Zhang D. Potential genetic biomarkers are found to be associated with both cognitive function and blood pressure: A bivariate genome-wide association analysis. Mech Ageing Dev 2022; 204:111671. [PMID: 35364053 DOI: 10.1016/j.mad.2022.111671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 03/17/2022] [Accepted: 03/25/2022] [Indexed: 11/15/2022]
Abstract
A bivariate genome-wide association study was conducted in 137 pairs of twins to explore the shared genetic loci between cognition and blood pressure (BP). Before SNPs imputation, rs72815554 is significantly (P < 5 × 10-8) associated with the cognition-pulse pressure (PP) phenotype, while after imputation, 4 and 9 SNPs are significantly associated with the cognition-SBP phenotype, and cognition-PP phenotype, respectively, including rs72815554. There existed SNPs with highly linkage disequilibrium (LD) of rs10998339, rs72815554, rs11665292, and rs10823231. Besides, rs10998347, rs12153038, and rs10998295 had higher RegulomeDB scores and are located in the transcription factors binding regions. Rs7574283 and rs58113664 are located in the super-enhancer regions which are expressed highly in the adrenal gland, artery, atrial tissue, brain, nerves, etc. There are 1108, 1154, 1071, and 1102 genes associated with cognition-SBP, cognition-DBP, cognition-PP, and cognition-mean arterial pressure (MAP) phenotypes at the suggestive significant association level (P < 0.05), respectively. Furthermore, 641, 630, 900, and 555 pathways are associated with cognition-SBP, cognition-DBP, cognition-PP, and cognition-MAP phenotypes at the suggestive significant association level (P < 0.05), respectively.
Collapse
Affiliation(s)
- Liming Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao 266021, China.
| | - Weijing Wang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao 266021, China.
| | - Chunsheng Xu
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao 266021, China.
| | - Haiping Duan
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao 266021, China.
| | - Xiaocao Tian
- Qingdao Municipal Centre for Disease Control and Prevention, Qingdao 266021, China.
| | - Dongfeng Zhang
- Department of Epidemiology and Health Statistics, The School of Public Health of Qingdao University, No.308 Ningxia Road, Qingdao 266021, China.
| |
Collapse
|
19
|
Niu X, Song H, Xiao X, Yu J, Yu J, Yang Y, Huang Q, Zang L, Han T, Zhang D, Li W. Tectoridin alleviates lipopolysaccharide -induced inflammaion via inhibiting TLR4-NF-κB/NLRP3 signaling in vivo and in vitro. Immunopharmacol Immunotoxicol 2022; 44:641-655. [PMID: 35506641 DOI: 10.1080/08923973.2022.2073890] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUD Tectoridin, widely extracted and separated from the rhizome of Iris tectorum Maxium, is extensively reported to have affluent bioactivity, but rarely reported to have anti-inflammatory effects. In this study, we aim to investigate the anti-inflammatory effects and the underlying mechanisms of tectoridin. METHODS Here, RAW264.7 macrophages were stimulated with Lipopolysaccharide (LPS) for the inflammation model in vitro. Experimental animals received tectoridin and Dexamethasone (DEX) before LPS injection for endotoxic shock mouse model in vivo. The pro-inflammatory mediators and cytokines in the cell supernatant and serum were detected by ELISA kits. The tissue damages were assessed by biochemical indexes and H&E staining. Immunohistochemistry and Western blot were performed for the detection of proteins. RESULTS Our data showed that tectoridin attenuated the LPS-up-regulated nitric oxide (NO), interleukin-6, (IL-6) and interleukin-18, (IL-18) from macrophages and tumor necrosis factor-α, (TNF-α); (IL-6) and (IL-1β) in the serum levels. Besides, our histopathological study showed that the damages caused by LPS in the lung, liver and kidney tissues were decreased. Furthermore, our results demonstrated that tectoridin inhibited the activation of TLR4-NF-κB/NLRP3 signaling proved by immunohistochemistry assay and Western blot. CONCLUSION Taken all together, tectoridin might have the potential ability of anti-inflammatory effects and the possible mechanism may be relevant to its inhibition of TLR4-NF-κB/NLRP3 signaling.
Collapse
Affiliation(s)
- Xiaofeng Niu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Huixin Song
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Xin Xiao
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jinjin Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Jiabao Yu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yajie Yang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qiuxia Huang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lulu Zang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Tengfei Han
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, Shaanxi, China
| | - Dezhu Zhang
- Shaanxi Panlong Pharmaceutical Group Limited by Share LTD, Xi'an, Shaanxi, China
| | - Weifeng Li
- School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| |
Collapse
|
20
|
Oxidative Stress and Ginsenosides: An Update on the Molecular Mechanisms. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:9299574. [PMID: 35498130 PMCID: PMC9045968 DOI: 10.1155/2022/9299574] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 12/18/2022]
Abstract
Ginsenosides are a class of active components extracted from ginseng plants (such as Panax ginseng, Panax quinquefolium, and Panax notoginseng). Ginsenosides have significant protective effects on the nervous system, cardiovascular system, and immune system, so they have been widely used in the treatment of related diseases. Entry of a variety of endogenous or exogenous harmful substances into the body can lead to an imbalance between the antioxidant defense system and reactive oxygen species, thus producing toxic effects on a variety of tissues and cells. In addition, oxidative stress can alter multiple signaling pathways, including the Keap1/Nrf2/ARE, PI3K/AKT, Wnt/β-catenin, and NF-κB pathways. With the deepening of research in this field, various ginsenoside monomers have been reported to exert antioxidant effects through multiple signaling pathways and thus have good application prospects. This article summarized the research advancements regarding the antioxidative effects and related mechanisms of ginsenosides, providing a theoretical basis for experimental research on and clinical treatment with ginsenosides.
Collapse
|
21
|
Lv X, Hu H, Shen C, Zhang X, Yan L, Zhang S, Guo Y. Risk Factors Associated With Lower Bone Mineral Density in Primary Aldosteronism Patients. Front Endocrinol (Lausanne) 2022; 13:884302. [PMID: 35784563 PMCID: PMC9245341 DOI: 10.3389/fendo.2022.884302] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
PURPOSE The association between primary aldosteronism (PA) and lower bone mineral density (BMD) has raised a concern, but the contributing factors remain unclear. We aim to explore the risk factors for lower BMD in PA patients. METHODS We analyzed and compared the data of 60 PA patients with 60 matched essential hypertension (EH) patients. BMD, bone metabolites, and several oxidative stress and inflammation indicators-including C-reactive protein (CRP), superoxide dismutase (SOD), total bilirubin (TBIL), mean platelet volume (MPV), etc.-were assessed and compared in PA and EH patients. Bivariate correlation analysis and multivariate linear regression analysis were performed to explore the factors associated with BMD in PA patients. RESULTS The BMD measured by quantitative computed tomography in PA patients was lower than that in EH patients (141.9 ± 34.0 vs. 158.9 ± 55.9 g/cm3, p = 0.047), especially in patients less than 50 years old. BMD was independently negatively associated with age (standardized β = -0.581, p < 0.001), serum phosphorus (standardized β = -0.203, p = 0.008), urinary calcium excretion (standardized β = -0.185, p = 0.031), and MPV (standardized β = -0.172, p = 0.043) and positively associated with SOD (standardized β = 0.205, p = 0.011) and TBIL (standardized β = 0.212, p = 0.015). CONCLUSIONS The PA patients showed a lower BMD than the EH patients, which was associated with age, serum phosphorus, urinary calcium excretion, MPV, SOD, and TBIL. These variables might be potential markers for the assessment of bone loss and efficacy of treatments in PA patients.
Collapse
Affiliation(s)
- Xiaomei Lv
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Huijun Hu
- Department of Radiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chuyu Shen
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyun Zhang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Li Yan
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Shaoling Zhang
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ying Guo
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- *Correspondence: Ying Guo,
| |
Collapse
|
22
|
Fan XZ, Wang YY, Cui ZY, Cheng ZH, Zhang HL, Gamper N, Zhang F, Han M. Kv7.4 channel is a key regulator of vascular inflammation and remodeling in neointimal hyperplasia and abdominal aortic aneurysms. Free Radic Biol Med 2022; 178:111-124. [PMID: 34863875 DOI: 10.1016/j.freeradbiomed.2021.11.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Revised: 11/08/2021] [Accepted: 11/30/2021] [Indexed: 12/16/2022]
Abstract
Inflammation has recently emerged as an important contributor for cardiovascular disease development and participates pivotally in the development of neointimal hyperplasia and abdominal aortic aneurysms (AAA) formation. Kv7.4/KCNQ4, a K+ channel, is one of the important regulators of vascular function but its role in vascular inflammation is unexplored. Here, we showed that the expression of Kv7.4 channel was elevated in the neointima and AAA tissues from mice and humans. Genetic deletion or pharmacological inhibition of Kv7.4 channel in mice alleviated neointimal hyperplasia and AAA formation via downregulation of a set of vascular inflammation-related genes, matrix metalloproteinases (MMP) 2/9, and intercellular adhesion molecule (ICAM-1). Furthermore, genetic deletion or inhibition of Kv7.4 channel suppressed the activation of tumor necrosis factor receptor 1 (TNFR1)-nuclear factor (NF)-κB signaling pathway via blockade of interaction between TNFR1 and TNFR1-associated death domain protein (TRADD) in vascular smooth muscle cells (VSMCs). Knockdown of Kv7.4 in vivo identified VSMC-expressed Kv7.4 as a major factor in vascular inflammation. Collectively, our findings suggest that Kv7.4 channel aggravates vascular inflammatory response, which promotes the neointimal hyperplasia and AAA formation. Inhibition of Kv7.4 channel may be a novel therapeutic strategy for vascular inflammatory diseases.
