1
|
Yang S, Wang D, Wang H, Shao J, Tang C. Design, synthesis, and biological evaluation of TRPV4-KCa2.3 coupling enhancers as novel therapeutic agents for hypertension. Eur J Med Chem 2025; 294:117724. [PMID: 40409057 DOI: 10.1016/j.ejmech.2025.117724] [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: 03/24/2025] [Revised: 04/28/2025] [Accepted: 05/04/2025] [Indexed: 05/25/2025]
Abstract
Although current treatment strategies for hypertension are well-developed, there remains a group of patients who do not respond adequately to available medications. As a result, the identification of new therapeutic targets and the design of target-specific drugs are crucial directions for the future management of hypertension. Our previous research identified the TRPV4-KCa2.3 complex as a novel target for hypertension treatment, leading to the discovery of the positive compound JNc-440. Using JNc-440 as a lead molecule, 21 compounds were designed and synthesized across five distinct series. Among these, representative compounds IB-2 and II-9 demonstrated the ability to restore the coupling of the decoupled complex under hypertensive conditions and significantly reduced blood pressure in a high salt-induced hypertensive mouse model. This work lays a foundation for the future development of novel therapeutics targeting hypertension.
Collapse
Affiliation(s)
- Shaying Yang
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
| | - Dongyu Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Huabing Wang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Junlan Shao
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China
| | - Chunlei Tang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi, China.
| |
Collapse
|
2
|
Barbosa HE, da Silva AB, Nazar PHO, Bertoloni RR, de Oliveira-Filho AGS, Nikolaou S. Reactivity of trinuclear ruthenium acetates with nitrite and nitric oxide ligands in aqueous media. Dalton Trans 2025; 54:9388-9398. [PMID: 40407811 DOI: 10.1039/d5dt00630a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/11/2025]
Abstract
The chemical reactivity of nitrosyl- and nitrite-coordinated compounds in an aqueous environment is a vital part of understanding the action of these compounds as potential nitric oxide-releasing molecules (NORMs). This work reports the behaviour of the [Ru3O(CH3COO)6(py)2NO2] (1) complex, which is an isomeric mixture of nitrite-N and nitrite-O, and the nitrosyl complex [Ru3O(CH3COO)6(py)2NO]PF6 (2) in aqueous medium with and without light irradiation. NO release under light irradiation was detected through chronoamperometry, which showed that nitrite complex 1 produces NO but is less effective than nitrosyl complex 2. This difference is due to the mechanism of NO production by complex 1, which depends on the nitrite-O isomer, present in minor proportion in the synthetic sample, as shown by computational and NMR data. The reactivity of these compounds in the dark was investigated under various pH values. The nitrite complex 1 had the coordinated nitrite converted to NO+, with a pK = 4.2. NO+ was readily released, yielding the solvate species [Ru3O(CH3COO)6(py)2S]+. For the nitrosyl complex 2, two successive nucleophilic attacks by hydroxide ions were observed producing the [Ru3O(CH3COO)6(py)2HNO2] (3) and [Ru3O(CH3COO)6(py)2NO2]- (4) compounds, with pK values of 9.8 and 12.3, respectively. In buffered solutions (TRIS.HCl and PBS), the kinetic trace for the conversion of 2 to 3 suggested an induction period followed by the complete conversion to [Ru3O(CH3COO)6(py)2HNO2] at pH values where the nitrosyl [Ru3O(CH3COO)6(py)2NO]+ should be the major species. Based on these observations, our data suggest a sequence of steps in which compound 3 accumulates and then, with the aid of the buffer components, increases the rate of its own formation.
Collapse
Affiliation(s)
- Hugo E Barbosa
- Departamento de Química, LABiQSC2 - Laboratório de Atividade Biológica e Química Supramolecular de Compostos de Coordenação, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, ZIPCODE 14040-901, Ribeirão Preto-SP, Brazil.
| | - Amanda B da Silva
- Departamento de Química, LABiQSC2 - Laboratório de Atividade Biológica e Química Supramolecular de Compostos de Coordenação, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, ZIPCODE 14040-901, Ribeirão Preto-SP, Brazil.
| | - Pedro H O Nazar
- Departamento de Química, LABiQSC2 - Laboratório de Atividade Biológica e Química Supramolecular de Compostos de Coordenação, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, ZIPCODE 14040-901, Ribeirão Preto-SP, Brazil.
| | - Renan R Bertoloni
- Departamento de Química, LABiQSC2 - Laboratório de Atividade Biológica e Química Supramolecular de Compostos de Coordenação, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, ZIPCODE 14040-901, Ribeirão Preto-SP, Brazil.
| | - Antonio G S de Oliveira-Filho
- Departamento de Química, LABiQSC2 - Laboratório de Atividade Biológica e Química Supramolecular de Compostos de Coordenação, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, ZIPCODE 14040-901, Ribeirão Preto-SP, Brazil.
