1
|
Cai D, Li C, Zhang Y, He S, Guo Y, Liao W, Liao Y, Bin J, He X. CircHipk3 serves a dual role in macrophage pyroptosis by promoting NLRP3 transcription and inhibition of autophagy to induce abdominal aortic aneurysm formation. Clin Transl Med 2024; 14:e70102. [PMID: 39601144 PMCID: PMC11599875 DOI: 10.1002/ctm2.70102] [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: 05/30/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
AIMS CircRNAs could regulate macrophage pyroptosis, which has the potential in promoting the synergistic effect of inflammation and matrix metalloproteinase (MMP) activity in abdominal aortic aneurysm (AAA). But the roles of circRNAs in modulating macrophage pyroptosis in the AAA remain unknown. This study explored the contribution to AAA of circHipk3, which was macrophage pyroptosis promoter, and the underlying mechanism. METHODS AND RESULTS CircHipk3 was markedly upregulated in aortic aneurysms compared with that in normal arteries. In mice treated with circHipk3 contributed to macrophage pyroptosis, subsequently promoting the synergistic effect of inflammation and MMP synthesis, and significantly accelerated angiotensin (Ang) II- and porcine pancreatic elastase (PPE)-induced AAA formation. Mechanically, chromatin isolation by RNA purification (ChIRP) indicated that circHipk3 facilitated macrophage pyroptosis by interaction with Stat3, increase the NLRP3 level in the aorta, and by binding Snd1 to promote Ptbp1 mRNA degradation to inhibit autophagy. Therefore, our study revealed the important role of circHipk3 in macrophage pyroptosis and thus significantly improved the outcome of AAA. CONCLUSIONS CircHipk3 serves a dual role in augmenting macrophage pyroptosis by interaction with Stat3, increase the NLRP3 level, and by binding Snd1 to promote Ptbp1 mRNA degradation to inhibit autophagy, thereby inducing aneurysm formation and progression. KEY POINTS CircHipk3 is significantly upregulated in abdominal aortic aneurysms (AAA) compared to normal arteries, contributing to macrophage pyroptosis. CircHipk3 promotes the synergistic effect of inflammation and matrix metalloproteinase (MMP) activity, accelerating Angiotensin II- and porcine pancreatic elastase-induced AAA formation in mice. Mechanistically, CircHipk3 interacts with Stat3 to elevate NLRP3 levels and binds Snd1 to promote Ptbp1 mRNA degradation, inhibiting autophagy. CircHipk3's dual role in enhancing NLRP3 inflammasome activation and inhibiting autophagy makes it a critical regulator in AAA development and rupture. Targeting CircHipk3 may offer a novel therapeutic strategy to prevent pyroptosis and AAA development, positioning it as a potential treatment target.
Collapse
Affiliation(s)
- Donghua Cai
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhouChina
| | - Chuling Li
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhouChina
| | - Yingyuan Zhang
- Department of Cardiology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Sisi He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhouChina
| | - Yihai Guo
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhouChina
| | - Wangjun Liao
- Department of Oncology, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
| | - Yulin Liao
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhouChina
| | - Jianping Bin
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhouChina
| | - Xiang He
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Nanfang HospitalSouthern Medical UniversityGuangzhouChina
- Guangdong Provincial Key Laboratory of Cardiac Function and MicrocirculationGuangzhouChina
| |
Collapse
|
2
|
Castillo RL, Farías J, Sandoval C, González-Candia A, Figueroa E, Quezada M, Cruz G, Llanos P, Jorquera G, Kostin S, Carrasco R. Role of NLRP3 Inflammasome in Heart Failure Patients Undergoing Cardiac Surgery as a Potential Determinant of Postoperative Atrial Fibrillation and Remodeling: Is SGLT2 Cotransporter Inhibition an Alternative for Cardioprotection? Antioxidants (Basel) 2024; 13:1388. [PMID: 39594530 PMCID: PMC11591087 DOI: 10.3390/antiox13111388] [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: 09/06/2024] [Revised: 10/29/2024] [Accepted: 11/11/2024] [Indexed: 11/28/2024] Open
Abstract
In heart failure (HF) patients undergoing cardiac surgery, an increased activity of mechanisms related to cardiac remodeling may determine a higher risk of postoperative atrial fibrillation (POAF). Given that atrial fibrillation (AF) has a negative impact on the course and management of HF, including the need for anticoagulation therapy, identifying the factors associated with AF occurrence after cardiac surgery is crucial for the prognosis of these patients. POAF is thought to occur when various clinical and biochemical triggers act on susceptible cardiac tissue (first hit), with oxidative stress and inflammation during cardiopulmonary bypass (CPB) surgery being potential contributing factors (second hit). However, the molecular mechanisms involved in these processes remain poorly characterized. Recent research has shown that patients who later develop POAF often have pre-existing abnormalities in calcium handling and activation of NLRP3-inflammasome signaling in their atrial cardiomyocytes. These molecular changes may make cardiomyocytes more susceptible to spontaneous Ca2+-releases and subsequent arrhythmias, particularly when exposed to inflammatory mediators. Additionally, some clinical studies have linked POAF with elevated preoperative inflammatory markers, but there is a need for further research in order to better understand the impact of CPB surgery on local and systemic inflammation. This knowledge would make it possible to determine whether patients susceptible to POAF have pre-existing inflammatory conditions or cellular electrophysiological factors that make them more prone to developing AF and cardiac remodeling. In this context, the NLRP3 inflammasome, expressed in cardiomyocytes and cardiac fibroblasts, has been identified as playing a key role in the development of HF and AF, making patients with pre-existing HF with reduced ejection fraction (HFrEF) the focus of several clinical studies with interventions that act at this level. On the other hand, HFpEF has been linked to metabolic and non-ischemic risk factors, but more research is needed to better characterize the myocardial remodeling events associated with HFpEF. Therefore, since ventricular remodeling may differ between HFrEF and HFpEF, it is necessary to perform studies in both groups of patients due to their pathophysiological variations. Clinical evidence has shown that pharmacological therapies that are effective for HFrEF may not provide the same anti-remodeling benefits in HFpEF patients, particularly compared to traditional adrenergic and renin-angiotensin-aldosterone system inhibitors. On the other hand, there is growing interest in medications with pleiotropic or antioxidant/anti-inflammatory effects, such as sodium-glucose cotransporter 2 inhibitors (SGLT-2is). These drugs may offer anti-remodeling effects in both HFrEF and HFpEF by inhibiting pro-inflammatory, pro-oxidant, and NLRP3 signaling pathways and their mediators. The anti-inflammatory, antioxidant, and anti-remodeling effects of SGLT-2 i have progressively expanded from HFrEF and HFpEF to other forms of cardiac remodeling. However, these advances in research have not yet encompassed POAF despite its associations with inflammation, oxidative stress, and remodeling. Currently, the direct or indirect effects of NLRP3-dependent pathway inhibition on the occurrence of POAF have not been clinically assessed. However, given that NLRP3 pathway inhibition may also indirectly affect other pathways, such as inhibition of NF-kappaB or inhibition of matrix synthesis, which are strongly linked to POAF and cardiac remodeling, it is reasonable to hypothesize that this type of intervention could play a role in preventing these events.
Collapse
Affiliation(s)
- Rodrigo L. Castillo
- Departamento de Medicina Interna Oriente, Facultad de Medicina, Universidad de Chile, Santiago 7500922, Chile
- Unidad de Paciente Crítico, Hospital del Salvador, Santiago 7500922, Chile
| | - Jorge Farías
- Departamento de Ingeniería Química, Facultad de Ingeniería y Ciencias, Universidad de La Frontera, Temuco 4811230, Chile
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Departamento de Medicina Interna, Facultad de Medicina, Universidad de La Frontera, Temuco 4811230, Chile
| | - Alejandro González-Candia
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile; (A.G.-C.); (E.F.)
| | - Esteban Figueroa
- Instituto de Ciencias de la Salud, Universidad de O’Higgins, Rancagua 2841959, Chile; (A.G.-C.); (E.F.)