Collapse
Affiliation(s)
- Xi-Zhenzi Fan
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Ying-Ying Wang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Yang Cui
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Zi-Hao Cheng
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Hai-Lin Zhang
- Department of Pharmacology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, PR China; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, PR China
| | - Nikita Gamper
- Department of Pharmacology, Key Laboratory of Neural and Vascular Biology, Ministry of Education, PR China; Key Laboratory of New Drug Pharmacology and Toxicology, Hebei Medical University, Shijiazhuang, 050017, PR China; School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, UK
| | - Fan Zhang
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| | - Mei Han
- Department of Biochemistry and Molecular Biology, College of Basic Medicine, Key Laboratory of Medical Biotechnology of Hebei Province, Key Laboratory of Neural and Vascular Biology of Ministry of Education, Hebei Medical University, Shijiazhuang, 050017, PR China.
| |
Collapse
|
23
|
Luo M, Cao C, Niebauer J, Yan J, Ma X, Chang Q, Zhang T, Huang X, Liu G. Effects of different intensities of continuous training on vascular inflammation and oxidative stress in spontaneously hypertensive rats. J Cell Mol Med 2021; 25:8522-8536. [PMID: 34331512 PMCID: PMC8419160 DOI: 10.1111/jcmm.16813] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 07/05/2021] [Accepted: 07/09/2021] [Indexed: 12/17/2022] Open
Abstract
We aimed to study the effects and underlying mechanism of different intensities of continuous training (CT) on vascular inflammation and oxidative stress in spontaneously hypertensive rats (SHR). Rats were divided into five groups (n = 12): Wistar‐Kyoto rats sedentary group (WKY‐S), sedentary group (SHR‐S), low‐intensity CT group (SHR‐L), medium‐intensity CT group (SHR‐M) and high‐intensity CT group (SHR‐H). Changes in body mass, heart rate and blood pressure were recorded. The rats were euthanized after 14 weeks, and blood and vascular tissue samples were collected. Haematoxylin and Eosin staining was used to observe the aortic morphology, and Western blot was used to detect the expression of mesenteric artery proteins. After CT, the mean arterial pressures improved in SHR‐L and SHR‐M and increased in SHR‐H compared with those in SHR‐S. Vascular inflammation and oxidative stress levels significantly subsided in SHR‐L and SHR‐M (p < 0.05), whereas in SHR‐H, only vascular inflammation significantly subsided (p < 0.05), and oxidative stress remained unchanged (p > 0.05). AMPK and SIRT1/3 expressions in SHR‐L and SHR‐M were significantly up‐regulated than those in SHR‐S (p < 0.05). These results indicated that low‐ and medium‐intensity CT can effectively reduce the inflammatory response and oxidative stress of SHR vascular tissue, and high‐intensity CT can improve vascular tissue inflammation but not oxidative stress.
Collapse
Affiliation(s)
- Minghao Luo
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Chunmei Cao
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Josef Niebauer
- University Institute of Sports Medicine, Prevention and Rehabilitation, Paracelsus Medical University, Salzburg, Austria
| | - Jianghong Yan
- Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Xindong Ma
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Qing Chang
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| | - Ting Zhang
- The Fifth Affiliated Hospital of Sun Yat-sen University, Guangdong, China
| | - Xiaoxiao Huang
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| | - Guochun Liu
- The College of Exercise Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
24
|
Stenvinkel P, Chertow GM, Devarajan P, Levin A, Andreoli SP, Bangalore S, Warady BA. Chronic Inflammation in Chronic Kidney Disease Progression: Role of Nrf2. Kidney Int Rep 2021; 6:1775-1787. [PMID: 34307974 PMCID: PMC8258499 DOI: 10.1016/j.ekir.2021.04.023] [Citation(s) in RCA: 128] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022] Open
Abstract
Despite recent advances in the management of chronic kidney disease (CKD), morbidity and mortality rates in these patients remain high. Although pressure-mediated injury is a well-recognized mechanism of disease progression in CKD, emerging data indicate that an intermediate phenotype involving chronic inflammation, oxidative stress, hypoxia, senescence, and mitochondrial dysfunction plays a key role in the etiology, progression, and pathophysiology of CKD. A variety of factors promote chronic inflammation in CKD, including oxidative stress and the adoption of a proinflammatory phenotype by resident kidney cells. Regulation of proinflammatory and anti-inflammatory factors through NF-κB- and nuclear factor, erythroid 2 like 2 (Nrf2)-mediated gene transcription, respectively, plays a critical role in the glomerular and tubular cell response to kidney injury. Chronic inflammation contributes to the decline in glomerular filtration rate (GFR) in CKD. Whereas the role of chronic inflammation in diabetic kidney disease (DKD) has been well-elucidated, there is now substantial evidence indicating unresolved inflammatory processes lead to fibrosis and eventual end-stage kidney disease (ESKD) in several other diseases, such as Alport syndrome, autosomal-dominant polycystic kidney disease (ADPKD), IgA nephropathy (IgAN), and focal segmental glomerulosclerosis (FSGS). In this review, we aim to clarify the mechanisms of chronic inflammation in the pathophysiology and disease progression across the spectrum of kidney diseases, with a focus on Nrf2.
Collapse
Affiliation(s)
- Peter Stenvinkel
- Department of Renal Medicine M99, Karolinska University Hospital at Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Glenn M Chertow
- Division of Nephrology, Stanford University, Stanford, California, USA
| | - Prasad Devarajan
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Adeera Levin
- Department of Medicine, The University of British Columbia, Vancouver, Canada
| | - Sharon P Andreoli
- Department of Pediatrics, Indiana University School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - Sripal Bangalore
- Division of Cardiology, New York University, New York, New York, USA
| | - Bradley A Warady
- Division of Pediatric Nephrology, Children's Mercy Kansas City, Kansas City, Missouri, USA
| |
Collapse
|
25
|
Liu S, Wang C, Lu J, Dai G, Che H, He W. Long-term inhibition of UCHL1 decreases hypertension and retinopathy in spontaneously hypertensive rats. J Int Med Res 2021; 49:3000605211020641. [PMID: 34130526 PMCID: PMC8212382 DOI: 10.1177/03000605211020641] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To investigate the role of the deubiquitinase ubiquitin C-terminal hydrolase L1 (UCHL1) in hypertension and retinopathy in the spontaneously hypertensive rat (SHR). METHODS Wistar-Kyoto (WKY) rats and SHRs were administered the UCHL1 inhibitor LDN57444 (20 μg/kg/day) for 4 months. Pathological changes were detected with hematoxylin and eosin, immunofluorescence, and dihydroethidium staining. The mRNA and protein expression of UCHL1 were examined by real-time PCR and immunoblotting analysis. RESULTS At 6 months of age, SHRs showed significantly increased mRNA and protein levels of UCHL1 in the retina compared with WKY rats. Moreover, SHRs exhibited significantly increased central retinal thickness, inflammation, and reactive oxygen species production compared with WKY rats, and these effects were markedly attenuated by systemic administration of the UCHL1 inhibitor LDN57444. The beneficial effects of LDN57444 were possibly associated with reduced blood pressure and the inactivation of several signaling pathways. CONCLUSION UCHL1 is involved in hypertension and retinopathy in SHRs, suggesting that UCHL1 may be used as a potential therapeutic target for treating hypertensive retinopathy.