- Instituto de Química de São Carlos, Universidade de São Paulo, Av. Trabalhador São-Carlense, 400, 13566-590, São Carlos-SP, Brazil
| | - Sofia Nikolaou
- Departamento de Química, LABiQSC2 - Laboratório de Atividade Biológica e Química Supramolecular de Compostos de Coordenação, Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, Universidade de São Paulo, ZIPCODE 14040-901, Ribeirão Preto-SP, Brazil.
| |
Collapse
|
3
|
Fan H, Yang Z, Ying H, Zhao J, Wang X, Gong J, Li L, Liu X, Gong T, Ke Q, Zhuang L, Liang P. iPSC-derived cardiomyocytes and engineered heart tissues reveal suppressed JAK2/STAT3 signaling in LMNA-related emery-dreifuss muscular dystrophy. Redox Biol 2025; 83:103638. [PMID: 40286437 PMCID: PMC12059692 DOI: 10.1016/j.redox.2025.103638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Revised: 04/09/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025] Open
Abstract
LMNA mutation related Emery-Dreifuss muscular dystrophy (LMNA-related EDMD), is a rare genetic disorder often involving life-threatening cardiac complications. However, the molecular links between LMNA mutations and their related EDMD cardiac phenotypes have remained unclear. Here, using EDMD patient-specific and genome-edited induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs), we link the LMNA L204P mutation with the pathogenic phenotypes of arrhythmia and contractile dysfunction. Using multi-omics analysis, we then show that LMNA L204P results in decreased chromatin accessibility, leading to the downregulation of JAK2 in EDMD iPSC-CMs. Mechanistically, JAK2/STAT3 signaling pathway suppression in EDMD iPSC-CMs is shown to cause mitochondrial dysfunction and oxidative stress, ultimately resulting in the above phenotypes. Conversely, pharmacological or genetic activation of JAK2/STAT3 signaling effectively rescues both the arrhythmic and contractile dysfunction phenotypes in EDMD iPSC-CMs via improvements in mitochondrial function. In addition, whilst EDMD engineered heart tissues (EHTs) display dysfunctional contractile force generation, this can also be significantly alleviated by STAT3 activation. Taken together, we present chromatin compartment change-mediated JAK2/STAT3 suppression as a novel mechanism underlying cardiac pathogenic phenotypes in LMNA-related EDMD. Our findings indicate that activating the JAK2/STAT3 signaling pathway may hold the potential to serve as a novel therapeutic strategy for this condition.
Collapse
Affiliation(s)
- Hangping Fan
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Zongkuai Yang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Hangying Ying
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, 310016, China
| | - Jiuxiao Zhao
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xiaochen Wang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China
| | - Junhao Gong
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518052, China
| | - Lingying Li
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518052, China
| | - Xujie Liu
- Shenzhen Key Laboratory of Cardiovascular Disease, Fuwai Hospital Chinese Academy of Medical Sciences, Shenzhen, 518052, China
| | - Tingyu Gong
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, 310015, China.
| | - Qing Ke
- Department of Neurology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Lenan Zhuang
- Department of Veterinary Medicine, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China.
| | - Ping Liang
- Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China; Institute of Translational Medicine, Zhejiang University, Hangzhou, 310029, China.
| |
Collapse
|
4
|
Zhu L, Wang Q, Du J, Li X, Meng Q, Lu J, Miao Y, Li Y. Non-central symmetric 2D bismuth-based perovskites for piezoelectric-enhanced sonodynamic immunotherapy. J Colloid Interface Sci 2025; 687:386-401. [PMID: 39965436 DOI: 10.1016/j.jcis.2025.02.071] [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: 01/06/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/20/2025]
Abstract
Sonodynamic therapy (SDT), an emerging treatment modality, exhibits great potential in cancer therapy owing to its excellent tissue penetration, immune activation ability, and relatively low side effects. The lattice distortion of inorganic perovskite is challenging to control, which leads to an unsatisfactory SDT effect. This study presents a two-dimensional bismuth-based halide perovskite material, MA3Bi2Cl9-PEG (MBCP), with favorable piezoelectric properties, being first applied to tumor sonodynamic immunotherapy. By introducing methylamine cations, the central symmetry of MBC is effectively disrupted, resulting in a non-centrosymmetric crystal structure. This structural modification remarkably enhances the piezoelectric performance, enabling more robust charge separation effects under ultrasound excitation and thus facilitating the efficient generation of reactive oxygen species (ROS). Moreover, the generated ROS triggers immunogenic cell death in tumor cells, through the depletion of excessive glutathione and the inhibition of glutathione peroxidase 4, induces ferroptosis. The combined therapeutic strategy substantially enhances the anti-tumor efficacy and effectively suppresses lung metastasis. This research offers a promising example of the application of perovskite piezoelectric materials in sonodynamic immunotherapy.
Collapse
Affiliation(s)
- Luna Zhu
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qian Wang
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jun Du
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xueyu Li
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Qingxuan Meng
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Jiacheng Lu
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yuqing Miao
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yuhao Li
- School of Materials and Chemistry, Institute of Bismuth Science, Shanghai Collaborative Innovation Center of Energy Therapy for Tumors, University of Shanghai for Science and Technology, Shanghai 200093, China.
| |
Collapse
|
5
|
Li Z, Lu Y, Wang L, Shi L, Wang T. Reactive oxygen species-dependent nanomedicine therapeutic modalities for gastric cancer. NANOSCALE ADVANCES 2025; 7:3210-3227. [PMID: 40308560 PMCID: PMC12038724 DOI: 10.1039/d5na00321k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025]
Abstract
Reactive oxygen species (ROS) play a double-edged role in gastric cancer (GC). Higher levels of ROS in tumor cells compared to normal cells facilitate tumor progression. Once ROS concentrations rise rapidly to toxic levels, they cause GC cell death, which is instead beneficial for GC treatment. Based on these functions, nano-delivery systems taking the therapeutic advantages of ROS have been widely employed in tumor therapy in recent years, overcoming the drawbacks of conventional drug delivery techniques, such as non-specific systemic effects. In this review, the precise impacts of ROS on GC have been detailed, along with ROS-based nanomedicine therapeutic schemes. These strategies mainly focused on the use of excess ROS in the tumor microenvironment for controlled drug release and a substantial enhancement of ROS concentrations for tumor killing. The challenges and opportunities for the advancement of these anticancer therapies are also emphasized.