| | - Mauricio Quezada
- Facultad de Medicina, Universidad Finis Terrae, Santiago 7501015, Chile;
| | - Gonzalo Cruz
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
| | - Paola Llanos
- Centro de Estudios en Ejercicio, Metabolismo y Cáncer, Facultad de Medicina, Universidad de Chile, Santiago 8380453, Chile;
- Facultad de Odontología, Instituto de Investigación en Ciencias Odontológicas, Universidad de Chile, Santiago 8380544, Chile
| | - Gonzalo Jorquera
- Instituto de Fisiología, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso 2360102, Chile;
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), Universidad de Chile, Santiago 8331051, Chile;
| | - Sawa Kostin
- Faculty of Health Sciences, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany;
| | - Rodrigo Carrasco
- Departamento de Cardiología, Clínica Alemana de Santiago, Santiago 7500922, Chile;
| |
Collapse
|
3
|
Yin Z, Zhang J, Shen Z, Qin J, Wan J, Wang M. Regulated vascular smooth muscle cell death in vascular diseases. Cell Prolif 2024; 57:e13688. [PMID: 38873710 PMCID: PMC11533065 DOI: 10.1111/cpr.13688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 05/13/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024] Open
Abstract
Regulated cell death (RCD) is a complex process that involves several cell types and plays a crucial role in vascular diseases. Vascular smooth muscle cells (VSMCs) are the predominant elements of the medial layer of blood vessels, and their regulated death contributes to the pathogenesis of vascular diseases. The types of regulated VSMC death include apoptosis, necroptosis, pyroptosis, ferroptosis, parthanatos, and autophagy-dependent cell death (ADCD). In this review, we summarize the current evidence of regulated VSMC death pathways in major vascular diseases, such as atherosclerosis, vascular calcification, aortic aneurysm and dissection, hypertension, pulmonary arterial hypertension, neointimal hyperplasia, and inherited vascular diseases. All forms of RCD constitute a single, coordinated cell death system in which one pathway can compensate for another during disease progression. Pharmacologically targeting RCD pathways has potential for slowing and reversing disease progression, but challenges remain. A better understanding of the role of regulated VSMC death in vascular diseases and the underlying mechanisms may lead to novel pharmacological developments and help clinicians address the residual cardiovascular risk in patients with cardiovascular diseases.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Zican Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Juan‐Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Center for Healthy Aging, Wuhan University School of NursingWuhanChina
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of GeriatricsZhongnan Hospital of Wuhan University, Wuhan UniversityWuhanChina
- Cardiovascular Research Institute, Wuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
4
|
Balahura Stămat LR, Dinescu S. Inhibition of NLRP3 inflammasome contributes to paclitaxel efficacy in triple negative breast cancer treatment. Sci Rep 2024; 14:24753. [PMID: 39433537 PMCID: PMC11494052 DOI: 10.1038/s41598-024-75805-3] [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: 08/05/2024] [Accepted: 10/08/2024] [Indexed: 10/23/2024] Open
Abstract
Chronic inflammation and NLRP3 inflammasome activation are among the determining factors of breast malignancies. Paclitaxel (PTX) is a drug used in breast cancer treatment which sustains prolonged inflammation, reducing the effectiveness of chemotherapy. Considering the impact of inflammatory processes in cancer progression, there is a strong concern to develop therapeutic strategy targeting NLRP3 inflammasome for triple-negative breast cancer (TNBC) treatment. Therefore, the aim of this study was to evaluate the potential of PTX and NLRP3 inflammasome modulation to counterbalance TNBC by inducing programmed cell death and inhibiting the activity of pro-inflammatory cytokines. The obtained results suggested the strong interaction between NLRP3 inflammasome and TNBC and revealed that pharmacological inhibition, using NLRP3-specific inhibitor MCC950, and genetic silencing of NLRP3 inflammasome using specific small interfering RNA, reduced inflammatory responses and facilitated PTX-determined tumor cell death. Thus, NLRP3 inflammasome manipulation in combination with anti-tumor drugs opens up new therapeutic perspectives for TNBC therapy.
Collapse
Affiliation(s)
- Liliana-Roxana Balahura Stămat
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, 050095, Romania.
- Research Institute of the University of Bucharest, Bucharest, 050663, Romania.
| |
Collapse
|
5
|
Wang JX, Xu XY, Wang YM, Chen AD, Li YH, Zhu GQ, Xiong XQ. Superior cervical ganglionectomy attenuates vascular remodeling in spontaneously hypertensive rats. J Hypertens 2024:00004872-990000000-00564. [PMID: 39445597 DOI: 10.1097/hjh.0000000000003883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 09/05/2024] [Indexed: 10/25/2024]
Abstract
BACKGROUND Sympathetic hyperactivity contributes to the pathogenesis of hypertension. However, it is unclear whether the excessive sympathetic activity is an independent and crucial factor for vascular remodeling in hypertension. This study focused on the effect of local sympathetic denervation with superior cervical ganglionectomy (SCGx) on vascular remodeling. METHODS Surgical bilateral SCGx was performed in 9-week-old male Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Control rats received sham-operation without SCGx. All measurements were made 4 weeks after the surgery. RESULTS The effectiveness of SCGx was confirmed by the eye features of Horner syndrome, greatly reduced tyrosine hydroxylase (TH) contents in the superior cervical ganglion (SCG)-innervated arteries in the head. Although SCGx had no significant effects on blood pressure and heart rate in WKY and SHR, it attenuated vascular remodeling of facial artery and superficial temporal artery in SHR, two representative SCG-innervated extracranial arteries, without significant effects on non-SCG-innervated thoracic aorta and mesenteric artery. SCGx-treated SHR had more auricular blood flow and retina microvasculature than sham-operated SHR. However, SCGx had only a mild effect in attenuating the vascular remodeling of basilar artery and middle cerebral artery, two representative SCG-innervated intracranial arteries, in SHR. SCGx-treated SHR exhibited upregulation of α-smooth muscle actin, downregulation of proliferating cell nuclear antigen, and attenuation of oxidative stress and inflammation in facial artery and superficial temporal artery. CONCLUSIONS Sympathetic denervation by SCGx in SHR attenuated local vascular remodeling, suggesting that sympathetic overactivity is a crucial pathogenic factor of vascular remodeling in SHR.
Collapse
Affiliation(s)
- Jing-Xiao Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology
| | - Xiao-Yu Xu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology
| | - Yi-Ming Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology
| | - Ai-Dong Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao-Qing Xiong
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology
| |
Collapse
|
6
|
Satheesan A, Kumar J, Leela KV, Murugesan R, Chaithanya V, Angelin M. Review on the role of nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3) inflammasome pathway in diabetes: mechanistic insights and therapeutic implications. Inflammopharmacology 2024; 32:2753-2779. [PMID: 39160391 DOI: 10.1007/s10787-024-01556-2] [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: 07/04/2024] [Accepted: 08/10/2024] [Indexed: 08/21/2024]
Abstract
This review explores the pivotal role of the nucleotide-binding oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome in the pathogenesis of diabetes and its complications, highlighting the therapeutic potential of various oral hypoglycemic drugs targeting this pathway. NLRP3 inflammasome activation, triggered by metabolic stressors like hyperglycemia, hyperlipidemia, and free fatty acids (FFAs), leads to the release of pro-inflammatory cytokines interleukin-1β and interleukin-18, driving insulin resistance, pancreatic β-cell dysfunction, and systemic inflammation. These processes contribute to diabetic complications such as nephropathy, neuropathy, retinopathy, and cardiovascular diseases (CVD). Here we discuss the various transcriptional, epigenetic, and gut microbiome mediated regulation of NLRP3 activation in diabetes. Different classes of oral hypoglycemic drugs modulate NLRP3 inflammasome activity through various mechanisms: sulfonylureas inhibit NLRP3 activation and reduce inflammatory cytokine levels; sodium-glucose co-transporter 2 inhibitors (SGLT2i) suppress inflammasome activity by reducing oxidative stress and modulating intracellular signaling pathways; dipeptidyl peptidase-4 inhibitors mitigate inflammasome activation, protecting against renal and vascular complications; glucagon-like peptide-1 receptor agonists attenuate NLRP3 activity, reducing inflammation and improving metabolic outcomes; alpha-glucosidase inhibitors and thiazolidinediones exhibit anti-inflammatory properties by directly inhibiting NLRP3 activation. Agents that specifically target NLRP3 and inhibit their activation have been identified recently such as MCC950, Anakinra, CY-09, and many more. Targeting the NLRP3 inflammasome, thus, presents a promising strategy for managing diabetes and its complications, with oral hypoglycemic drugs offering dual benefits of glycemic control and inflammation reduction. Further research into the specific mechanisms and long-term effects of these drugs on NLRP3 inflammasome activity is warranted.
Collapse
Affiliation(s)
- Abhishek Satheesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Janardanan Kumar
- Department of General Medicine, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India.
| | - Kakithakara Vajravelu Leela
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Ria Murugesan
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Venkata Chaithanya
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| | - Matcha Angelin
- Department of Microbiology, SRM Medical College Hospital and Research Centre, SRMIST, Kattankulathur, Chengalpattu, Tamil Nadu, 603203, India
| |
Collapse
|
7
|
Liu X, Chen S, Luo W, Yu C, Yan S, Lei L, Qiu S, Lin X, Feng T, Shi J, Zhang Q, Liang H, Liu X, Lee APW, Zheng L, Zhang X, Xiu J. LncRNA MFRL regulates the phenotypic switch of vascular smooth muscle cells to attenuate arterial remodeling by encoding a novel micropeptide MFRLP. Transl Res 2024; 272:54-67. [PMID: 38838852 DOI: 10.1016/j.trsl.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Revised: 05/14/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Arterial remodeling is a common pathophysiological change in the pathogenesis of cardiovascular diseases in which the phenotypic switch of vascular smooth muscle cells (VSMC) plays an important role. Recently, an increasing number of long non-coding RNAs(lncRNAs) have been shown to encode micropeptides that play biological roles and have great clinical transformation potential. However, the role of micropeptides encoded by lncRNAs in arterial remodeling has not been well studied and requires further exploration. METHODS AND RESULTS Through bioinformatic analysis and experimental verification, we found that a new lncRNA, the mitochondrial function-related lncRNA (MFRL), encodes a 64-amino acid micropeptide, MFRLP. MFRL and MFRLP play important roles in the phenotypic switch of VSMC. Further experiments showed that MFRLP interacts with mitochondrial cytochrome b to reduce accumulation of reactive oxygen species, suppress mitophagy and inhibit the VSMC switch from contractile to synthetic phenotype. CONCLUSIONS LncRNA MFRL encodes the micropeptide MFRLP, which interacts with mitochondrial cytochrome b to inhibit the VSMC switch from contractile to synthetic phenotype and improve arterial remodeling.