Collapse
Affiliation(s)
- Shasha Liu
- The Second Clinical College, Dalian Medical University, Dalian, P.R. China.,Health Management Center, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Chengfang Wang
- Health Management Center, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Jianmin Lu
- Department of Ophthalmology, First Affiliated Hospital of Dalian Medical University, Dalian, P.R. China
| | - Guangzheng Dai
- Clinical Research Center, He Eye Specialists Hospitals, Shenyang, P.R. China
| | - Huixin Che
- Clinical Research Center, He Eye Specialists Hospitals, Shenyang, P.R. China
| | - Wei He
- The Second Clinical College, Dalian Medical University, Dalian, P.R. China.,Clinical Research Center, He Eye Specialists Hospitals, Shenyang, P.R. China
| |
Collapse
|
26
|
康 瀚, 王 羽, 钟 晓, 殷 伟, 李 芳, 蒋 建. [Effect of salt intake on residual renal function in rats receiving peritoneal dialysis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2021; 41:264-271. [PMID: 33624601 PMCID: PMC7905259 DOI: 10.12122/j.issn.1673-4254.2021.02.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Indexed: 01/23/2023]
Abstract
OBJECTIVE To assess the effect of salt intake on residual renal function in rats and explore the possible mechanism. OBJECTIVE SD rats were 5/6-nephrectomized to induce chronic renal failure followed by peritoneal dialysis for 4 weeks (n=18) or without dialysis treatment (control group; n=18). In both groups, the rats were divided into 3 subgroups and were given lowsalt diet (0.02% NaCl), normal salt diet (0.4% NaCl), and high-salt diet (4% NaCl). After 8 and 12weeks, blood pressure and creatinine and sodium levels in the blood, urine, and peritoneal dialysate of the rats were examined. Glomerular sclerosis, tubulointerstitial fibrosis, and protein expression levels of RAS components (ACE-1, AGT, and AT-1) in renal cortical tissue of the rats were evaluated. OBJECTIVE The residual renal function of the rats all decreased especially in rats with high salt intake for 8and 12 weeks. In peritoneal dialysis group, the rats with high-salt diet showed signficiantly increased renal interstitial fibrosis score (P=0.036), glomerular sclerosis index (P=0.045), systolic blood pressure (P=0.004), diastolic blood pressure (P=0.048), and renal expressions of AGT, ACE-1, and AT1 (P < 0.05) as compared with those with normal salt intake. In the rats fed the same high-salt diet, the renal interstitial fibrosis score, glomerular sclerosis index, diastolic blood pressure increase, and renal AGT and ACE-1 expression levels were significantly lower in the peritoneal dialysis group than in the control group (P < 0.05). A positive correlation was noted between the reduction of residual renal function and sodium intake in the rats. OBJECTIVE In rats with chronic renal failure, high salt intake promotes the activation of the renal RAS system, increases blood pressure, and agrevates renal fibrosis to accelerate the decline of residual renal function, and peritoneal dialysis partially reduces the damage of residual renal function induced by high-salt diets by removing excessive sodium.
Collapse
Affiliation(s)
- 瀚 康
- 南方医科大学南方医院肾内科,广东 广州 510515Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 羽 王
- 南方医科大学南方医院肾内科,广东 广州 510515Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 晓红 钟
- 南方医科大学南方医院肾内科,广东 广州 510515Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 伟 殷
- 南方医科大学动物实验管理中心,广东 广州 510515Laboratory Animal Center, Southern Medical University, Guangzhou 510515, China
| | - 芳 李
- 南方医科大学南方医院肾内科,广东 广州 510515Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - 建平 蒋
- 南方医科大学南方医院肾内科,广东 广州 510515Department of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
27
|
Guo Y, Liu Z, Wang M. NFKB1-mediated downregulation of microRNA-106a promotes oxidative stress injury and insulin resistance in mice with gestational hypertension. Cytotechnology 2021; 73:115-126. [PMID: 33505119 DOI: 10.1007/s10616-020-00448-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 12/06/2020] [Indexed: 12/11/2022] Open
Abstract
This study intended to investigate the role of NFKB1 in oxidative stress injury and insulin resistance in gestational hypertension (GH) mice. Following establishment of a GH mouse model by high-fat diet, NFKB1, miR-106a, and FLOT2 expression was detected in liver of mice. After NFKB1, miR-106a, and FLOT2 were altered in GH mice by lentiviral vector, oxidative stress markers in liver tissues were examined by colorimetry, and insulin resistance was assessed by fasting blood glucose and fasting insulin levels. Next, hepatocytes were isolated from GH mice and treated with miR-106a mimic, inhibitor or siRNA, followed by determination of hepatocyte apoptosis and the expression of inflammation- and apoptosis-related factors. Evaluation of the correlations among NFKB1, miR-106a, and FLOT2 were conducted. Liver of GH mice harbored NFKB1 and FLOT2 upregulation and miR-106a downregulation. miR-106a was transcriptionally inhibited by NFKB1, and negatively targeted FLOT2. Oxidative stress injury and insulin resistance in GH mice and apoptosis and inflammation of hepatocytes from GH mice were decreased after silencing NFKB1 or FLOT2 or overexpressing miR-106a. These findings provided evidence demonstrating the inhibitory effect of NFKB1 silencing on oxidative stress injury and insulin resistance in GH mice via miR-106a upregulation and FLOT2 downregulation.
Collapse
Affiliation(s)
- Yunxia Guo
- Department of Obstetrics, The Second People's Hospital of Liaocheng, Liaocheng, 252600 Shandong People's Republic of China
| | - Zhaofang Liu
- Department of Obstetrics, The Second People's Hospital of Liaocheng, Liaocheng, 252600 Shandong People's Republic of China
| | - Ming Wang
- Department of Obstetrics and Gynecology, Maternity and Child Health Care of Zaozhuang, No. 25, Wenhua East Road, Shizhong District, Zaozhuang, 277100 Shandong People's Republic of China
| |
Collapse
|
28
|
Li J, Sun L, Li Y. Regulation of dimethylarginine dimethylaminohydrolase 2 expression by NF-κB acetylation. Exp Ther Med 2020; 21:114. [PMID: 33335577 PMCID: PMC7739820 DOI: 10.3892/etm.2020.9546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Accepted: 10/08/2020] [Indexed: 12/20/2022] Open
Abstract
Nitric oxide (NO) serves a crucial role in the kidney and is synthesized by NO synthase (NOS). Asymmetrical dimethylarginine is an endogenous inhibitor of NOS that is metabolized by dimethylarginine dimethylaminohydrolase (DDAH). To investigate the role of acetylation in DDAH2 expression, 293 cells were treated with trichostatin A (TSA), a deacetylase inhibitor and the mRNA and protein levels were assessed using quantitative PCR and western blotting respectively. Its promoter activity was detected using a luciferase assay. The effect of TSA on NF-κB acetylation was tested after immunoprecipitation. The binding of NF-κB to the DDAH2 promoter was analyzed using an electrophoretic mobility shift assay and chromatin immunoprecipitation. TSA upregulated DDAH2 expression and transcriptional activity of the DDAH2 promoter through a NF-κB responsive element, which is located at the -1582 to -1573 position of the DDAH2 promoter. Furthermore, TSA treatment promoted NF-κB acetylation, resulting in enhanced NF-κB binding affinity to its binding site both in vitro and in vivo. Taken together, the present study demonstrated that NF-κB acetylation upregulated DDAH2 expression by enhancing the binding ability of NF-κB to the DDAH2 promoter, resulting in increased promoter activity. The results provided a possible mechanism underlying the regulation of NO production in renal cells and a potential target for treating certain NO-associated renal disorders.
Collapse
Affiliation(s)
- Jiaqi Li
- Department of Medical Genetics, School of Life Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| | - Lu Sun
- Department of Medical Genetics, School of Life Science, China Medical University, Shenyang, Liaoning 110122, P.R. China.,Department of Clinical Genetics, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110004, P.R. China
| | - Yinghui Li
- Department of Medical Genetics, School of Life Science, China Medical University, Shenyang, Liaoning 110122, P.R. China
| |
Collapse
|
29
|
Moskalev A, Stambler I, Caruso C. Innate and Adaptive Immunity in Aging and Longevity: The Foundation of Resilience. Aging Dis 2020; 11:1363-1373. [PMID: 33269094 PMCID: PMC7673842 DOI: 10.14336/ad.2020.0603] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 06/03/2020] [Indexed: 12/15/2022] Open
Abstract
The interrelation of the processes of immunity and senescence now receives an unprecedented emphasis during the COVID-19 pandemic, which brings to the fore the critical need to combat immunosenescence and improve the immune function and resilience of older persons. Here we review the historical origins and the current state of the science of innate and adaptive immunity in aging and longevity. From the modern point of view, innate and adaptive immunity are not only affected by aging but also are important parts of its underlying mechanisms. Excessive levels or activity of antimicrobial peptides, C-reactive protein, complement system, TLR/NF-κB, cGAS/STING/IFN 1,3 and AGEs/RAGE pathways, myeloid cells and NLRP3 inflammasome, declined levels of NK cells in innate immunity, thymus involution and decreased amount of naive T-cells in adaptive immunity, are biomarkers of aging and predisposition factors for cellular senescence and aging-related pathologies. Long-living species, human centenarians, and women are characterized by less inflamm-aging and decelerated immunosenescence. Despite recent progress in understanding, the harmonious theory of immunosenescence is still developing. Geroprotectors targeting these mechanisms are just emerging and are comprehensively discussed in this article.