Collapse
Affiliation(s)
- Zhiyan Li
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Yanjun Lu
- Division of Gastric Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Lulu Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| | - Liuyi Shi
- Yangzhou University Medical College Yangzhou 225001 China
| | - Tao Wang
- Department of Thoracic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School Nanjing 210008 China
| |
Collapse
|
6
|
Fang G, Tian Y, You L, Xu R, Gao S. KLF15 prevents ferroptosis in vascular smooth muscle cells via interacting with p53. Biochem Biophys Res Commun 2025; 770:152029. [PMID: 40382847 DOI: 10.1016/j.bbrc.2025.152029] [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: 01/18/2025] [Revised: 05/06/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
The formation of intracranial aneurysm (IA) is intimately linked to the progressive loss of vascular smooth muscle cells (VSMCs). Reactive oxygen species (ROS) play a pivotal role in inducing VSMC death during IA progression. Krüppel-like factor 15 (KLF15) plays a crucial role in preserving vascular homeostasis. However, the potential impact of KLF15 on ROS-triggered VSMC death remains unexplored. Analysis of microarray datasets from the GEO database suggests reduced KLF15 levels in human IA tissues. This study further confirms decreased KLF15 expression in ROS-treated human brain VSMCs (HBVSMCs). An unbiased examination of the transcriptome in HBVSMCs transfected with siKLF15 reveals that KLF15 regulates the ferroptosis pathway upon ROS stress. Silencing KLF15 results in the upregulation of genes promoting ferroptosis, such as SAT1, HMOX1, and MAP1LC3B, while downregulation of the ferroptosis regulatory gene SLC7A11. Cell death increases in KLF15-silenced HBVSMCs and is rescued by the ferroptosis inhibitor frerrostain-1. Co-immunoprecipitation and in situ proximity ligation assay indicate that KLF15 interacts with p53. Knockdown of p53 rescues the effects of siKLF15 on ROS-induced ferroptosis, including elevated cell death, lipid ROS levels, and the malondialdehyde content, as well as reduced SLC7A11protin levels in HBVSMCs. These findings suggest that KLF15 may lower cell sensitivity to ferroptosis by interacting with p53 and preventing p53-mediated transcriptional repression of SLC7A11. Overall, our results reveal a protective function of KLF15 in preventing ROS-induced ferroptosis in HBVSMCs.
Collapse
Affiliation(s)
- Guangming Fang
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Yexuan Tian
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China
| | - Lili You
- The Second Hospital of Chaoyang, Chaoyang, 122000, Liaoning Province, China
| | - Ruixue Xu
- The First Affiliated Hospital of Dalian Medical University, Dalian, 116011, China.
| | - Shijuan Gao
- Beijing Anzhen Hospital, Capital Medical University, Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing, 100029, China.
| |
Collapse
|
7
|
Wang H, Ortiz PA, Romero CA. Luminal flow in the connecting tubule induces afferent arteriole vasodilation. Clin Exp Nephrol 2025; 29:541-547. [PMID: 39800794 DOI: 10.1007/s10157-024-02615-2] [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: 09/25/2024] [Accepted: 12/19/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND Renal autoregulatory mechanisms modulate renal blood flow. Connecting tubule glomerular feedback (CNTGF) is a vasodilator mechanism in the connecting tubule (CNT), triggered paracrinally when high sodium levels are detected via the epithelial sodium channel (ENaC). The primary activation factor of CNTGF-whether NaCl concentration, independent luminal flow, or the combined total sodium delivery-is still unclear. We hypothesized that increasing luminal flow in the CNT induces CNTGF via O2- generation and ENaC activation. METHODS Rabbit afferent arterioles (Af-Arts) with adjacent CNTs were microperfused ex-vivo with variable flow rates and sodium concentrations ranging from < 1 to 80 mM and from 5 to 40 nL/min flow rates. RESULTS Perfusion of the CNT with 5 mM NaCl and increasing flow rates from 5 to 10, 20, and 40 nL/min caused a flow-rate-dependent dilation of the Af-Art (P < 0.001). Adding the ENaC blocker benzamil inhibited flow-induced Af-Art dilation, indicating a CNTGF response. In contrast, perfusion of the CNT with < 1 mM NaCl did not result in flow-induced CNTGF vasodilation (P > 0.05). Multiple linear regression modeling (R2 = 0.51; P < 0.001) demonstrated that tubular flow (β = 0.163 ± 0.04; P < 0.001) and sodium concentration (β = 0.14 ± 0.03; P < 0.001) are independent variables that induce afferent arteriole vasodilation. Tempol reduced flow-induced CNTGF, and L-NAME did not influence this effect. CONCLUSION Increased luminal flow in the CNT induces CNTGF activation via ENaC, partially due to flow-stimulated O2- production and independent of nitric oxide synthase (NOS) activity.