Collapse
Affiliation(s)
- Xiaocong Liu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Siyu Chen
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Wei Luo
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Chen Yu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Shaohua Yan
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Li Lei
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Shifeng Qiu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xinxin Lin
- Department of Critical Care Medicine, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350000, PR China
| | - Ting Feng
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Jinglin Shi
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Qiuxia Zhang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Hongbin Liang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xuewei Liu
- Affiliated Dongguan Hospital, Southern Medical University, Dongguan 523059, PR China
| | - Alex Pui-Wai Lee
- Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong 999077, PR China
| | - Lei Zheng
- Department of Laboratory Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China
| | - Xinlu Zhang
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| | - Jiancheng Xiu
- Department of Cardiology, State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Nanfang Hospital, Southern Medical University, Guangzhou 510515, PR China.
| |
Collapse
|
8
|
Tiwari P, Elgazzaz M, Lazartigues E, Hanif K. Effect of Diminazene Aceturate, an ACE2 activator, on platelet CD40L signaling induced glial activation in rat model of hypertension. Int Immunopharmacol 2024; 139:112654. [PMID: 38996777 DOI: 10.1016/j.intimp.2024.112654] [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: 10/24/2023] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.
Collapse
Affiliation(s)
- Priya Tiwari
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mona Elgazzaz
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Eric Lazartigues
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
9
|
Hsu CY, Mustafa MA, Moath Omar T, Taher SG, Ubaid M, Gilmanova NS, Nasrat Abdulraheem M, Saadh MJ, Athab AH, Mirzaei R, Karampoor S. Gut instinct: harnessing the power of probiotics to tame pathogenic signaling pathways in ulcerative colitis. Front Med (Lausanne) 2024; 11:1396789. [PMID: 39323474 PMCID: PMC11422783 DOI: 10.3389/fmed.2024.1396789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 08/22/2024] [Indexed: 09/27/2024] Open
Abstract
Ulcerative colitis (UC) is a chronic inflammatory bowel disease (IBD) marked by persistent inflammation of the mucosal lining of the large intestine, leading to debilitating symptoms and reduced quality of life. Emerging evidence suggests that an imbalance of the gut microbiota plays a crucial role in UC pathogenesis, and various signaling pathways are implicated in the dysregulated immune response. Probiotics are live microorganisms that confer health benefits to the host, have attracted significant attention for their potential to restore gut microbial balance and ameliorate inflammation in UC. Recent studies have elucidated the mechanisms by which probiotics modulate these signaling pathways, often by producing anti-inflammatory molecules and promoting regulatory immune cell function. For example, probiotics can inhibit the nuclear factor-κB (NF-κB) pathway by stabilizing Inhibitor of kappa B alpha (IκBα), dampening the production of proinflammatory cytokines. Similarly, probiotics can modulate the Janus kinase/signal transducer and activator of transcription (JAK/STAT) signaling pathway, suppressing the activation of STAT1 and STAT3 and thus reducing the inflammatory response. A better understanding of the underlying mechanisms of probiotics in modulating pathogenic signaling pathways in UC will pave the way for developing more effective probiotic-based therapies. In this review, we explore the mechanistic role of probiotics in the attenuation of pathogenic signaling pathways, including NF-κB, JAK/STAT, mitogen-activated protein kinases (MAPKs), Wnt/β-catenin, the nucleotide-binding domain (NOD)-, leucine-rich repeat (LRR)- and pyrin domain-containing protein 3 (NLRP3) inflammasome, Toll-like receptors (TLRs), interleukin-23 (IL-23)/IL-17 signaling pathway in UC.
Collapse
Affiliation(s)
- Chou-Yi Hsu
- Department of Pharmacy, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
- Thunderbird School of Global Management, Arizona State University Tempe Campus, Phoenix, AZ, United States
| | - Mohammed Ahmed Mustafa
- Department of Medical Laboratory Technology, Imam Jaafar AL-Sadiq University, Baghdad, Iraq
- Department of Pathological Analyzes, College of Applied Sciences, University of Samarra, Samarra, Iraq
| | - Thabit Moath Omar
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq
| | - Sada Gh Taher
- Department of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Mohammed Ubaid
- Department of MTL, Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Nataliya S Gilmanova
- Department of Prosthetic Dentistry, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | | | | | - Aya H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Rasoul Mirzaei
- Venom and Biotherapeutics Molecules Lab, Medical Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Sajad Karampoor
- Gastrointestinal and Liver Diseases Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Verlinden SF. The genetic advantage of healthy centenarians: unraveling the central role of NLRP3 in exceptional healthspan. FRONTIERS IN AGING 2024; 5:1452453. [PMID: 39301197 PMCID: PMC11410711 DOI: 10.3389/fragi.2024.1452453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Despite extensive research into extending human healthspan (HS) and compressing morbidity, the mechanisms underlying aging remain elusive. However, a better understanding of the genetic advantages responsible for the exceptional HS of healthy centenarians (HC), who live in good physical and mental health for one hundred or more years, could lead to innovative health-extending strategies. This review explores the role of NLRP3, a critical component of innate immunity that significantly impacts aging. It is activated by pathogen-associated signals and self-derived signals that increase with age, leading to low-grade inflammation implicated in age-related diseases. Furthermore, NLRP3 functions upstream in several molecular aging pathways, regulates cellular senescence, and may underlie the robust health observed in HC. By targeting NLRP3, mice exhibit a phenotype akin to that of HC, the HS of monkeys is extended, and aging symptoms are reversed in humans. Thus, targeting NLRP3 could offer a promising approach to extend HS. Additionally, a paradigm shift is proposed. Given that the HS of the broader population is 30 years shorter than that of HC, it is postulated that they suffer from a form of accelerated aging. The term 'auto-aging' is suggested to describe accelerated aging driven by NLRP3.
Collapse
|
11
|
Li Y, Yishajiang S, Chen Y, Tulahong G, Wen W, Wang M, Li Z. TRPC5-mediated NLRP3 inflammasome activation contributes to myocardial cell pyroptosis in chronic intermittent hypoxia rats. Acta Cardiol 2024; 79:796-804. [PMID: 39377158 DOI: 10.1080/00015385.2024.2408137] [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: 11/13/2023] [Revised: 05/29/2024] [Accepted: 09/17/2024] [Indexed: 10/09/2024]
Abstract
BACKGROUND Chronic intermittent hypoxia (CIH) is the primary cause of myocardial inflammation in obstructive sleep apnea-hypopnea syndrome (OSAHS). Pyroptosis is a newly discovered form of programmed cell death accompanying inflammatory reactions. Our previous study showed that TRPC5 is upregulated in the myocardial injury of CIH rats. The present study aimed to explore the role of TRPC5 in CIH-induced myocardial cell pyroptosis. METHODS A model of CIH in OSA rats was established. SD rats were randomly divided into control group(8rats) and OSA group(8rats). Scanning electron microscope(SEM) was performed on left ventricular tissues slides. Western blot were used to detect the expression levels of pyroptosis-related factors and TRPC5 and its downstream proteins in myocardia tissue. RESULTS The pyroptosis of myocardial cells by SEM revealed damaged cell membrane integrity of OSA group rats, with fibrous tissue attached to the cell membrane surface, and vesicular protrusions and pyroptotic bodies were observed. Compared to the control group, the expression of pyroptosis-related proteins, such as caspase1, pro-IL-1β, IL-1β, IL-18, GSDMD, and GSDMD-N was upregulated in the OSA group (p < 0.05). Compared to the control group, the expression of TRPC5, NLPR3, p-CaMKIIβ + δ+γ, and HDAC4 was higher in the OSA group (p < 0.05). CONCLUSIONS These findings indicated that the pyroptosis response increases in CIH-induced myocardial injury, and the mechanism that TRPC5 is upregulated, promoting the expression of NLRP3 and inflammasome formation through CaMKII phosphorylation and HDAC4 cytoplasmic translocation. This might be a potential target for the treatment of OSA-induced myocardial injury.