Collapse
Affiliation(s)
- Alexey Moskalev
- Institute of Biology of FRC of Komi Scientific Center of Ural Branch of Russian Academy of Sciences, Syktyvkar, 167982, Russia.
| | - Ilia Stambler
- Vetek (Seniority), The Movement for Longevity and Quality of Life, Israel.
| | - Calogero Caruso
- Laboratory of Immunopathology and Immunosenescence, Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| |
Collapse
|
30
|
Chen Q, Zhang P, Xiao QX, Liu Q, Zhang Y. Protective effect of Shengmai injection on myocardial endothelial cell glycoprotein detachment after myocardial ischemia-reperfusion injury in isolated rat hearts. Perfusion 2020; 36:757-765. [PMID: 33070762 DOI: 10.1177/0267659120965921] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To investigate effects of Shengmai injection (SMI) postconditioning on myocardial ischemia-reperfusion injury (MIRI) in isolated rat hearts. MATERIALS AND METHODS A total of thirty isolated hearts were randomly divided into three groups: Sham group, I/R group and SMI group. Sham group was continuously perfused with K-H solution for 120 minutes. I/R group and SMI group were given balanced perfusion for 30 min followed by reperfusion for 60 min, with an interval of 30 min, and those in the SMI group were given postconditioning with 1% SMI during the first 10 min of reperfusion. The left ventricular function, markers of myocardial injury, endothelial cell injury and oxidative stress injury were measured at 30 minutes after equilibration (t0), 30 minutes after ischemia (t2) and 60 minutes after reperfusion (t3). RESULTS The results showed that there was no significant difference for all observation indexes at t0. Compared with the Sham group, real portfolio project and coronary arterial flow rate and the activity of superoxide dismutase were significantly decreased in the I/R group, whereas those in the SMI group were significantly higher. Left ventricular end-diastolic pressure, the concentrate of malondialdehyde, lactate dehydrogenase, cTn-I, hyaluronic acid, heparin sulphate, syndecan-1 in the I/R group were markedly higher than those in the Sham group, whereas those in the SMI group were significantly lower. CONCLUSION In summary, the present study indicated that 1% SMI postconditioning can alleviate the detachment of endothelial cell glycoprotein envelope induced by myocardial ischemia-reperfusion injury, and its mechanism is probably related to the inhibition of the oxidative stress injury.
Collapse
Affiliation(s)
- Qi Chen
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ping Zhang
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qiu-Xia Xiao
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Qing Liu
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ying Zhang
- Department of Anesthesiology, Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
31
|
Comparisons of skin microvascular changes in patients with primary aldosteronism and essential hypertension. Hypertens Res 2020; 43:1222-1230. [PMID: 32488215 DOI: 10.1038/s41440-020-0475-4] [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: 02/23/2020] [Revised: 04/23/2020] [Accepted: 04/28/2020] [Indexed: 11/08/2022]
Abstract
The aim of our cross-sectional study was to evaluate skin microvascular alterations in patients with hypertension secondary to primary aldosteronism (PA) and in subjects with essential hypertension (EH). Skin microcirculation was detected by nailfold videocapillaroscopy (NVC) and laser Doppler perfusion imaging (LDPI), both noninvasive techniques for the evaluation of digital capillaroscopic damage and hand skin blood perfusion. From September 2018 to April 2019, we consecutively enrolled 80 patients, of whom 42 had PA and 38 had EH. A morphological and structural study of cutaneous microcirculation was carried out through NVC, while functional evaluation of the peripheral microcirculation was carried out with LDPI. Using LDPI indices, dermal perfusion gradients were calculated in various regions of interest at the level of the back of the hand (ROI1 and ROI2). No differences between the two groups in NVC parameters were found. In contrast, LDPI showed worse skin perfusion parameters in patients with PA compared with patients with EH (ROI1: 143.9 ± 29.9 pU vs 163.3 ± 35.2 pU, p = 0.01; perfusion gradient ROI1-ROI2: 62.4 ± 28.8 pU vs 79.3 ± 33.5 pU, p = 0.019). Furthermore, the ROI1-ROI2 (proximal-distal) perfusion gradient was negatively correlated with aldosterone plasma levels (r -0.269; p = 0.017). Multivariate analysis showed that aldosterone was significantly associated with the ROI1-ROI2 perfusion gradient (b -0.220; p = 0.044). Patients with PA showed altered skin perfusion and greater microvascular dysfunction compared with the EH group. Our results are consistent with the hypothesis that aldosterone may have a pathophysiological role in microvascular remodeling in patients with PA, with predominant functional dysfunction.
Collapse
|
32
|
Xu GR, Zhang C, Yang HX, Sun JH, Zhang Y, Yao TT, Li Y, Ruan L, An R, Li AY. Modified citrus pectin ameliorates myocardial fibrosis and inflammation via suppressing galectin-3 and TLR4/MyD88/NF-κB signaling pathway. Biomed Pharmacother 2020; 126:110071. [DOI: 10.1016/j.biopha.2020.110071] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 02/28/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
|
33
|
Shuai W, Kong B, Yang H, Fu H, Huang H. Loss of myeloid differentiation protein 1 promotes atrial fibrillation in heart failure with preserved ejection fraction. ESC Heart Fail 2020; 7:626-638. [PMID: 31994333 PMCID: PMC7160510 DOI: 10.1002/ehf2.12620] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/18/2019] [Accepted: 01/03/2020] [Indexed: 12/19/2022] Open
Abstract
AIMS Myeloid differentiation protein 1 (MD1) is expressed in the mammalian heart and exerts an anti-arrhythmic effect. Atrial fibrillation (AF) is closely related to heart failure with preserved ejection fraction (HFpEF). The potential impact of MD1 on AF vulnerability in an HFpEF model is not clear. METHODS AND RESULTS MD1 knock-out and wild-type (WT) mice were subjected to uninephrectomy and continuous saline or d-aldosterone infusion and given 1% sodium chloride drinking water for 4 weeks. Echocardiographic and haemodynamic measurements, electrophysiological studies, Masson staining, and molecular analysis were performed. Aldosterone-infused WT mice develop HFpEF with left ventricular hypertrophy, moderate hypertension, pulmonary congestion, and diastolic dysfunction. Aldosterone infusion increased the vulnerability of WT mice to AF, as shown by a prolonged interatrial conduction time, shortened effective refractory period, and higher incidence of AF. In addition, aldosterone infusion increased myocardial fibrosis and inflammation, decreased sarcoplasmic reticulum Ca2+ -ATPase 2a protein expression and the phosphorylation of phospholamban at Thr17, and increased sodium/calcium exchanger 1 protein expression and the phosphorylation of ryanodine receptor 2 in WT mice. All of the above adverse effects of aldosterone infusion were further exacerbated in MD1 knock-out mice compare with WT mice. Mechanistically, MD1 deletion increased the activation of the toll-like receptor 4/calmodulin-dependent protein kinase II signalling pathway in in vivo and in vitro experiments. CONCLUSIONS MD1 deficiency increases the vulnerability of HFpEF mice to AF. This is mainly caused by aggravated maladaptive left atrial fibrosis and inflammation and worsened dysregulation of calcium handling, which is induced by the enhanced activation of the toll-like receptor 4/calmodulin-dependent protein kinase II signalling pathway.