Collapse
Affiliation(s)
- Hong Wang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Pablo A Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, MI, USA
| | - Cesar A Romero
- Renal Medicine Division, Department of Medicine, Emory University School of Medicine, 101 Woodruff Circle, Woodruff Memorial Research Building, Office 338A, Atlanta, GA, 30322, USA.
| |
Collapse
|
8
|
Puel EM, Taruhn LF, Damé-Teixeira N, Stefani CM, Lataro RM. Is there a link between the abundance of nitrate-reducing bacteria and arterial hypertension? A systematic review. Nitric Oxide 2025; 157:19-33. [PMID: 40220988 DOI: 10.1016/j.niox.2025.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
CONTEXT Nitric oxide is a vasodilator molecule that acts on blood pressure (BP) control, and its production can occur through the reduction of nitrates by oral or intestinal nitrate-reducing bacteria. However, the relationship between nitrate-reducing bacteria and arterial hypertension (HTN) remains under debate. OBJECTIVE Systematically review if there is an association between the abundance of oral and intestinal nitrate-reducing bacteria and the occurrence of HTN in humans. DATABASES AND ELIGIBILITY CRITERIA MEDLINE, Scopus, Cochrane Library, EMBASE, LILACS, Web of Science, Livivo, ProQuest Dissertations, and Google Scholar were searched for eligible articles until February 10th, 2024. Studies were included if they: (1) were observational studies or clinical trials; (2) included adults (≥18 years old) with HTN (systolic BP ≥ 130 mmHg and/or diastolic BP > 80 mmHg and/or use of BP lowering medication); (3) compared (or not) to no-HTN adults; and (4) used next-generation sequencing microbiome analysis to identify bacterial taxa in the oral and/or gut nitrate-reducing bacteria. RESULTS The search identified 9365 articles, and 28 were included in the study after applying the inclusion and exclusion criteria; 23 articles assessed the gut microbiota, 4 assessed the oral microbiota, and 1 assessed both. Depletion of nitrate-reducing bacteria was not consistently shown in the studies. The included studies reported reduction, increase, and no change in the nitrate-reducing bacteria genera or species in oral or gut microbiota. CONCLUSION We found no association between the abundance of oral and gut nitrate-reducing bacteria and the occurrence of HTN in humans. REGISTRATION PROSPERO identification number CRD42022315891.
Collapse
Affiliation(s)
- Esthela M Puel
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Lillian F Taruhn
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil
| | - Nailê Damé-Teixeira
- Department of Dentistry, School of Health Sciences, University of Brasília, Brasília, Brazil
| | - Cristine M Stefani
- Department of Dentistry, School of Health Sciences, University of Brasília, Brasília, Brazil
| | - Renata M Lataro
- Department of Physiological Sciences, Center of Biological Sciences, Federal University of Santa Catarina, Florianópolis, Santa Catarina, Brazil.
| |
Collapse
|
9
|
Shi J, Yu Y, Yuan H, Li Y, Xue Y. Mitochondrial dysfunction in AMI: mechanisms and therapeutic perspectives. J Transl Med 2025; 23:418. [PMID: 40211347 PMCID: PMC11987341 DOI: 10.1186/s12967-025-06406-5] [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: 01/03/2025] [Accepted: 03/20/2025] [Indexed: 04/13/2025] Open
Abstract
Acute myocardial infarction (AMI) and the myocardial ischemia-reperfusion injury (MI/RI) that typically ensues represent a significant global health burden, accounting for a considerable number of deaths and disabilities. In the context of AMI, percutaneous coronary intervention (PCI) is the preferred treatment option for reducing acute ischemic damage to the heart. Despite the modernity of PCI therapy, pathological damage to cardiomyocytes due to MI/RI remains an important target for intervention that affects the long-term prognosis of patients. In recent years, mitochondrial dysfunction during AMI has been increasingly recognized as a critical factor in cardiomyocyte death. Damaged mitochondria play an active role in the formation of an inflammatory environment by triggering key signaling pathways, including those mediated by cyclic GMP-AMP synthase, NOD-like receptors and Toll-like receptors. This review emphasizes the dual role of mitochondria as both contributors to and regulators of inflammation. The aim is to explore the complex mechanisms of mitochondrial dysfunction in AMI and its profound impact on immune dysregulation. Specific interventions including mitochondrial-targeted antioxidants, membrane-stabilizing peptides, and mitochondrial transplantation therapies have demonstrated efficacy in preclinical AMI models.
Collapse
Affiliation(s)
- Jingle Shi
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yiding Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huajing Yuan
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yan Li
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| | - Yitao Xue
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China.
| |
Collapse
|
10
|
Song G, Yang Z, Cheng J, Lin H, Huang Y, Lv F, Bai H, Wang S. Chemiluminescence Resonance Energy Transfer for Targeted Photoactivation of Ion Channels in Living Cells. Angew Chem Int Ed Engl 2025; 64:e202423792. [PMID: 39888216 DOI: 10.1002/anie.202423792] [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: 12/06/2024] [Revised: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 02/01/2025]
Abstract
The regulation of oxidative stress in living cells is essential for maintaining cellular processes and signal transduction. However, developing straightforward strategies to activate oxidative stress-sensitive membrane channels in situ poses significant challenges. In this study, we present a chemiluminescence resonance energy transfer (CRET) system based on a conjugated oligomer, oligo(p-phenylenevinylene)-imidazolium (OPV-Im), designed for the activation of transient receptor potential melastatin 2 (TRPM2) calcium channels in situ by superoxide anion (O2⋅-) without requiring external light sources. The OPV-Im oligomer targeted the cell membrane efficiently, leading to the activation of TRPM2 channels in situ by the CRET process and subsequent intracellular calcium overload. This cascade resulted in mitochondrial damage and inhibition of autophagy, ultimately inducing cell apoptosis. Additionally, this strategy could be applied for the selective killing of tumor cells that overexpress TRPM2 ion channels and for inhibiting the growth of three-dimensional (3D) tumor spheroids. Our system offers a novel approach for regulating ion channel activity and oxidative stress in living cells compared to optogenetics and photodynamic therapy.