Collapse
Affiliation(s)
- Yu Li
- Second Department of General Internal Medicine, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Sharezhati Yishajiang
- Department of Hypertension, The People's Hospital of Xinjiang Uygur Autonomous Region, Urumqi, Xinjiang, China
| | - Yulan Chen
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Gulinazi Tulahong
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Wen Wen
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Mengmeng Wang
- Department of Hypertension, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Zhiqiang Li
- Animal Experiments Center, Xinjiang Medical University, Urumqi, Xinjiang, China
| |
Collapse
|
12
|
Zheng F, Ye C, Lei JZ, Ge R, Li N, Bo JH, Chen AD, Zhang F, Zhou H, Wang JJ, Chen Q, Li YH, Zhu GQ, Han Y. Intervention of Asprosin Attenuates Oxidative Stress and Neointima Formation in Vascular Injury. Antioxid Redox Signal 2024; 41:488-504. [PMID: 38814824 DOI: 10.1089/ars.2023.0383] [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] [Indexed: 06/01/2024]
Abstract
Aims: Asprosin, a newly discovered hormone, is linked to insulin resistance. This study shows the roles of asprosin in vascular smooth muscle cell (VSMC) proliferation, migration, oxidative stress, and neointima formation of vascular injury. Methods: Mouse aortic VSMCs were cultured, and platelet-derived growth factor-BB (PDGF-BB) was used to induce oxidative stress, proliferation, and migration in VSMCs. Vascular injury was induced by repeatedly moving a guidewire in the lumen of the carotid artery in mice. Results: Asprosin overexpression promoted VSMC oxidative stress, proliferation, and migration, which were attenuated by toll-like receptor 4 (TLR4) knockdown, antioxidant (N-Acetylcysteine, NAC), NADPH oxidase 1 (NOX1) inhibitor ML171, or NOX2 inhibitor GSK2795039. Asprosin overexpression increased NOX1/2 expressions, whereas asprosin knockdown increased heme oxygenase-1 (HO-1) and NADPH quinone oxidoreductase-1 (NQO-1) expressions. Asprosin inhibited nuclear factor E2-related factor 2 (Nrf2) nuclear translocation. Nrf2 activator sulforaphane increased HO-1 and NQO-1 expressions and prevented asprosin-induced NOX1/2 upregulation, oxidative stress, proliferation, and migration. Exogenous asprosin protein had similar roles to asprosin overexpression. PDGF-BB increased asprosin expressions. PDGF-BB-induced oxidative stress, proliferation, and migration were enhanced by Nrf2 inhibitor ML385 but attenuated by asprosin knockdown. Vascular injury increased asprosin expression. Local asprosin knockdown in the injured carotid artery promoted HO-1 and NQO-1 expressions but attenuated the NOX1 and NOX2 upregulation, oxidative stress, neointima formation, and vascular remodeling in mice. Innovation and Conclusion: Asprosin promotes oxidative stress, proliferation, and migration of VSMCs via TLR4-Nrf2-mediated redox imbalance. Inhibition of asprosin expression attenuates VSMC proliferation and migration, oxidative stress, and neointima formation in the injured artery. Asprosin might be a promising therapeutic target for vascular injury. Antioxid. Redox Signal. 41, 488-504.
Collapse
Affiliation(s)
- Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Chao Ye
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jian-Zhen Lei
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Rui Ge
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Na Li
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jin-Hua Bo
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ai-Dong Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Feng Zhang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Hong Zhou
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Jue-Jin Wang
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, People's Republic China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, People's Republic China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| | - Ying Han
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, and Department of Physiology, Nanjing Medical University, Nanjing, People's Republic of China
| |
Collapse
|
13
|
Carrillo-Gálvez AB, Zurita F, Guerra-Valverde JA, Aguilar-González A, Abril-García D, Padial-Molina M, Olaechea A, Martín-Morales N, Martín F, O’Valle F, Galindo-Moreno P. NLRP3 and AIM2 inflammasomes expression is modified by LPS and titanium ions increasing the release of active IL-1β in alveolar bone-derived MSCs. Stem Cells Transl Med 2024; 13:826-841. [PMID: 39013640 PMCID: PMC11328940 DOI: 10.1093/stcltm/szae042] [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: 12/30/2023] [Accepted: 05/19/2024] [Indexed: 07/18/2024] Open
Abstract
Periodontitis and peri-implantitis are inflammatory diseases of infectious etiology that lead to the destruction of the supporting tissues located around teeth or implants. Although both pathologies share several characteristics, it is also known that they show important differences which could be due to the release of particles and metal ions from the implant surface. The activation of the inflammasome pathway is one of the main triggers of the inflammatory process. The inflammatory process in patients who suffer periodontitis or peri-implantitis has been mainly studied on cells of the immune system; however, it is also important to consider other cell types with high relevance in the regulation of the inflammatory response. In that context, mesenchymal stromal cells (MSCs) play an essential role in the regulation of inflammation due to their ability to modulate the immune response. This study shows that the induction of NLRP3 and absent in melanoma 2 (AIM2) inflammasome pathways mediated by bacterial components increases the secretion of active IL-1β and the pyroptotic process on human alveolar bone-derived mesenchymal stromal cells (hABSCs). Interestingly, when bacterial components are combined with titanium ions, NLRP3 expression is further increased while AIM2 expression is reduced. Furthermore, decrease of NLRP3 or AIM2 expression in hABSCs partially reverses the negative effect observed on the progression of the inflammatory process as well as on cell survival. In summary, our data suggest that the progression of the inflammatory process in peri-implantitis could be more acute due to the combined action of organic and inorganic components.
Collapse
Affiliation(s)
- Ana Belén Carrillo-Gálvez
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| | - Federico Zurita
- Department of Genetics and Institute of Biotechnology, University of Granada, 18071 Granada, Spain
| | - José Antonio Guerra-Valverde
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- PhD Program in Clinical Medicine and Public Health, University of Granada,18071 Granada, Spain
| | - Araceli Aguilar-González
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada, 18016 Granada, Spain
- Department of Medicinal and Organic Chemistry and Excellence Research Unit of Chemistry Applied to Bio-Medicine and the Environment, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain
| | - Darío Abril-García
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| | - Miguel Padial-Molina
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| | - Allinson Olaechea
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- PhD Program in Clinical Medicine and Public Health, University of Granada,18071 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada, 18016 Granada, Spain
| | - Natividad Martín-Morales
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- PhD Program in Biomedicine, University of Granada, 18071 Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, 18071 Granada, Spain
| | - Francisco Martín
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government PTS Granada, 18016 Granada, Spain
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18071 Granada, Spain
- Excellence Research Unit “Modeling Nature” (MNat), University of Granada, 18071 Granada, Spain
| | - Francisco O’Valle
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
- Department of Pathology, School of Medicine, University of Granada, 18071 Granada, Spain
- Institute of Biopathology and Regenerative Medicine (IBIMER, CIBM), University of Granada, 18071 Granada, Spain
| | - Pablo Galindo-Moreno
- Department of Oral Surgery and Implant Dentistry, School of Dentistry, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria (IBS) de Granada, 18012 Granada, Spain
| |
Collapse
|
14
|
He R, He Z, Zhang T, Liu B, Gao M, Li N, Geng Q. HDAC3 in action: Expanding roles in inflammation and inflammatory diseases. Cell Prolif 2024:e13731. [PMID: 39143689 DOI: 10.1111/cpr.13731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/14/2024] [Accepted: 07/27/2024] [Indexed: 08/16/2024] Open
Abstract
Inflammation serves as the foundation for numerous physiological and pathological processes, driving the onset and progression of various diseases. Histone deacetylase 3 (HDAC3), an essential chromatin-modifying protein within the histone deacetylase superfamily, exerts its transcriptional inhibitory role through enzymatic histone modification to uphold normal physiological function, growth, and development of the body. With both enzymatic and non-enzymatic activities, HDAC3 plays a pivotal role in regulating diverse transcription factors associated with inflammatory responses and related diseases. This review examines the involvement of HDAC3 in inflammatory responses while exploring its therapeutic potential as a target for treating inflammatory diseases, thereby offering valuable insights for clinical applications.
Collapse
Affiliation(s)
- Ruyuan He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhuokun He
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Tianyu Zhang
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Bohao Liu
- Department of Thoracic Surgery, Jilin University, Changchun, China
| | - Minglang Gao
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ning Li
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Geng
- Department of Thoracic Surgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
15
|
Zhang J, Liu S, Ding W, Wan J, Qin JJ, Wang M. Resolution of inflammation, an active process to restore the immune microenvironment balance: A novel drug target for treating arterial hypertension. Ageing Res Rev 2024; 99:102352. [PMID: 38857706 DOI: 10.1016/j.arr.2024.102352] [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: 11/24/2023] [Revised: 05/11/2024] [Accepted: 05/27/2024] [Indexed: 06/12/2024]
Abstract
The resolution of inflammation, the other side of the inflammatory response, is defined as an active and highly coordinated process that promotes the restoration of immune microenvironment balance and tissue repair. Inflammation resolution involves several key processes, including dampening proinflammatory signaling, specialized proresolving lipid mediator (SPM) production, nonlipid proresolving mediator production, efferocytosis and regulatory T-cell (Treg) induction. In recent years, increasing attention has been given to the effects of inflammation resolution on hypertension. Furthermore, our previous studies reported the antihypertensive effects of SPMs. Therefore, in this review, we aim to summarize and discuss the detailed association between arterial hypertension and inflammation resolution. Additional, the association between gut microbe-mediated immune and hypertension is discussed. This findings suggested that accelerating the resolution of inflammation can have beneficial effects on hypertension and its related organ damage. Exploring novel drug targets by focusing on various pathways involved in accelerating inflammation resolution will contribute to the treatment and control of hypertensive diseases in the future.