Collapse
Affiliation(s)
- Wei Shuai
- Department of CardiologyRenmin Hospital of Wuhan University238 Jiefang RoadWuhanHubei430060China
- Cardiovascular Research Institute of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Bin Kong
- Department of CardiologyRenmin Hospital of Wuhan University238 Jiefang RoadWuhanHubei430060China
- Cardiovascular Research Institute of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Hongjie Yang
- Department of CardiologyRenmin Hospital of Wuhan University238 Jiefang RoadWuhanHubei430060China
- Cardiovascular Research Institute of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Hui Fu
- Department of CardiologyRenmin Hospital of Wuhan University238 Jiefang RoadWuhanHubei430060China
- Cardiovascular Research Institute of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - He Huang
- Department of CardiologyRenmin Hospital of Wuhan University238 Jiefang RoadWuhanHubei430060China
- Cardiovascular Research Institute of Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
34
|
Lu G, Li J, Zhai Y, Li Q, Xie D, Zhang J, Xiao Y, Gao X. Spironolactone suppresses aldosterone-induced Kv1.5 expression by attenuating mineralocorticoid receptor-Nox1/2/4-mediated ROS generation in neonatal rat atrial myocytes. Biochem Biophys Res Commun 2019; 520:379-384. [DOI: 10.1016/j.bbrc.2019.10.039] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 10/03/2019] [Indexed: 10/25/2022]
|
35
|
Moss ME, Carvajal B, Jaffe IZ. The endothelial mineralocorticoid receptor: Contributions to sex differences in cardiovascular disease. Pharmacol Ther 2019; 203:107387. [PMID: 31271793 PMCID: PMC6848769 DOI: 10.1016/j.pharmthera.2019.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 06/26/2019] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease remains the leading cause of death for both men and women. The observation that premenopausal women are protected from cardiovascular disease relative to age-matched men, and that this protection is lost with menopause, has led to extensive study of the role of sex steroid hormones in the pathogenesis of cardiovascular disease. However, the molecular basis for sex differences in cardiovascular disease is still not fully understood, limiting the ability to tailor therapies to male and female patients. Therefore, there is a growing need to investigate molecular pathways outside of traditional sex hormone signaling to fully understand sex differences in cardiovascular disease. Emerging evidence points to the mineralocorticoid receptor (MR), a steroid hormone receptor activated by the adrenal hormone aldosterone, as one such mediator of cardiovascular disease risk, potentially serving as a sex-dependent link between cardiovascular risk factors and disease. Enhanced activation of the MR by aldosterone is associated with increased risk of cardiovascular disease. Emerging evidence implicates the MR specifically within the endothelial cells lining the blood vessels in mediating some of the sex differences observed in cardiovascular pathology. This review summarizes the available clinical and preclinical literature concerning the role of the MR in the pathophysiology of endothelial dysfunction, hypertension, atherosclerosis, and heart failure, with a special emphasis on sex differences in the role of endothelial-specific MR in these pathologies. The available data regarding the molecular mechanisms by which endothelial-specific MR may contribute to sex differences in cardiovascular disease is also summarized. A paradigm emerges from synthesis of the literature in which endothelial-specific MR regulates vascular function in a sex-dependent manner in response to cardiovascular risk factors to contribute to disease. Limitations in this field include the relative paucity of women in clinical trials and, until recently, the nearly exclusive use of male animals in preclinical investigations. Enhanced understanding of the sex-specific roles of endothelial MR could lead to novel mechanistic insights underlying sex differences in cardiovascular disease incidence and outcomes and could identify additional therapeutic targets to effectively treat cardiovascular disease in men and women.
Collapse
Affiliation(s)
- M Elizabeth Moss
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America
| | - Brigett Carvajal
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, MA, United States of America; Sackler School of Graduate Biomedical Sciences, Tufts University School of Medicine, Boston, MA, United States of America.
| |
Collapse
|
36
|
Pantelidis P, Sideris M, Viigimaa M, Avranas K, Deligkaris P, Zografou I, Lovic D. The Mechanisms of Actions of Aldosterone and its Antagonists in Cardiovascular Disease. Curr Pharm Des 2019; 24:5491-5499. [DOI: 10.2174/1381612825666190215100502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 02/10/2019] [Indexed: 11/22/2022]
Abstract
Background:
Aldosterone, through its actions on Mineralcorticosteroid Receptors (MR), controls fluid
and electrolyte balance, but also exerts various direct deleterious actions on the vasculature. A number of aldosterone
antagonists have been manufactured to reverse these effects.
Objective:
A comprehensive review of the underlying mechanisms of the actions of aldosterone and its antagonists
in cardiovascular disease.
Method:
The relevant studies indexed in PubMed, Scopus and Google Scholar databases, published from 2003 to
May 2018 were identified and reported.
Results:
Aldosterone binds to MR, activating them as intracellular transcription factors. Moreover, aldosterone,
through its actions on MR, as well as on another not fully explored class of receptors, triggers several signaling
pathways that produce rapid, non-genomic actions. In the vasculature, all these changes favor the establishment of
inflammation and cardiovascular dysfunction, which, in turn, lead to or exacerbate various cardiovascular diseases.
Mineralcorticosteroid Antagonists (MRA) are compounds that antagonize the action of aldosterone on MR.
Spironolactone was the first steroidal MRA to be commercially used. It showed beneficial clinical results, but
also a number of adverse effects. The next generation of steroidal MRA, exhibited lower potency but did not
induce many of these adverse reactions, due to their high selectivity for MR. The third generation of MRA compromises
the newly introduced non-steroidal MRA, which have a completely different chemical structure, they
induce different and more drastic changes to MR, they are much more specific and currently under clinical trials.
Conclusion:
New MRA, which block the aldosterone induced pathways in the vasculature, hold promising results
for the treatment of cardiovascular disease.
Collapse
Affiliation(s)
- Panteleimon Pantelidis
- 2nd Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Michail Sideris
- Women Health Research Unit, Queen Mary University of London, London, United Kingdom
| | - Margus Viigimaa
- Centre of Cardiology, North Estonia Medical Centre, Tallinn, Estonia; Institute of Health Technologies, Tallinn University of Technology, Tallinn, Estonia
| | - Konstantinos Avranas
- 2nd Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Pavlos Deligkaris
- 2nd Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Ioanna Zografou
- 2nd Propaedeutic Department of Internal Medicine, Aristotle University of Thessaloniki, Thessaloniki, Greece
| | - Dragan Lovic
- Clinic for Internal Disease Intermedica, Cardiology department, Hypertension Center, Nis, Serbia
| |
Collapse
|
37
|
Lynch SM, Ward M, McNulty H, Angel CZ, Horigan G, Strain JJ, Purvis J, Tackett M, McKenna DJ. Serum levels of miR-199a-5p correlates with blood pressure in premature cardiovascular disease patients homozygous for the MTHFR 677C > T polymorphism. Genomics 2019; 112:669-676. [PMID: 31029863 DOI: 10.1016/j.ygeno.2019.04.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 04/12/2019] [Accepted: 04/24/2019] [Indexed: 02/02/2023]
Abstract
This investigation profiled circulating serum concentrations of microRNAs (miRNAs) in premature cardiovascular disease (CVD) patients screened for the 677C > T polymorphism in methylenetetrahydrofolate reductase (MTHFR), a risk factor for hypertension. Serum samples from 75 premature CVD patients of known MTHFR genotype were analysed for CVD-related miRNA expression, to identify those that were associated with blood pressure. Samples were collected at baseline and following intervention with riboflavin as part of a randomized controlled trial. In patients with the MTHFR 677TT genotype, expression of miR-199a-5p in serum was inversely correlated with hypertension at baseline, and with change in blood pressure in TT genotype patients who responded to riboflavin intervention. These correlations were not observed in MTHFR 677CC genotype patients. In vitro experiments and in silico data analysis provided evidence that miR-199a-5p targets SMAD4. This is the first study to link miR-199a-5p expression with hypertension in a genetically at-risk cohort of premature CVD patients.
Collapse
Affiliation(s)
- Seodhna M Lynch
- Genomic Medicine Research Group, Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK
| | - Mary Ward
- Nutrition Innovation Centre for Food and Health (NICHE), Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK
| | - Helene McNulty
- Nutrition Innovation Centre for Food and Health (NICHE), Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK
| | - C Zoe Angel
- Genomic Medicine Research Group, Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK
| | - Geraldine Horigan
- Nutrition Innovation Centre for Food and Health (NICHE), Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK
| | - J J Strain
- Nutrition Innovation Centre for Food and Health (NICHE), Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK
| | - John Purvis
- Department of Cardiology, Altnagelvin Area Hospital, Western Health and Social Care Trust, Derry, Northern Ireland, UK
| | - Mike Tackett
- Abcam, One Kendall Square Suite B2304, Cambridge, MA 02139, USA
| | - Declan J McKenna
- Genomic Medicine Research Group, Biomedical Sciences Research Institute, Ulster University, Cromore Road, Coleraine BT52 1SA, Northern Ireland, UK.
| |
Collapse
|
38
|
Cao Z, Jia Y, Zhu B. BNP and NT-proBNP as Diagnostic Biomarkers for Cardiac Dysfunction in Both Clinical and Forensic Medicine. Int J Mol Sci 2019; 20:ijms20081820. [PMID: 31013779 PMCID: PMC6515513 DOI: 10.3390/ijms20081820] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/09/2019] [Accepted: 04/11/2019] [Indexed: 12/23/2022] Open
Abstract
Currently, brain natriuretic peptide (BNP) and N-terminal proBNP (NT-proBNP) are widely used as diagnostic biomarkers for heart failure (HF) and cardiac dysfunction in clinical medicine. They are also used as postmortem biomarkers reflecting cardiac function of the deceased before death in forensic medicine. Several previous studies have reviewed BNP and NT-proBNP in clinical medicine, however, few articles have reviewed their application in forensic medicine. The present article reviews the biological features, the research and application status, and the future research prospects of BNP and NT-proBNP in both clinical medicine and forensic medicine, thereby providing valuable assistance for clinicians and forensic pathologists.