Collapse
Affiliation(s)
- Gang Song
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Zhiwen Yang
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Junjie Cheng
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Hongrui Lin
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| | - Yiming Huang
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Fengting Lv
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Haotian Bai
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Shu Wang
- Key Laboratory of Organic Solids, Beijing National Laboratory for Molecular Sciences, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
- College of Chemistry, University of Chinese Academy of Sciences., Beijing, 100049, P. R. China
| |
Collapse
|
11
|
Chen Y, Wu S, Liu H, Zhong Z, Bucci T, Wang Y, Zhao M, Liu Y, Yang Z, Gue Y, McDowell G, Huang B, Lip GYH. Role of oxidative balance score in staging and mortality risk of cardiovascular-kidney-metabolic syndrome: Insights from traditional and machine learning approaches. Redox Biol 2025; 81:103588. [PMID: 40073760 PMCID: PMC11950999 DOI: 10.1016/j.redox.2025.103588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 03/14/2025] Open
Abstract
OBJECTIVES To evaluate the roles of oxidative balance score (OBS) in staging and mortality risk of cardiovascular-kidney-metabolic syndrome (CKM). METHODS Data of this study were from the National Health and Nutrition Examination Survey 1999-2018. We performed cross-sectional analyses using multinomial logistic regression to investigate the relationship between OBS and CKM staging. Cox proportional hazards models were used to assess the impact of OBS on mortality outcomes in CKM patients. Additionally, mediation analyses were performed to explore whether OBS mediated the relationships between specific predictors (Life's Simple 7 score [LS7], systemic immune-inflammation index [SII], frailty score) and mortality outcomes. Then, machine learning models were developed to classify CKM stages 3/4 and predict all-cause mortality, with SHapley Additive exPlanations values used to interpret the contribution of OBS components. RESULTS 21,609 participants were included (20,319 CKM, median [IQR] age: 52.0 [38.0-65.0] years, 54.3% male, median [IQR] follow-up: 9.4 [5.3-14.1] years). Lower OBS quartiles were associated with advanced CKM staging. Moreover, lower OBS quartiles were related to increased mortality risk, compared to Q4 of OBS (all-cause mortality: Q1: HR 1.31, 95% CI 1.18-1.46, Q2: HR 1.27, 95% CI 1.14-1.42, Q3: HR 1.18, 95% CI 1.06-1.32; cardiovascular mortality: Q1: HR 1.44, 95% CI 1.16-1.79, Q2: HR 1.39, 95% CI 1.11-1.74, Q3: HR 1.26, 95% CI 1.01-1.57; non-cardiovascular mortality, Q1: HR 1.27, 95% CI 1.12-1.44, Q2: HR 1.23, 95% CI 1.08-1.40, Q3: HR 1.16, 95% CI 1.02-1.31), with optimal risk stratification threshold for OBS was 22. Additionally, OBS mediated (ranging 4.25%-32.85 %) effects of SII, LS7, frailty scores on mortality outcomes. Moreover, light gradient boosting machine achieved the highest performance for predicting advanced CKM staging (area under curve: 0.905) and all-cause mortality (area under curve: 0.875). Cotinine increased risk, while magnesium, vitamin B6, physical activity were protective. CONCLUSIONS This study highlights OBS as a risk stratification tool for CKM, emphasizing oxidative stress's role in CKM staging and mortality risk management.
Collapse
Affiliation(s)
- Yang Chen
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Department of Cardiovascular and Metabolic Medicine, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, United Kingdom.
| | - Shuang Wu
- National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Hongyu Liu
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Ziyi Zhong
- Department of Musculoskeletal Ageing and Science, Institute of Life Course and Medical Sciences, University of Liverpool, United Kingdom
| | - Tommaso Bucci
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Department of Clinical Internal, Anaesthesiologic and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Yimeng Wang
- National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China; National Clinical Research Center of Cardiovascular Diseases, National Center for Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Manlin Zhao
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, Engineering Research Center of Medical Devices for Cardiovascular Diseases, Ministry of Education, National Clinical Research Center for Cardiovascular Diseases, Beijing, People's Republic of China
| | - Yang Liu
- Department of Cardiovascular Medicine, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People's Republic of China
| | - Zhengkun Yang
- Department of Cardiology, Tianjin Medical University General Hospital, Tianjin, People's Republic of China
| | - Ying Gue
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - Garry McDowell
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool, United Kingdom
| | - Bi Huang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China.