Collapse
Affiliation(s)
- Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Siqi Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China
| | - Wen Ding
- Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China; Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| | - Juan-Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Center for Healthy Aging, Wuhan University School of Nursing, Wuhan, China.
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan University, Wuhan University, Wuhan, China; Department of Cardiology, Renmin Hospital of Wuhan University; Cardiovascular Research Institute, Wuhan University; Hubei Key Laboratory of Cardiology, Wuhan, China.
| |
Collapse
|
16
|
Xu ZQ, Li XZ, Zhu R, Ge R, Wei H, Shi HW, Wang Z, Yang C, Yang YW, Lu XJ, Chen AD, Zhu GQ, Tan X. Asprosin contributes to vascular remodeling in hypertensive rats via superoxide signaling. J Hypertens 2024; 42:1427-1439. [PMID: 38690935 DOI: 10.1097/hjh.0000000000003751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2024]
Abstract
OBJECTIVE Proliferation and migration of vascular smooth muscle cells (VSMCs) contribute to vascular remodeling. Asprosin, a newly discovered protein hormone, is involved in metabolic diseases. Little is known about the roles of asprosin in cardiovascular diseases. This study focused on the role and mechanism of asprosin on VSMC proliferation and migration, and vascular remodeling in a rat model of hypertension. METHODS AND RESULTS VSMCs were obtained from the aortic media of 8-week-old male Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR). Asprosin was upregulated in the VSMCs of SHR. For in vitro studies, asprosin promoted VSMC proliferation and migration of WKY and SHR, and increased Nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX) activity, NOX1/2/4 protein expressions and superoxide production. Knockdown of asprosin inhibited the proliferation, migration, NOX activity, NOX1/2 expressions and superoxide production in the VSMCs of SHR. The roles of asprosin in promoting VSMC proliferation and migration were not affected by hydrogen peroxide scavenger, but attenuated by superoxide scavenger, selective NOX1 or NOX2 inhibitor. Toll-like receptor 4 (TLR4) was upregulated in SHR, TLR4 knockdown inhibited asprosin overexpression-induced proliferation, migration and oxidative stress in VSMCs of WKY and SHR. Asprosin was upregulated in arteries of SHR, and knockdown of asprosin in vivo not only attenuated oxidative stress and vascular remodeling in aorta and mesentery artery, but also caused a subsequent persistent antihypertensive effect in SHR. CONCLUSIONS Asprosin promotes VSMC proliferation and migration via NOX-mediated superoxide production. Inhibition of endogenous asprosin expression attenuates VSMC proliferation and migration, and vascular remodeling of SHR.
Collapse
Affiliation(s)
- Zhi-Qin Xu
- Emergency Department
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University
| | - Xiu-Zhen Li
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University
| | | | - Rui Ge
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | | | | | | | | | | | - Xue-Juan Lu
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University
| | - Ai-Dong Chen
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center of Translational Medicine for Cardiovascular Disease, and Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiao Tan
- Emergency Department
- Department of Cardiology, The Second Affiliated Hospital of Nanjing Medical University
| |
Collapse
|
17
|
Liu K, Li Y, Yin F, Wu X, Zhang X, Jiang D, Wang J, Zhang Z, Wang R, Chen C, Han Y. Elucidating thoracic aortic dissection pathogenesis: The interplay of m1A-related gene expressions and miR-16-5p/YTHDC1 Axis in NLRP3-dependent pyroptosis. Int J Biol Macromol 2024; 274:133293. [PMID: 38925173 DOI: 10.1016/j.ijbiomac.2024.133293] [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: 01/21/2024] [Revised: 05/23/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
The underlying molecular mechanisms of thoracic aortic dissection (TAD) remain incompletely understood. Recent insights into RNA methylation and microRNA-mediated gene regulation offer new avenues for exploring how these processes contribute to the pathophysiology of TAD, particularly through the modulation of pyroptosis and smooth muscle cell viability. This research aimed to elucidate the interplay of m1A-related gene expressions and miR-16-5p/YTHDC1 Axis in NLRP3-dependent pyroptosis, a mechanism implicated in the pathogenesis of TAD. We collected tissue samples from 28 human TAD patients and 8 healthy aortic group, as well as utilized a mouse model to replicate the disease. A combination of computational, in vitro, and in vivo methods was applied, including CIBERSORTx analysis, Pearson correlation, gene transfection using antagomiR-16-5p, siRNA, and several staining as well as cell culture techniques. Our analysis indicated two differentially expressed genes, ALKBH2 and YTHDC1. We found significant upregulation of has-miR-16-5p and downregulation of YTHDC1 at mRNA level in AD samples. Immune cell infiltration in TAD samples was examined using the CIBERSORTx database. We confirmed that YTHDC1 was a target of miR-16-5p, as evidenced by an inhibitory effect on luciferase activity. Inhibition of miR-16-5p enhanced SMC proliferation and promoted cell viability whilst downregulating NLRP3-pyroptosis. YTHDC1 expression was increased, and NLRP3-pyroptosis expressions were inhibited, suggesting miR-16-5p/YTHDC1 axis may involve the NLRP3-pyroptosis of the SMC. In vivo analyses confirmed the prevention of NLRP3-pyroptosis in middle layer of the thoracic aorta, implying that the miR-16-5p/YTHDC1 axis regulated SMC proliferation via NLRP3-pyroptosis signaling. Our findings underscored the anti-pyroptotic role of miR-16-5p/YTHDC1 axis in the pathogenesis of TAD, suggesting a potential therapeutic strategy via targeting YTHDC1 and suppressing miR-16-5p to inhibit NLRP3-dependent pyroptosis. Although further investigation is needed, these results relating to SMC proliferation are a significant step forward in understanding TAD.
Collapse
Affiliation(s)
- Kun Liu
- Department of Cardiac Surgery, Affiliated Hospital, Guizhou Medical University, Guiyang, China
| | - Yuemeng Li
- Department of Vascular Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Fanxing Yin
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Xiaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaoxu Zhang
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Deying Jiang
- Department of Vascular Surgery, Central Hospital of Dalian University of Technology, Dalian, China
| | - Jian Wang
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Zhaoxuan Zhang
- School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China
| | - Ruihua Wang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, Brisbane, QLD, Australia
| | - Yanshuo Han
- Department of Vascular Surgery, Central Hospital of Dalian University of Technology, Dalian, China; School of Life and Pharmaceutical Science, Dalian University of Technology, Panjin, China.
| |
Collapse
|
18
|
Lu Y, Liang X, Song J, Guan Y, Yang L, Shen R, Niu Y, Guo Z, Zhu N. Niclosamide modulates phenotypic switch and inflammatory responses in human pulmonary arterial smooth muscle cells. Mol Cell Biochem 2024:10.1007/s11010-024-05061-6. [PMID: 38980591 DOI: 10.1007/s11010-024-05061-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 06/29/2024] [Indexed: 07/10/2024]
Abstract
Excessive proliferation and migration of pulmonary arterial smooth muscle cells (PASMCs) represent key steps of pulmonary vascular remodeling, leading to the development of pulmonary arterial hypertension (PAH) and right ventricular failure. Niclosamide (NCL), an FDA-approved anthelmintic, has been shown to regulate cell proliferation, migration, invasion, and apoptosis through a variety of signaling pathways. However, its role on modulating the phenotypic switch and inflammatory responses in PASMCs remains unclear. In this study, cell proliferation assay showed that NCL inhibited PDGF-BB induced proliferation of human PASMCs in a dose-dependent manner. Western blot analysis further confirmed a notable reduction in the expression of cyclin D1 and PCNA proteins. Subsequently, flow cytometry analysis demonstrated that NCL induced an increased percentage of cells in the G1 phase while promoting apoptosis in PASMCs. Moreover, both scratch wound assay and transwell assay confirmed that NCL decreased PDGF-BB-induced migration of PASMCs. Mechanistically, western blot revealed that pretreatment of PASMCs with NCL markedly restored the protein levels of SMA, SM22, and calponin, while reducing phosphorylation of P38/STAT3 signaling in the presence of PDGF-BB. Interestingly, macrophages adhesion assay showed that NCL markedly reduced recruitment of Calcein-AM labeled RAW264.7 by TNFα-stimulated PASMCs. Western blot revealed that NCL suppressed TNFα-induced expression of both of VCAM-1 and ICAM-1 proteins. Furthermore, pretreatment of PASMCs with NCL significantly inhibited NLRP3 inflammasome activity through reducing NLRP3, AIM2, mature interleukin-1β (IL-β), and cleaved Caspase-1 proteins expression. Together, these results suggested versatile effects of NCL on controlling of proliferation, migration, and inflammatory responses in PASMCs through modulating different pathways, indicating that repurposing of NCL may emerge as a highly effective drug for PAH treatment.