Collapse
Affiliation(s)
- Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang 110122, China.
| | - Yuqing Jia
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang 110122, China.
| | - Baoli Zhu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang 110122, China.
| |
Collapse
|
39
|
Musolino V, Palus S, Latouche C, Gliozzi M, Bosco F, Scarano F, Nucera S, Carresi C, Scicchitano M, von Haehling S, Jaisser F, Hasenfuss G, Anker SD, Mollace V, Springer J. Cardiac expression of neutrophil gelatinase-associated lipocalin in a model of cancer cachexia-induced cardiomyopathy. ESC Heart Fail 2019; 6:89-97. [PMID: 30367561 PMCID: PMC6352893 DOI: 10.1002/ehf2.12372] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 09/24/2018] [Indexed: 12/13/2022] Open
Abstract
AIMS Cachexia is a severe consequence of cancer. Although cancer-induced heart atrophy leads to cardiac dysfunction and heart failure (HF), biomarkers for their diagnosis have not been identified. Neutrophil gelatinase-associated lipocalin (NGAL) is an aldosterone-responsive gene increased in HF. We studied NGAL and its association with aldosterone levels in a model of cancer cachexia-induced cardiomyopathy. METHODS AND RESULTS Rats were injected with Yoshida 108 AH-130 hepatoma cells to induce tumour. Cachectic rats were treated daily, for 16 days, with placebo or with 5 or 50 mg/kg/day of spironolactone. Cardiac function was analysed by echocardiography at baseline and at Day 11. Weight loss and atrophy of lean body and fat mass of cachectic rats were significantly attenuated by spironolactone. Cardiac dysfunction of tumour-bearing rats was improved by spironolactone. Plasma aldosterone was up-regulated from 337 ± 7 pg/mL in sham animals to 591 ± 31 pg/mL in the cachectic rats (P < 0.001 vs. sham). Treatment with 50 or 5 mg/kg/day of spironolactone reduced plasma aldosterone to 396 ± 22 and 391 ± 25 pg/mL (P < 0.01 vs. placebo). Plasma levels of NGAL were also increased in cachectic rats (1.462 ± 0.3603 μg/mL) than in controls (0.0936 ± 6 μg/mL, P < 0.001). Spironolactone treatment (50 mg/kg/day) significantly reduced cardiac mRNA and protein NGAL levels (P < 0.05 and P < 0.001 vs. placebo, respectively). NGAL mRNA and protein levels were overexpressed in cachectic animal hearts treated with placebo, compared with control (P < 0.05 and P < 0.01 vs. sham). Spironolactone treatment at 50 mg/kg/day reduced significantly cardiac NGAL (P < 0.05 and P < 0.001 vs. placebo). CONCLUSIONS Cancer cachexia induced increased levels of aldosterone and NGAL, contributing to worsening cardiac damage in cancer cachexia-induced cardiomyopathy. Spironolactone treatment may greatly attenuate cardiac dysfunction and lean mass atrophy associated with cancer cachexia.
Collapse
Affiliation(s)
- Vincenzo Musolino
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Sandra Palus
- Department of CardiologyUniversity Medical Center Göttingen (UMG)GöttingenGermany
| | | | - Micaela Gliozzi
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Francesca Bosco
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Federica Scarano
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Saverio Nucera
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Cristina Carresi
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Miriam Scicchitano
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Stephan von Haehling
- Department of CardiologyUniversity Medical Center Göttingen (UMG)GöttingenGermany
| | | | - Gerd Hasenfuss
- Department of CardiologyUniversity Medical Center Göttingen (UMG)GöttingenGermany
| | - Stefan D. Anker
- Division of Cardiology and Metabolism – Heart Failure, Cachexia & Sarcopenia, Department of Cardiology (CVK); and Berlin‐Brandenburg Center for Regenerative Therapies (BCRT); Deutsches Zentrum für Herz‐Kreislauf‐Forschung (DZHK) Berlin; Charité Universitätsmedizin BerlinBerlinGermany
| | - Vincenzo Mollace
- Institute of Research for Food Safety & Health (IRC‐FSH)University of Catanzaro “Magna Graecia”CatanzaroItaly
| | - Jochen Springer
- Department of CardiologyUniversity Medical Center Göttingen (UMG)GöttingenGermany
| |
Collapse
|
40
|
Buonafine M, Bonnard B, Jaisser F. Mineralocorticoid Receptor and Cardiovascular Disease. Am J Hypertens 2018; 31:1165-1174. [PMID: 30192914 DOI: 10.1093/ajh/hpy120] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 08/09/2018] [Indexed: 12/14/2022] Open
Abstract
Activation of the mineralocorticoid receptor (MR) in the distal nephron by its ligand, aldosterone, plays an important role in sodium reabsorption and blood pressure regulation. However, expression of the MR goes beyond the kidney. It is expressed in a variety of other tissues in which its activation could lead to tissue injury. Indeed, MR activation in the cardiovascular (CV) system has been shown to promote hypertension, fibrosis, and inflammation. Pharmacological blockade of the MR has protective effects in several animal models of CV disease. Furthermore, the use of MR antagonists is beneficial for heart failure patients, preventing mortality and morbidity. A better understanding of the implications of the MR in the setting of CV diseases is critical for refining treatments and improving patient care. The mechanisms involved in the deleterious effects of MR activation are complex and include oxidative stress, inflammation, and fibrosis. This review will discuss the pathological role of the MR in the CV system and the major mechanisms underlying it.
Collapse
Affiliation(s)
- Mathieu Buonafine
- INSERM, UMRS, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France
- Paris Descartes University, Paris, France
| | - Benjamin Bonnard
- INSERM, UMRS, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France
- Paris Descartes University, Paris, France
| | - Frédéric Jaisser
- INSERM, UMRS, Centre de Recherche des Cordeliers, Sorbonne University, Paris, France
- Paris Descartes University, Paris, France
- INSERM, Clinical Investigation Centre, French-Clinical Research Infrastructure Network (F-CRIN) INI-CRCT, RHU Fight-HF, Nancy, France
| |
Collapse
|
41
|
Wu F, Lin Y, Liu Q. The emerging role of aldosterone/mineralocorticoid receptors in the pathogenesis of erectile dysfunction. Endocrine 2018; 61:372-382. [PMID: 29721801 DOI: 10.1007/s12020-018-1610-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 04/17/2018] [Indexed: 12/26/2022]
Abstract
PURPOSE Aldosterone is an old hormone that has been discovered for more than fifty years. The clinical application of its receptors' inhibitors, especially spirolactone, has benifited patients for decades worldwide. In this review, we briefly summarized the molecular mechanism of aldosterone/mineralocorticoid receptors (Ald-MRs) signaling in cardiovascular diseases and its emerging role in erectile dysfunction. METHODS We searched PubMed, Web of Science, and Scopus for manuscripts published prior to December 2017 using key words " aldosterone " AND " erectile dysfunction " OR " cardiovascular disease " OR " mineralocorticoid receptors ". Related literature and clinical perspectives were collated, summarized and discussed in this review. RESULTS The increase of reactive oxygen species production, inhibition of endothelial nitric oxide synthase system, and induction of inflammation are ubiquitous in vascular endothelial cells or vascular smooth muscle cells after the activation of Ald-MRs pathway. In addition, in cardiovascular diseases with over-active Ald-MRs signaling, MRs blockade could reverse the injury and improve the prognosis. Notably, multiple studies have correlated aldosterone and MRs to the pathogenesis of erectile function, while the mechanism is largely unperfectly identified. CONCLUSION In conclusion, we summarize the current evidence to highlight the potential role of aldosterone in erectile dysfunction and provide critical insights into the treatment of the disease.
Collapse
Affiliation(s)
- Fei Wu
- Department of Urology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China.
| | - Yun Lin
- Department of Urology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China
| | - Qingyong Liu
- Department of Urology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250014, China.
| |
Collapse
|
42
|
Patel B, Mann GE, Chapple SJ. Concerted redox modulation by sulforaphane alleviates diabetes and cardiometabolic syndrome. Free Radic Biol Med 2018; 122:150-160. [PMID: 29427794 DOI: 10.1016/j.freeradbiomed.2018.02.004] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/01/2018] [Accepted: 02/03/2018] [Indexed: 02/07/2023]
Abstract
Diabetes and cardiometabolic disorders such as hypertension and obesity are major risk factors for the development of cardiovascular disease, with a wealth of evidence suggesting that oxidative stress is linked to the initiation and pathogenesis of these disease processes. With yearly increases in the global incidence of cardiovascular diseases (CVD) and diabetes, numerous studies have focused on characterizing whether upregulating antioxidant defenses through exogenous antioxidants (e.g. vitamin E, vitamin C) or activation of endogenous defenses (e.g. the Nuclear factor erythroid 2-related factor 2 (Nrf2) antioxidant defense pathway) may be of benefit. The dietary isothiocyanate sulforaphane (SFN) is currently the subject of several clinical trials for a variety of disease states, including the evaluation of its therapeutic potential to ameliorate diabetic and cardiometabolic complications. SFN is a well characterized and potent Nrf2 inducer, however recent studies suggest its protective actions may be in part mediated by its modulation of various pro-inflammatory (e.g. Nuclear factor-kappa B (NFκB)) and metabolic (e.g. Peroxisome Proliferator-Activator Receptor Gamma (PPARγ)) signaling pathways. The focus of this review is to provide a detailed analysis of the known mechanisms by which SFN modulates Nrf2, NFκB and PPARγ signaling and crosstalk and to provide a critical evaluation of the evidence linking these transcriptional pathways with diabetic and cardiometabolic complications and SFN mediated cytoprotection. To allow comparison between rodent and human studies, we discuss the published bioavailability of SFN metabolites achieved in rodents and man in the context of Nrf2, NFκB and PPARγ signaling. Furthermore, we provide an update on the functional outcomes and implicated signaling pathways reported in recent clinical trials with SFN in Type 2 diabetic patients.