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science at University of Liverpool, Liverpool John Moores University and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom; Danish Centre for Health Services Research, Department of Clinical Medicine, Aalborg University, Aalborg, DK-9220, Denmark; Medical University of Bialystok, Bialystok, Poland.
| |
Collapse
|
12
|
Penna C, Pagliaro P. Endothelial Dysfunction: Redox Imbalance, NLRP3 Inflammasome, and Inflammatory Responses in Cardiovascular Diseases. Antioxidants (Basel) 2025; 14:256. [PMID: 40227195 PMCID: PMC11939635 DOI: 10.3390/antiox14030256] [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: 01/26/2025] [Revised: 02/17/2025] [Accepted: 02/20/2025] [Indexed: 04/15/2025] Open
Abstract
Endothelial dysfunction (ED) is characterized by an imbalance between vasodilatory and vasoconstrictive factors, leading to impaired vascular tone, thrombosis, and inflammation. These processes are critical in the development of cardiovascular diseases (CVDs) such as atherosclerosis, hypertension and ischemia/reperfusion injury (IRI). Reduced nitric oxide (NO) production and increased oxidative stress are key contributors to ED. Aging further exacerbates ED through mitochondrial dysfunction and increased oxidative/nitrosative stress, heightening CVD risk. Antioxidant systems like superoxide-dismutase (SOD), glutathione-peroxidase (GPx), and thioredoxin/thioredoxin-reductase (Trx/TXNRD) pathways protect against oxidative stress. However, their reduced activity promotes ED, atherosclerosis, and vulnerability to IRI. Metabolic syndrome, comprising insulin resistance, obesity, and hypertension, is often accompanied by ED. Specifically, hyperglycemia worsens endothelial damage by promoting oxidative stress and inflammation. Obesity leads to chronic inflammation and changes in perivascular adipose tissue, while hypertension is associated with an increase in oxidative stress. The NLRP3 inflammasome plays a significant role in ED, being triggered by factors such as reactive oxygen and nitrogen species, ischemia, and high glucose, which contribute to inflammation, endothelial injury, and exacerbation of IRI. Treatments, such as N-acetyl-L-cysteine, SGLT2 or NLRP3 inhibitors, show promise in improving endothelial function. Yet the complexity of ED suggests that multi-targeted therapies addressing oxidative stress, inflammation, and metabolic disturbances are essential for managing CVDs associated with metabolic syndrome.
Collapse
Affiliation(s)
- Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy;
- National Institute for Cardiovascular Research (INRC), 40126 Bologna, Italy
| |
Collapse
|
13
|
Abidin BM, Rios FJ, Montezano AC, Touyz RM. Transient receptor potential melastatin 7 cation channel, magnesium and cell metabolism in vascular health and disease. Acta Physiol (Oxf) 2025; 241:e14282. [PMID: 39801180 DOI: 10.1111/apha.14282] [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: 06/16/2024] [Revised: 12/07/2024] [Accepted: 01/01/2025] [Indexed: 01/30/2025]
Abstract
Preserving the balance of metabolic processes in endothelial cells (ECs) and vascular smooth muscle cells (VSMCs), is crucial for optimal vascular function and integrity. ECs are metabolically active and depend on aerobic glycolysis to efficiently produce energy for their essential functions, which include regulating vascular tone. Impaired EC metabolism is linked to endothelial damage, increased permeability and inflammation. Metabolic alterations in VSMCs also contribute to vascular dysfunction in atherosclerosis and hypertension. Magnesium (Mg2+) is the second most abundant intracellular divalent cation and influences molecular processes that regulate vascular function, including vasodilation, vasoconstriction, and release of vasoactive substances. Mg2+ is critically involved in maintaining cellular homeostasis and metabolism since it is an essential cofactor for ATP, nucleic acids and hundreds of enzymes involved in metabolic processes. Low Mg2+ levels have been linked to endothelial dysfunction, increased vascular tone, vascular inflammation and arterial remodeling. Growing evidence indicates an important role for the transient receptor potential melastatin-subfamily member 7 (TRPM7) cation channel in the regulation of Mg2+ homeostasis in EC and VSMCs. In the vasculature, TRPM7 deficiency leads to impaired endothelial function, increased vascular contraction, phenotypic switching of VSMCs, inflammation and fibrosis, processes that characterize the vascular phenotype in hypertension. Here we provide a comprehensive overview on TRPM7/Mg2+ in the regulation of vascular function and how it influences EC and VSMC metabolism such as glucose and energy homeostasis, redox regulation, phosphoinositide signaling, and mineral metabolism. The putative role of TRPM7/Mg2+ and altered cellular metabolism in vascular dysfunction and hypertension is also discussed.
Collapse
Affiliation(s)
- Belma Melda Abidin
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Francisco J Rios
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Augusto C Montezano
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
| | - Rhian M Touyz
- Cardiovascular Health Across the Life Span, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- Department of Family Medicine, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
14
|
Picone F, Giudice V, Iside C, Venturini E, Di Pietro P, Vecchione C, Selleri C, Carrizzo A. Lymphocyte Subset Imbalance in Cardiometabolic Diseases: Are T Cells the Missing Link? Int J Mol Sci 2025; 26:868. [PMID: 39940640 PMCID: PMC11816853 DOI: 10.3390/ijms26030868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
Cardiometabolic and cardiovascular diseases (CVDs) remain the leading cause of death worldwide, with well-established risk factors such as smoking, obesity, and diabetes contributing to plaque formation and chronic inflammation. However, emerging evidence suggests that the immune system plays a more significant role in the development and progression of CVD than previously thought. Specifically, the finely tuned regulation of lymphocyte subsets governs post-injury inflammation and tissue damage resolution and orchestrates the functions and activation of endothelial cells, cardiomyocytes, and fibroblasts in CVD-associated lesions (e.g., atherosclerotic plaques). A deeper understanding of the immune system's involvement in CVD development and progression will provide new insights into disease biology and uncover novel therapeutic targets aimed at re-establishing immune homeostasis. In this review, we summarize the current state of knowledge on the distribution and involvement of lymphocyte subsets in CVD, including atherosclerosis, diabetes, hypertension, myocardial infarction, and stroke.