Collapse
Affiliation(s)
- Yuwen Lu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Xiaogan Liang
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Jingwen Song
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yugen Guan
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Liang Yang
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Rongrong Shen
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Yunpu Niu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China
| | - Zhifu Guo
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| | - Ni Zhu
- Department of Cardiology, Changhai Hospital, Naval Medical University, 168 Changhai Road, Shanghai, 200433, China.
| |
Collapse
|
19
|
Chen N, Wu S, Zhi K, Zhang X, Guo X. ZFP36L1 controls KLF16 mRNA stability in vascular smooth muscle cells during restenosis after vascular injury. J Mol Cell Cardiol 2024; 192:13-25. [PMID: 38653384 DOI: 10.1016/j.yjmcc.2024.04.012] [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: 09/10/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 04/25/2024]
Abstract
The RNA-binding zinc finger protein 36 (ZFP36) family participates in numerous physiological processes including transition and differentiation through post-transcriptional regulation. ZFP36L1 is a member of the ZFP36 family. This study aimed to evaluate the role of ZFP36L1 in restenosis. We found that the expression of ZFP36L1 was inhibited in VSMC-phenotypic transformation induced by TGF-β, PDGF-BB, and FBS and also in the rat carotid injury model. In addition, we found that the overexpression of ZFP36L1 inhibited the proliferation and migration of VSMCs and promoted the expression of VSMC contractile genes; whereas ZFP36L1 interference promoted the proliferation and migration of VSMCs and suppressed the expression of contractile genes. Furthermore, the RNA binding protein immunoprecipitation and double luciferase reporter gene experiments shows that ZFP36L1 regulates the phenotypic transformation of VSMCs through the posttranscriptional regulation of KLF16. Finally, our research results in the rat carotid balloon injury animal model further confirmed that ZFP36L1 regulates the phenotypic transformation of VSMCs through the posttranscriptional regulation of KLF16 and further plays a role in vascular injury and restenosis in vivo.
Collapse
Affiliation(s)
- Ningheng Chen
- Department of Vascular surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shiyong Wu
- Department of Vascular surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kangkang Zhi
- Department of Vascular surgery, Second Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Xiaoping Zhang
- Clinical Nuclear Medicine Center, Imaging Clinical Medical Center, Institute of Nuclear Medicine, Department of Nuclear Medicine, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai 200072, China.
| | - Xueli Guo
- Department of Vascular surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
20
|
Dos Passos RR, Santos CV, Priviero F, Briones AM, Tostes RC, Webb RC, Bomfim GF. Immunomodulatory Activity of Cytokines in Hypertension: A Vascular Perspective. Hypertension 2024; 81:1411-1423. [PMID: 38686582 PMCID: PMC11168883 DOI: 10.1161/hypertensionaha.124.21712] [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] [Indexed: 05/02/2024]
Abstract
Cytokines play a crucial role in the structure and function of blood vessels in hypertension. Hypertension damages blood vessels by mechanisms linked to shear forces, activation of the renin-angiotensin-aldosterone and sympathetic nervous systems, oxidative stress, and a proinflammatory milieu that lead to the generation of neoantigens and damage-associated molecular patterns, ultimately triggering the release of numerous cytokines. Damage-associated molecular patterns are recognized by PRRs (pattern recognition receptors) and activate inflammatory mechanisms in endothelial cells, smooth muscle cells, perivascular nerves, and perivascular adipose tissue. Activated vascular cells also release cytokines and express factors that attract macrophages, dendritic cells, and lymphocytes to the blood vessels. Activated and differentiated T cells into Th1, Th17, and Th22 in secondary lymphoid organs migrate to the vessels, releasing specific cytokines that further contribute to vascular dysfunction and remodeling. This chronic inflammation alters the profile of endothelial and smooth muscle cells, making them dysfunctional. Here, we provide an overview of how cytokines contribute to hypertension by impacting the vasculature. Furthermore, we explore clinical perspectives about the modulation of cytokines as a potential therapeutic intervention to specifically target hypertension-linked vascular dysfunction.
Collapse
Affiliation(s)
- Rinaldo R Dos Passos
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
| | - Cintia V Santos
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil (C.V.S., R.C.T.)
| | - Fernanda Priviero
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Biomedical Engineering, College of Engineering and Computing (F.P., R.C.W.), University of South Carolina, Columbia
| | - Ana M Briones
- Department of Pharmacology, Facultad de Medicina, Universidad Autónoma de Madrid, Spain (A.M.B.)
- Instituto de Investigación Sanitaria del Hospital Universitario La Paz (IdiPAZ), Madrid, Spain (A.M.B.)
- CIBER Cardiovascular, Madrid, Spain (A.M.B.)
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of Sao Paulo, Brazil (C.V.S., R.C.T.)
| | - R Clinton Webb
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- Department of Biomedical Engineering, College of Engineering and Computing (F.P., R.C.W.), University of South Carolina, Columbia
| | - Gisele F Bomfim
- Cardiovascular Translational Research Center, School of Medicine (R.R.d.P., C.V.S., F.P., R.C.W., G.F.B.), University of South Carolina, Columbia
- NUPADS - Health Education and Research Center, Institute of Health Sciences, Federal University of Mato Grosso, Sinop, Brazil (G.F.B.)
| |
Collapse
|
21
|
Di C, Ji M, Li W, Liu X, Gurung R, Qin B, Ye S, Qi R. Pyroptosis of Vascular Smooth Muscle Cells as a Potential New Target for Preventing Vascular Diseases. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07578-w. [PMID: 38822974 DOI: 10.1007/s10557-024-07578-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/03/2024]
Abstract
Vascular remodeling is the adaptive response of the vessel wall to physiological and pathophysiological changes, closely linked to vascular diseases. Vascular smooth muscle cells (VSMCs) play a crucial role in this process. Pyroptosis, a form of programmed cell death characterized by excessive release of inflammatory factors, can cause phenotypic transformation of VSMCs, leading to their proliferation, migration, and calcification-all of which accelerate vascular remodeling. Inhibition of VSMC pyroptosis can delay this process. This review summarizes the impact of pyroptosis on VSMCs and the pathogenic role of VSMC pyroptosis in vascular remodeling. We also discuss inhibitors of key proteins in pyroptosis pathways and their effects on VSMC pyroptosis. These findings enhance our understanding of the pathogenesis of vascular remodeling and provide a foundation for the development of novel medications that target the control of VSMC pyroptosis as a potential treatment strategy for vascular diseases.
Collapse
Affiliation(s)
- Chang Di
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, Haidian District, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing, 100191, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Peking University, Beijing, 100191, China.
| | - Meng Ji
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, Haidian District, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Peking University, Beijing, 100191, China
| | - Wenjin Li
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, Haidian District, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Peking University, Beijing, 100191, China
| | - Xiaoyi Liu
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, Haidian District, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Peking University, Beijing, 100191, China
| | - Rijan Gurung
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Boyang Qin
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, Haidian District, China
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing, 100191, China
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Peking University, Beijing, 100191, China
| | - Shu Ye
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
| | - Rong Qi
- Department of Pharmacology, School of Basic Medical Sciences, Peking University Health Science Center, 38 Xueyuan Road, Beijing, 100191, Haidian District, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China.
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Peking University, Beijing, 100191, China.
- Beijing Key Laboratory of Molecular Pharmaceutics and New Drug Delivery Systems, Peking University, Beijing, 100191, China.
- Cardiovascular Disease Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
| |
Collapse
|
22
|
Rios FJ, de Ciuceis C, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Lopreiato M, Mavraganis G, Mengozzi A, Montezano AC, Stavropoulos K, Winklewski PJ, Wolf J, Costantino S, Doumas M, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Virdis A. Mechanisms of Vascular Inflammation and Potential Therapeutic Targets: A Position Paper From the ESH Working Group on Small Arteries. Hypertension 2024; 81:1218-1232. [PMID: 38511317 DOI: 10.1161/hypertensionaha.123.22483] [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: 03/22/2024]
Abstract
Inflammatory responses in small vessels play an important role in the development of cardiovascular diseases, including hypertension, stroke, and small vessel disease. This involves various complex molecular processes including oxidative stress, inflammasome activation, immune-mediated responses, and protein misfolding, which together contribute to microvascular damage. In addition, epigenetic factors, including DNA methylation, histone modifications, and microRNAs influence vascular inflammation and injury. These phenomena may be acquired during the aging process or due to environmental factors. Activation of proinflammatory signaling pathways and molecular events induce low-grade and chronic inflammation with consequent cardiovascular damage. Identifying mechanism-specific targets might provide opportunities in the development of novel therapeutic approaches. Monoclonal antibodies targeting inflammatory cytokines and epigenetic drugs, show promise in reducing microvascular inflammation and associated cardiovascular diseases. In this article, we provide a comprehensive discussion of the complex mechanisms underlying microvascular inflammation and offer insights into innovative therapeutic strategies that may ameliorate vascular injury in cardiovascular disease.