Collapse
Affiliation(s)
- Bijal Patel
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Giovanni E Mann
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom
| | - Sarah J Chapple
- King's BHF Centre of Research Excellence, School of Cardiovascular Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London SE1 9NH, United Kingdom.
| |
Collapse
|
43
|
Rodríguez-Lara SQ, García-Benavides L, Miranda-Díaz AG. The Renin-Angiotensin-Aldosterone System as a Therapeutic Target in Late Injury Caused by Ischemia-Reperfusion. Int J Endocrinol 2018; 2018:3614303. [PMID: 29849615 PMCID: PMC5904808 DOI: 10.1155/2018/3614303] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 01/09/2018] [Accepted: 02/07/2018] [Indexed: 12/19/2022] Open
Abstract
Ischemia-reperfusion (I/R) injury is a well-known phenomenon that involves different pathophysiological processes. Connection in diverse systems of survival brings about cellular dysfunction or even apoptosis. One of the survival systems of the cells, to the assault caused by ischemia, is the activation of the renin-angiotensin-aldosterone system (also known as an axis), which is focused on activating diverse signaling pathways to favor adaptation to the decrease in metabolic supports caused by the hypoxia. In trying to adapt to the I/R event, great changes occur that unchain cellular dysfunction with the capacity to lead to cell death, which translates into a poor prognosis due to the progression of dysfunction of the cellular activity. The search for the understanding of the diverse therapeutic alternatives in molecular coupling could favor the prognosis and evolution of patients who are subject to the I/R process.
Collapse
Affiliation(s)
- Simón Quetzalcóatl Rodríguez-Lara
- University of Guadalajara, Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, Guadalajara, JAL, Mexico
| | - Leonel García-Benavides
- University of Guadalajara, Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, Guadalajara, JAL, Mexico
| | - Alejandra Guillermina Miranda-Díaz
- University of Guadalajara, Institute of Experimental and Clinical Therapeutics, Department of Physiology, University Health Sciences Centre, Guadalajara, JAL, Mexico
| |
Collapse
|
44
|
Venter M, van der Westhuizen FH, Elson JL. The aetiology of cardiovascular disease: a role for mitochondrial DNA? Cardiovasc J Afr 2017; 29:122-132. [PMID: 28906532 PMCID: PMC6009096 DOI: 10.5830/cvja-2017-037] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Accepted: 07/17/2017] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular disease (CVD) is a world-wide cause of mortality in humans and its incidence is on the rise in Africa. In this review, we discuss the putative role of mitochondrial dysfunction in the aetiology of CVD and consequently identify mitochondrial DNA (mtDNA) variation as a viable genetic risk factor to be considered. We then describe the contribution and pitfalls of several current approaches used when investigating mtDNA in relation to complex disease. We also propose an alternative approach, the adjusted mutational load hypothesis, which would have greater statistical power with cohorts of moderate size, and is less likely to be affected by population stratification. We therefore address some of the shortcomings of the current haplogroup association approach. Finally, we discuss the unique challenges faced by studies done on African populations, and recommend the most viable methods to use when investigating mtDNA variation in CVD and other common complex disease.
Collapse
Affiliation(s)
- Marianne Venter
- Human Metabolomics, North-West University, Potchefstroom, South Africa.
| | | | - Joanna L Elson
- Human Metabolomics, North-West University, Potchefstroom, South Africa; Institute of Genetic Medicine, Newcastle University, United Kingdom
| |
Collapse
|
45
|
Paricalcitol Pretreatment Attenuates Renal Ischemia-Reperfusion Injury via Prostaglandin E 2 Receptor EP4 Pathway. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:5031926. [PMID: 28465762 PMCID: PMC5390586 DOI: 10.1155/2017/5031926] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 02/23/2017] [Indexed: 11/18/2022]
Abstract
The protective mechanism of paricalcitol remains unclear in renal ischemia-reperfusion (IR) injury. We investigated the renoprotective effects of paricalcitol in IR injury through the prostaglandin E2 (PGE2) receptor EP4. Paricalcitol was injected into IR-exposed HK-2 cells and mice subjected to bilateral kidney ischemia for 23 min and reperfusion for 24 hr. Paricalcitol prevented IR-induced cell death and EP4 antagonist cotreatment offset these protective effects. Paricalcitol increased phosphorylation of Akt and cyclic AMP responsive element binding protein (CREB) and suppressed nuclear factor-κB (NF-κB) in IR-exposed cells and cotreatment of EP4 antagonist or EP4 small interfering RNA blunted these signals. In vivo studies showed that paricalcitol improved renal dysfunction and tubular necrosis after IR injury and cotreatment with EP4 antagonist inhibited the protective effects of paricalcitol. Phosphorylation of Akt was increased and nuclear translocation of p65 NF-κB was decreased in paricalcitol-treated mice with IR injury, which was reversed by EP4 blockade. Paricalcitol decreased oxidative stress and apoptosis in renal IR injury. Paricalcitol also attenuated the infiltration of inflammatory cells and production of proinflammatory cytokines after IR injury. EP4 antagonist abolished these antioxidant, anti-inflammatory, and antiapoptotic effects. The EP4 plays a pivotal role in the protective effects of paricalcitol in renal IR injury.
Collapse
|
46
|
Erboga M, Kanter M, Aktas C, Sener U, Fidanol Erboga Z, Bozdemir Donmez Y, Gurel A. Thymoquinone Ameliorates Cadmium-Induced Nephrotoxicity, Apoptosis, and Oxidative Stress in Rats is Based on its Anti-Apoptotic and Anti-Oxidant Properties. Biol Trace Elem Res 2016; 170:165-72. [PMID: 26226832 DOI: 10.1007/s12011-015-0453-x] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 07/20/2015] [Indexed: 12/22/2022]
Abstract
Cadmium (Cd), an environmental and industrial pollutant, generates free radicals responsible for oxidative stress. Cd can also lead to various renal toxic damage such as the proximal tubules and glomerulus dysfunction. Thymoquinone (TQ) is the main constituent of the essential oil obtained from black seeds (Nigella sativa) and has various pharmacological effects. The aim of the present study was to examine the nephroprotective, anti-oxidant, and anti-apoptotic effect of the TQ against Cd-induced nephrotoxicity. A total of 24 male Wistar albino rats were divided into three groups: control, Cd-treated, and Cd-treated with TQ; each group contain eight animals. The Cd-treated group was injected subcutaneously with CdCl2 dissolved in saline in the amount of 2 ml/kg/day for 30 days, resulting in a dosage of 1 mg/kg Cd. The rats in TQ-treated groups were given TQ (50 mg/kg body weight) once a day orally together with first Cd injection during the study period. The histopathological studies in the kidney of rats also showed that TQ markedly reduced the toxicity of Cd and preserved the normal histological architecture of the renal tissue. Immunohistochemical analysis revealed that TQ significantly decreased the Cd-induced over expression of nuclear factor-κB in renal tissue. Furthermore, TQ treatment resulted in decreased the number of apoptotic cells. TQ significantly suppressed lipid peroxidation, compensated deficits in the anti-oxidant defenses (reduced superoxide dismutase, glutathione peroxidase and catalase activities) in renal tissue resulted from Cd administration. These findings suggest that the nephroprotective potential of TQ in Cd toxicity might be due to its anti-oxidant and anti-apoptotic properties, which could be useful for achieving optimum effects in Cd-induced nephrotoxicity.