Collapse
Affiliation(s)
- Francesca Picone
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Valentina Giudice
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Concetta Iside
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | | | - Paola Di Pietro
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
| | - Carmine Vecchione
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| | - Carmine Selleri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Hematology and Transplant Center, University Hospital “San Giovanni di Dio e Ruggi d’Aragona”, 84131 Salerno, Italy
| | - Albino Carrizzo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (F.P.); (C.I.); (P.D.P.); (C.V.); (C.S.)
- Vascular Physiopathology Unit, IRCCS Neuromed, 86077 Pozzilli, Italy;
| |
Collapse
|
15
|
Zhang Z, Wang Y, Chen X, Wu C, Zhou J, Chen Y, Liu X, Tang X. The aging heart in focus: The advanced understanding of heart failure with preserved ejection fraction. Ageing Res Rev 2024; 101:102542. [PMID: 39396676 DOI: 10.1016/j.arr.2024.102542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 09/25/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Heart failure with preserved ejection fraction (HFpEF) accounts for 50 % of heart failure (HF) cases, making it the most common type of HF, and its prevalence continues to increase in the aging society. HFpEF is a systemic syndrome resulting from many risk factors, such as aging, metabolic syndrome, and hypertension, and its clinical features are highly heterogeneous in different populations. HFpEF syndrome involves the dysfunction of multiple organs, including the heart, lung, muscle, and vascular system. The heart shows dysfunction of various cells, including cardiomyocytes, endothelial cells, fibroblasts, adipocytes, and immune cells. The complex etiology and pathobiology limit experimental research on HFpEF in animal models, delaying a comprehensive understanding of the mechanisms and making treatment difficult. Recently, many scientists and cardiologists have attempted to improve the clinical outcomes of HFpEF. Recent advances in clinically related animal models and systemic pathology studies have improved our understanding of HFpEF, and clinical trials involving sodium-glucose cotransporter 2 inhibitors have significantly enhanced our confidence in treating HFpEF. This review provides an updated comprehensive discussion of the etiology and pathobiology, molecular and cellular mechanisms, preclinical animal models, and therapeutic trials in animals and patients to enhance our understanding of HFpEF and improve clinical outcomes.
Collapse
Affiliation(s)
- Zhewei Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China; Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Yu Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China; West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Xiangqi Chen
- Department of Pharmacy, Institute of Metabolic Diseases and Pharmacotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chuan Wu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China
| | - Jingyue Zhou
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China
| | - Yan Chen
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaojing Liu
- Department of Cardiology and Laboratory of Cardiovascular Diseases, Institute of Cardiovascular Diseases, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, National Health Commission Key Laboratory of Chronobiology, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Children's Medicine Key Laboratory of Sichuan Province, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, No.2222 Xinchuan Road, Chengdu 610041, China.
| |
Collapse
|
16
|
Behers BJ, Behers BM, Stephenson-Moe CA, Vargas IA, Meng Z, Thompson AJ, Melchor J, Wojtas CN, Rosario MA, Baker JF, Deevers AC, Mouratidis RW, Sweeney MJ. Magnesium and Potassium Supplementation for Systolic Blood Pressure Reduction in the General Normotensive Population: A Systematic Review and Subgroup Meta-Analysis for Optimal Dosage and Treatment Length. Nutrients 2024; 16:3617. [PMID: 39519450 PMCID: PMC11547496 DOI: 10.3390/nu16213617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 10/14/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND/OBJECTIVES Studies have shown that consistent reductions of 2 mm Hg in systolic blood pressure (SBP) for the general normotensive population can result in significant decreases in mortality from heart disease and stroke. The purpose of this meta-analysis was to determine the optimal dose and duration of treatment for magnesium and potassium supplementation, having previously discovered that both reduce SBP by -2.79 and -2.10 mm Hg, respectively. METHODS Placebo-controlled, randomized clinical trials examining the effects of magnesium and potassium supplementation on SBP were identified. Pairwise meta-analyses with subgroups for dosage and treatment duration were run. RESULTS Magnesium at dosages of ≤360 mg/day and durations greater than 3 months reduced SBP by -3.03 and -4.31 mm Hg, respectively. Potassium at dosages of ≤60 mmol/day and durations greater than 1 month reduced SBP by -2.34 and -2.80 mm Hg, respectively. CONCLUSIONS Both supplements demonstrated greater reductions in SBP for the general population at lower dosages and longer treatment durations. Future studies are needed to validate these findings and provide tailored recommendations. These studies could investigate varying dosages over long-term follow-up to provide robust data on optimal dosages and treatment durations, as our findings were limited due to reliance on previously published trials.