Collapse
Affiliation(s)
- Francisco J Rios
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Carolina de Ciuceis
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Georgios Georgiopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Antonios Lazaridis
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Ryszard Nosalski
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - George Pavlidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Simon Tual-Chalot
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
| | - Claudia Agabiti-Rosei
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
| | - Livia L Camargo
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Edyta Dąbrowska
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Fosca Quarti-Trevano
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Marcin Hellmann
- Cardiac Diagnostics (M.H.), Medical University of Gdansk, Poland
| | - Stefano Masi
- Institute of Cardiovascular Science, University College London, United Kingdom (S.M.)
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Mariarosaria Lopreiato
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Georgios Mavraganis
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Alessandro Mengozzi
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- Health Science Interdisciplinary Center, Scuola Superiore Sant'Anna, Pisa (A.M.)
| | - Augusto C Montezano
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Konstantinos Stavropoulos
- Second Medical Department, Hippokration Hospital, Aristotle University of Thessaloniki, Greece (K. Stavropoulos)
| | - Pawel J Winklewski
- Departments of Human Physiology (P.J.W.), Medical University of Gdansk, Poland
| | - Jacek Wolf
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Sarah Costantino
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
| | - Michael Doumas
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Eugenia Gkaliagkousi
- Third Department of Internal Medicine, Aristotle University of Thessaloniki, Papageorgiou Hospital, Greece (A.L., E.G.)
| | - Guido Grassi
- Clinica Medica, Department of Medicine and Surgery, University of Milano-Bicocca, Milan, Italy (F.Q.-T., G.G.)
| | - Tomasz J Guzik
- Centre for Cardiovascular Sciences; Queen's Medical Research Institute, University of Edinburgh, United Kingdom (R.N., T.J.G.)
- Department of Internal Medicine, Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland (R.N., T.J.G.)
| | - Ignatios Ikonomidis
- Medical School (G.P., I.I.), National and Kapodistrian University of Athens
- Preventive Cardiology Laboratory and Clinic of Cardiometabolic Diseases, 2-Cardiology Department, Attikon Hospital, Athens, Greece (G.P., I.I.)
| | - Krzysztof Narkiewicz
- Department of Hypertension and Diabetology, Center of Translational Medicine (E.D., J.W., K.N.) and M.D.)
| | - Francesco Paneni
- Center for Translational and Experimental Cardiology, Department of Cardiology, University Hospital Zurich, University of Zurich, Switzerland (A.M., F.P.)
- University Heart Center (S.C., F.P.), University Hospital Zurich, Switzerland
- Department of Research and Education (F.P.), University Hospital Zurich, Switzerland
| | - Damiano Rizzoni
- Department of Clinical and Experimental Sciences, University of Brescia (C.d.C., C.A.-R., D.R.)
- Division of Medicine, Spedali Civili di Brescia, Italy (D.R.)
| | - Kimon Stamatelopoulos
- Angiology and Endothelial Pathophysiology Unit, Department of Clinical Therapeutics, Medical School (G.G., G.M., K. Stamatelopoulos), National and Kapodistrian University of Athens
| | - Konstantinos Stellos
- Biosciences Institute, Vascular Biology and Medicine Theme, Faculty of Medical Sciences, Newcastle University, United Kingdom (S.T.-C., K. Stellos)
- Department of Cardiovascular Research, European Center for Angioscience, Medical Faculty Mannheim (K. Stellos), Heidelberg University, Germany
- Department of Cardiology, University Hospital Mannheim (K. Stellos), Heidelberg University, Germany
- German Centre for Cardiovascular Research, Heidelberg/Mannheim Partner Site (K. Stellos)
| | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| | - Rhian M Touyz
- Research Institute of the McGill University Health Centre, McGill University, Montreal, Canada (F.J.R., L.L.C., A.C.M., R.M.T.)
| | - Agostino Virdis
- Department of Clinical and Experimental Medicine, University of Pisa, Italy (S.M., M.L., A.M., S.T., A.V.)
| |
Collapse
|
23
|
Liu Y, Wang L, Lei D, Tan X, Jin W, Hou M, Hu K, Yan Y, Wang H, Xiang C, Lai Y. Circ_0000006 and circ_0000160 regulate hsa-let-7e-5p/UBQLN4 axis in aortic dissection progression. PLoS One 2024; 19:e0304668. [PMID: 38820386 PMCID: PMC11142605 DOI: 10.1371/journal.pone.0304668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 05/15/2024] [Indexed: 06/02/2024] Open
Abstract
Aortic aneurysms (AA) and aorta dissection (AD) are life-threatening conditions with a rising incidence and high mortality rate. Recent research has linked non-coding RNAs to the regulation of AA and AD progression. In this study, we performed circRNA sequencing, microRNA (miRNA) sequencing, and messenger RNA (mRNA) sequencing on plasma samples from AA and AD patients to identify the key circRNA-miRNA-mRNA axis involved in the transition from AA to AD. Our results showed elevated levels of circ_0000006 and circ_0000160, along with decreased levels of hsa-let-7e-5p in AD samples compared to AA samples. Predictive analysis suggested that circ_0000006 and circ_0000160 potentially target hsa-let-7e-5p, which in turn may bind to the mRNA of Ubiquilin 4 (UBQLN4). In an AD cell model using vascular smooth muscle cells (VSMCs), silencing circ_0000006 and circ_0000160 attenuated the effects of platelet-derived growth factor (PDGF)-induced phenotypic changes, proliferation, and migration. This effect was partially reversed by inhibiting hsa-let-7e-5p. Furthermore, we found that overexpression of UBQLN4 counteracted the effects of hsa-let-7e-5p, suggesting UBQLN4 as a downstream mediator of hsa-let-7e-5p. In an animal model of AD, knockdown of circ_0000006 and circ_0000160 also showed protective effects against aortic septation. Overall, our findings indicate that the upregulation of circ_0000006 and circ_0000160 contributes to the progression from AA to AD by influencing abnormal phenotypic changes, migration, and proliferation of VSMCs. The Hsa-let-7e-5p/UBQLN4 axis may play a critical role in AD development. Targeting circ_0000006 and circ_0000160 could be a potential therapeutic strategy for preventing the progression of AD.
Collapse
Affiliation(s)
- Yong Liu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Liang Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Dongyun Lei
- Department of Dermatology, Tianjin Academy of Traditional Chinese Medicine Affiliated Hospital, Tianjin, China
| | - Xiong Tan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Weitao Jin
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ming Hou
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Kai Hu
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yu Yan
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Hao Wang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Chaohu Xiang
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Yinglong Lai
- Department of Cardiovascular Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
24
|
Ding P, Song Y, Yang Y, Zeng C. NLRP3 inflammasome and pyroptosis in cardiovascular diseases and exercise intervention. Front Pharmacol 2024; 15:1368835. [PMID: 38681198 PMCID: PMC11045953 DOI: 10.3389/fphar.2024.1368835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/02/2024] [Indexed: 05/01/2024] Open
Abstract
NOD-like receptor protein 3 (NLRP3) inflammasome is an intracellular sensing protein complex that possesses NACHT, leucine-rich repeat, and pyrin domain, playing a crucial role in innate immunity. Activation of the NLRP3 inflammasome leads to the production of pro-inflammatory cellular contents, such as interleukin (IL)-1β and IL-18, and induction of inflammatory cell death known as pyroptosis, thereby amplifying or sustaining inflammation. While a balanced inflammatory response is beneficial for resolving damage and promoting tissue healing, excessive activation of the NLRP3 inflammasome and pyroptosis can have harmful effects. The involvement of the NLRP3 inflammasome has been observed in various cardiovascular diseases (CVD). Indeed, the NLRP3 inflammasome and its associated pyroptosis are closely linked to key cardiovascular risk factors including hyperlipidemia, diabetes, hypertension, obesity, and hyperhomocysteinemia. Exercise compared with medicine is a highly effective measure for both preventing and treating CVD. Interestingly, emerging evidence suggests that exercise improves CVD and inhibits the activity of NLRP3 inflammasome and pyroptosis. In this review, the activation mechanisms of the NLRP3 inflammasome and its pathogenic role in CVD are critically discussed. Importantly, the purpose is to emphasize the crucial role of exercise in managing CVD by suppressing NLRP3 inflammasome activity and proposes it as the foundation for developing novel treatment strategies.