Collapse
Affiliation(s)
- Mustafa Erboga
- Department of Histology and Embryology, Faculty of Medicine, University of Namik Kemal, Tekirdag, Turkey.
| | - Mehmet Kanter
- Department of Histology and Embryology, Faculty of Medicine, University of Istanbul Medeniyet, Istanbul, Turkey
| | - Cevat Aktas
- Department of Histology and Embryology, Faculty of Medicine, University of Namik Kemal, Tekirdag, Turkey
| | - Umit Sener
- Department of Physiology, Faculty of Medicine, University of Namik Kemal, Tekirdag, Turkey
| | - Zeynep Fidanol Erboga
- Department of Histology and Embryology, Faculty of Medicine, University of Namik Kemal, Tekirdag, Turkey
| | - Yeliz Bozdemir Donmez
- Department of Histology and Embryology, Faculty of Medicine, University of Namik Kemal, Tekirdag, Turkey
| | - Ahmet Gurel
- Department of Biochemistry, Faculty of Medicine, University of Namik Kemal, Tekirdag, Turkey
| |
Collapse
|
47
|
Jaisser F, Farman N. Emerging Roles of the Mineralocorticoid Receptor in Pathology: Toward New Paradigms in Clinical Pharmacology. Pharmacol Rev 2016; 68:49-75. [PMID: 26668301 DOI: 10.1124/pr.115.011106] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The mineralocorticoid receptor (MR) and its ligand aldosterone are the principal modulators of hormone-regulated renal sodium reabsorption. In addition to the kidney, there are several other cells and organs expressing MR, in which its activation mediates pathologic changes, indicating potential therapeutic applications of pharmacological MR antagonism. Steroidal MR antagonists have been used for decades to fight hypertension and more recently heart failure. New therapeutic indications are now arising, and nonsteroidal MR antagonists are currently under development. This review is focused on nonclassic MR targets in cardiac, vascular, renal, metabolic, ocular, and cutaneous diseases. The MR, associated with other risk factors, is involved in organ fibrosis, inflammation, oxidative stress, and aging; for example, in the kidney and heart MR mediates hormonal tissue-specific ion channel regulation. Genetic and epigenetic modifications of MR expression/activity that have been documented in hypertension may also present significant risk factors in other diseases and be susceptible to MR antagonism. Excess mineralocorticoid signaling, mediated by aldosterone or glucocorticoids binding, now appears deleterious in the progression of pathologies that may lead to end-stage organ failure and could therefore benefit from the repositioning of pharmacological MR antagonists.
Collapse
Affiliation(s)
- F Jaisser
- INSERM UMR 1138 Team 1, Cordeliers Research Center, Pierre et Marie Curie University, Paris, France (F.J., N.F); and University Paris-Est Creteil, Creteil, France (F.J.)
| | - N Farman
- INSERM UMR 1138 Team 1, Cordeliers Research Center, Pierre et Marie Curie University, Paris, France (F.J., N.F); and University Paris-Est Creteil, Creteil, France (F.J.)
| |
Collapse
|
48
|
The accumulation of brain water-free sodium is associated with ischemic damage independent of the blood pressure in female rats. Brain Res 2015; 1616:37-44. [PMID: 25957792 DOI: 10.1016/j.brainres.2015.04.051] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Revised: 04/25/2015] [Accepted: 04/27/2015] [Indexed: 12/28/2022]
Abstract
Estrogen deficiency worsens ischemic stroke outcomes. In ovariectomized (OVX(+)) rats fed a high-salt diet (HSD), an increase in the body Na(+)/water ratio, which characterizes water-free Na(+) accumulation, was associated with detrimental vascular effects independent of the blood pressure (BP). We hypothesized that an increase in brain water-free Na(+) accumulation is associated with ischemic brain damage in OVX(+)/HSD rats. To test our hypothesis we divided female Wistar rats into 4 groups, OVX(+) and OVX(-) rats fed HSD or a normal diet (ND), and subjected them to transient cerebral ischemia. The brain Na(+)/water ratio was increased even in OVX(+)/ND rats and augmented in OVX(+)/HSD rats. The increase in the brain Na(+)/water ratio was positively correlated with expansion of the cortical infarct volume without affecting the BP. Interestingly, OVX(+) was associated with the decreased expression of ATP1α3, a subtype of the Na(+) efflux pump. HSD increased the expression of brain Na(+) influx-related molecules and the mineralocorticoid receptor (MR). The pretreatment of OVX(+)/HSD rats with the MR antagonist eplerenone reduced brain water-free Na(+) accumulation, up-regulated ATP1α3, down-regulated MR, and reduced the cortical infarct volume. Our findings show that the increase in the brain Na(+)/water ratio elicited by estrogen deficiency or HSD is associated with ischemic brain damage BP-independently, suggesting the importance of regulating the accumulation of brain water-free Na(+). The up-regulation of ATP1α3 and the down-regulation of MR may provide a promising therapeutic strategy to attenuate ischemic brain damage in postmenopausal women.
Collapse
|
49
|
Atkinson S, Crocker D, Houser D, Mashburn K. Stress physiology in marine mammals: how well do they fit the terrestrial model? J Comp Physiol B 2015; 185:463-86. [PMID: 25913694 DOI: 10.1007/s00360-015-0901-0] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 03/23/2015] [Accepted: 04/09/2015] [Indexed: 12/28/2022]
Abstract
Stressors are commonly accepted as the causal factors, either internal or external, that evoke physiological responses to mediate the impact of the stressor. The majority of research on the physiological stress response, and costs incurred to an animal, has focused on terrestrial species. This review presents current knowledge on the physiology of the stress response in a lesser studied group of mammals, the marine mammals. Marine mammals are an artificial or pseudo grouping from a taxonomical perspective, as this group represents several distinct and diverse orders of mammals. However, they all are fully or semi-aquatic animals and have experienced selective pressures that have shaped their physiology in a manner that differs from terrestrial relatives. What these differences are and how they relate to the stress response is an efflorescent topic of study. The identification of the many facets of the stress response is critical to marine mammal management and conservation efforts. Anthropogenic stressors in marine ecosystems, including ocean noise, pollution, and fisheries interactions, are increasing and the dramatic responses of some marine mammals to these stressors have elevated concerns over the impact of human-related activities on a diverse group of animals that are difficult to monitor. This review covers the physiology of the stress response in marine mammals and places it in context of what is known from research on terrestrial mammals, particularly with respect to mediator activity that diverges from generalized terrestrial models. Challenges in conducting research on stress physiology in marine mammals are discussed and ways to overcome these challenges in the future are suggested.
Collapse
Affiliation(s)
- Shannon Atkinson
- School of Fisheries and Ocean Sciences, Juneau Center, University of Alaska Fairbanks, 17101 Pt. Lena Loop Road, Juneau, AK, 99801, USA,
| | | | | | | |
Collapse
|
50
|
Gomez-Sanchez EP. Brain mineralocorticoid receptors in cognition and cardiovascular homeostasis. Steroids 2014; 91:20-31. [PMID: 25173821 PMCID: PMC4302001 DOI: 10.1016/j.steroids.2014.08.014] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2014] [Revised: 07/10/2014] [Accepted: 08/04/2014] [Indexed: 12/20/2022]
Abstract
Mineralocorticoid receptors (MR) mediate diverse functions supporting osmotic and hemodynamic homeostasis, response to injury and inflammation, and neuronal changes required for learning and memory. Inappropriate MR activation in kidneys, heart, vessels, and brain hemodynamic control centers results in cardiovascular and renal pathology and hypertension. MR binds aldosterone, cortisol and corticosterone with similar affinity, while the glucocorticoid receptor (GR) has less affinity for cortisol and corticosterone. As glucocorticoids are more abundant than aldosterone, aldosterone activates MR in cells co-expressing enzymes with 11β-hydroxydehydrogenase activity to inactivate them. MR and GR co-expressed in the same cell interact at the molecular and functional level and these functions may be complementary or opposing depending on the cell type. Thus the balance between MR and GR expression and activation is crucial for normal function. Where 11β-hydroxydehydrogenase 2 (11β-HSD2) that inactivates cortisol and corticosterone in aldosterone target cells of the kidney and nucleus tractus solitarius (NTS) is not expressed, as in most neurons, MR are activated at basal glucocorticoid concentrations, GR at stress concentrations. An exception may be pre-autonomic neurons of the PVN which express MR and 11β-HSD1 in the absence of hexose-6-phosphate dehydrogenase required to generate the requisite cofactor for reductase activity, thus it acts as a dehydrogenase. MR antagonists, valuable adjuncts to the treatment of cardiovascular disease, also inhibit MR in the brain that are crucial for memory formation and exacerbate detrimental effects of excessive GR activation on cognition and mood. 11β-HSD1 inhibitors combat metabolic and cognitive diseases related to glucocorticoid excess, but may exacerbate MR action where 11β-HSD1 acts as a dehydrogenase, while non-selective 11β-HSD1&2 inhibitors cause injurious disruption of MR hemodynamic control. MR functions in the brain are multifaceted and optimal MR:GR activity is crucial. Therefore selectively targeting down-stream effectors of MR specific actions may be a better therapeutic goal.
Collapse
Affiliation(s)
- Elise P Gomez-Sanchez
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS 39216, USA.
| |
Collapse
|