Collapse
Affiliation(s)
- Benjamin J Behers
- Department of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
- Florida State University Internal Medicine Residency at Sarasota Memorial Hospital, Sarasota, FL 34239, USA
| | - Brett M Behers
- Department of Clinical Research, University of South Florida College of Medicine, Tampa, FL 33602, USA
| | - Christoph A Stephenson-Moe
- Department of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| | - Ian A Vargas
- Florida State University Internal Medicine Residency at Sarasota Memorial Hospital, Sarasota, FL 34239, USA
| | - Zhuo Meng
- Department of Statistics, Florida State University, Tallahassee, FL 32306, USA
| | - Anthony J Thompson
- Department of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| | - Julian Melchor
- Department of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| | - Caroline N Wojtas
- Florida State University Internal Medicine Residency at Sarasota Memorial Hospital, Sarasota, FL 34239, USA
| | - Manuel A Rosario
- Florida State University Internal Medicine Residency at Sarasota Memorial Hospital, Sarasota, FL 34239, USA
| | - Joel F Baker
- Florida State University Internal Medicine Residency at Sarasota Memorial Hospital, Sarasota, FL 34239, USA
| | - Alexander C Deevers
- Department of Clinical Research, University of Florida, Gainesville, FL 32603, USA
| | - Roxann W Mouratidis
- Department of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| | - Michael J Sweeney
- Department of Clinical Sciences, Florida State University College of Medicine, Tallahassee, FL 32304, USA
| |
Collapse
|
17
|
Wang H, Ortiz PA, Romero CA. Luminal Flow in the Connecting Tubule induces Afferent Arteriole Vasodilation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612758. [PMID: 39345602 PMCID: PMC11429694 DOI: 10.1101/2024.09.12.612758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Background Renal autoregulatory mechanisms modulate renal blood flow. Connecting tubule glomerular feedback (CNTGF) is a vasodilator mechanism in the connecting tubule (CNT), triggered paracrinally when high sodium levels are detected via the epithelial sodium channel (ENaC). The primary activation factor of CNTGF-whether NaCl concentration, independent luminal flow, or the combined total sodium delivery-is still unclear. We hypothesized that increasing luminal flow in the CNT induces CNTGF via O2- generation and ENaC activation. Methods Rabbit afferent arterioles (Af-Arts) with adjacent CNTs were microperfused ex-vivo with variable flow rates and sodium concentrations ranging from <1 mM to 80 mM and from 5 to 40 nL/min flow rates. Results Perfusion of the CNT with 5 mM NaCl and increasing flow rates from 5 to 10, 20, and 40 nL/min caused a flow rate-dependent dilation of the Af-Art (p<0.001). Adding the ENaC blocker benzamil inhibited flow-induced Af-Art dilation, indicating a CNTGF response. In contrast, perfusion of the CNT with <1 mM NaCl did not result in flow-induced CNTGF vasodilation (p>0.05). Multiple linear regression modeling (R2=0.51;p<0.001) demonstrated that tubular flow (β=0.163 ± 0.04;p<0.001) and sodium concentration (β=0.14 ± 0.03;p<0.001) are independent variables that induce afferent arteriole vasodilation. Tempol reduced flow-induced CNTGF, and L-NAME did not influence this effect. Conclusion Increased luminal flow in the CNT induces CNTGF activation via ENaC, partially due to flow-stimulated O2- production and independent of nitric oxide synthase (NOS) activity.
Collapse
Affiliation(s)
- Hong Wang
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Pablo A. Ortiz
- Hypertension and Vascular Research Division, Department of Internal Medicine, Henry Ford Hospital, Detroit, Michigan
| | - Cesar A. Romero
- Renal Division, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| |
Collapse
|
18
|
Bautista-Pérez R, Franco M. Purinergic Receptor Antagonists: A Complementary Treatment for Hypertension. Pharmaceuticals (Basel) 2024; 17:1060. [PMID: 39204165 PMCID: PMC11357398 DOI: 10.3390/ph17081060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 08/01/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
The treatment of hypertension has improved in the last century; attention has been directed to restoring several altered pathophysiological mechanisms. However, regardless of the current treatments, it is difficult to control blood pressure. Uncontrolled hypertension is responsible for several cardiovascular complications, such as chronic renal failure, which is frequently observed in hypertensive patients. Therefore, new approaches that may improve the control of arterial blood pressure should be considered to prevent serious cardiovascular disorders. The contribution of purinergic receptors has been acknowledged in the pathophysiology of hypertension; this review describes the participation of these receptors in the alteration of kidney function in hypertension. Elevated interstitial ATP concentrations are essential for the activation of renal purinergic receptors; this becomes a fundamental pathway that leads to the development and maintenance of hypertension. High ATP levels modify essential mechanisms implicated in the long-term control of blood pressure, such as pressure natriuresis, the autoregulation of the glomerular filtration rate and renal blood flow, and tubuloglomerular feedback responses. Any alteration in these mechanisms decreases sodium excretion. ATP stimulates the release of vasoactive substances, causes renal function to decline, and induces tubulointerstitial damage. At the same time, a deleterious interaction involving angiotensin II and purinergic receptors leads to the deterioration of renal function.
Collapse
Affiliation(s)
- Rocio Bautista-Pérez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico;
| | - Martha Franco
- Department of Cardio-Renal Pathophysiology, Instituto Nacional de Cardiología Ignacio Chávez, México City 14080, Mexico
| |
Collapse
|