Collapse
Affiliation(s)
- Ping Ding
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yuanming Song
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yang Yang
- Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, China
| | - Cheng Zeng
- Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, China
- Key Specialty of Clinical Pharmacy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, China
- Guangdong Key Laboratory of Pharmaceutical Bioactive Substances, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
25
|
Qiu X, Zhang Y, Xu YJ, Liang ZD, Dai XC, Xiao WL, Zhang XJ, Li XL. Euphzycopins A - D, macrocyclic diterpenoids with potential anti-inflammatory activity from Euphorbia Helioscopia. Fitoterapia 2024; 173:105821. [PMID: 38211643 DOI: 10.1016/j.fitote.2024.105821] [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: 09/11/2023] [Revised: 12/06/2023] [Accepted: 01/08/2024] [Indexed: 01/13/2024]
Abstract
Four new diterpenoids (1-4) and four known diterpenoids (5-8) were purified from the whole plant of Euphorbia helioscopia L. Compounds 1 and 2 were jathophanes diterpenoids with a 5/12 polycyclic systems, compound 3 was rhamofolane diterpenoid with a 5/10 bicyclic skeleton and compound 4 was a rare class of euphorbia diterpenes featuring an unusual 5/10 fused ring system. Anti-inflammatory activity tests were conducted on the separated compounds, indicating that compound 4 had significant inhibitory effect on NLRP3 inflammasome with an IC50 value of 7.75 μM. Further, the inhibitory effect of 4 was determined using immunofluorescence assays.
Collapse
Affiliation(s)
- Xiong Qiu
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Yu Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Yao-Jun Xu
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Zhong-Dan Liang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Xiao-Chang Dai
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China
| | - Wei-Lie Xiao
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China.
| | - Xing-Jie Zhang
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China.
| | - Xiao-Li Li
- Key Laboratory of Medicinal Chemistry for Natural Resource of Ministry of Education, Yunnan Characteristic Plant Extraction Laboratory, Yunnan Research & Development Center for Natural Products, School of Chemical Science and Technology, and School of Pharmacy, State Key Laboratory for Conservation and Utilization of Bio-Resources in Yunnan, Yunnan University, Kunming 650500, People's Republic of China.
| |
Collapse
|
26
|
Liu P, Wang Y, Tian K, Bai X, Wang Y, Wang Y. Artesunate inhibits macrophage-like phenotype switching of vascular smooth muscle cells and attenuates vascular inflammatory injury in atherosclerosis via NLRP3. Biomed Pharmacother 2024; 172:116255. [PMID: 38325261 DOI: 10.1016/j.biopha.2024.116255] [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: 11/12/2023] [Revised: 01/23/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024] Open
Abstract
Inflammation is one of the main pathogenic factors of atherosclerosis (AS), and the phenotypic transformation of macrophages in human vascular smooth muscle cells (HVSMCs) contributes to the inflammatory injury of blood vessels and the formation of atherosclerotic plaques. Artesunate reportedly exerts anti-inflammatory activity against AS. Herein, we aimed to explore the artesunate-mediated anti-inflammatory and HVSMC phenotypic switch effects against AS and elucidate potential underlying mechanisms. In vitro, artesunate decreased expression of NLRP3, caspase-1, and interleukin (IL)- 1β. Artesunate significantly inhibited low-density lipoprotein (LDL) expression in HVSMCs and macrophages. In vivo, artesunate reduced atherosclerotic plaque formation in high-fat diet (HFD)-fed ApoE-/- mice, as well as decreased NLRP3 and CD68 expression in atherosclerotic plaques. Artesunate decreased serum levels of triglycerides and increased high-density lipoprotein levels in HFD-med mice; however, serum levels of total cholesterol and LDL were unaltered. Treatment with artesunate substantially increased α-smooth muscle actin expression in aortic tissues while inhibiting expression levels of NLRP3, IL-1β, heparinase, matrix metalloproteinase 9, and Krüppel-like factor 4 (KLF4). Collectively, our findings suggest that artesunate-mediated effects may involve inhibition of the ERK1/2/NF-κB/IL-1β pathway in HVSMCs via the downregulation of NLRP3 expression. Thus, artesunate could serve as a novel strategy to treat AS by inhibiting AS plaque formation and suppressing macrophage-like phenotype switching of HVSMCs.
Collapse
Affiliation(s)
- Ping Liu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Yuqi Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Keke Tian
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Xinyu Bai
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China
| | - Yaowen Wang
- Department of Cardiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing Cardiac Arrhythmias Therapeutic Service Center, Chongqing 400010, China.
| | - Yan Wang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi 563000, China.
| |
Collapse
|
27
|
Qiao H, Yang B, Lv X, Liu Y. Exposure to TCBPA stimulates the growth of arterial smooth muscle cells through the activation of the ROS/NF-κB/NLRP3 signaling pathway. Toxicology 2024; 503:153759. [PMID: 38369010 DOI: 10.1016/j.tox.2024.153759] [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: 01/25/2024] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 02/20/2024]
Abstract
Tetrachlorobisphenol A (TCBPA) and Tetrabromobisphenol S (TBBPS) are organic compounds widely used in industrial production, including in plastic and textile manufacturing. Presently, residual TCBPA is commonly detected in the environment as well as in human and animal sera. Therefore, it is imperative to assess the potential toxicological effects of TCBPA on organismal health. A series of biochemical experiments, including indirect immunofluorescence, ELISA, Western blot, MTT, etc, were conducted to analyze the effects of TCBPA on vascular smooth muscle cells. In this study, the biological impact of TCBPA on arterial smooth muscle cells (ASMCs) was investigated. CCK8 and EdU assays demonstrated significant proliferation of ASMCs following TCBPA treatment. Furthermore, TCBPA induced an inflammatory response in smooth muscle cells, as evidenced by the upregulated expression of inflammatory cytokines including IL-6, IL-1β, and MCP1. Additionally, we observed that TCBPA triggered an oxidative stress response in ASMCs by measuring ROS levels. To elucidate the underlying molecular mechanism of TCBPA-induced ASMC proliferation, we found that NLRP3 was essential for this process. Further investigation revealed that NLRP3 activation was mediated by NF-κB (which was activated by ROS). In summary, our findings suggest that TCBPA promotes the proliferation of ASMCs through the ROS/NF-κB/NLRP3 signaling cascade. This work indicates that TCBPA may represent a potential risk factor for the development of atherosclerosis, highlighting the need for judicious control of TCBPA usage.
Collapse
Affiliation(s)
- Huanyu Qiao
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Bo Yang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Xiaoshuo Lv
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China
| | - Yongmin Liu
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China; Beijing Institute of Heart Lung and Blood Vessel Diseases, Beijing, China.
| |
Collapse
|
28
|
Yin Z, Zhang J, Zhao M, Peng S, Ye J, Liu J, Xu Y, Xu S, Pan W, Wei C, Qin J, Wan J, Wang M. Maresin-1 ameliorates hypertensive vascular remodeling through its receptor LGR6. MedComm (Beijing) 2024; 5:e491. [PMID: 38463394 PMCID: PMC10924638 DOI: 10.1002/mco2.491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 12/30/2023] [Accepted: 01/11/2024] [Indexed: 03/12/2024] Open
Abstract
Hypertensive vascular remodeling is defined as the changes in vascular function and structure induced by persistent hypertension. Maresin-1 (MaR1), one of metabolites from Omega-3 fatty acids, has been reported to promote inflammation resolution in several inflammatory diseases. This study aims to investigate the effect of MaR1 on hypertensive vascular remodeling. Here, we found serum MaR1 levels were reduced in hypertensive patients and was negatively correlated with systolic blood pressure (SBP). The treatment of MaR1 reduced the elevation of blood pressure and alleviated vascular remodeling in the angiotensin II (AngII)-infused mouse model. In addition, MaR1-treated vascular smooth muscle cells (VSMCs) exhibited reduced excessive proliferation, migration, and phenotype switching, as well as impaired pyroptosis. However, the knockout of the receptor of MaR1, leucine-rich repeat-containing G protein-coupled receptor 6 (LGR6), was seen to aggravate pathological vascular remodeling, which could not be reversed by additional MaR1 treatment. The mechanisms by which MaR1 regulates vascular remodeling through LGR6 involves the Ca2+/calmodulin-dependent protein kinase II/nuclear factor erythroid 2-related factor 2/heme oxygenase-1 signaling pathway. Overall, supplementing MaR1 may be a novel therapeutic strategy for the prevention and treatment of hypertension.
Collapse
Affiliation(s)
- Zheng Yin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jishou Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Mengmeng Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Shanshan Peng
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jing Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Jianfang Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Yao Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Shuwan Xu
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Wei Pan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Cheng Wei
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Juan‐Juan Qin
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Center for Healthy AgingWuhan University School of NursingWuhanChina
| | - Jun Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| | - Menglong Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Department of Geriatrics, Zhongnan Hospital of Wuhan UniversityWuhan UniversityWuhanChina
- Cardiovascular Research InstituteWuhan UniversityWuhanChina
- Hubei Key Laboratory of CardiologyWuhanChina
| |
Collapse
|
29
|
Li J, Yao H, Zhao F, An J, Wang Q, Mu J, Liu Z, Zou MH, Xie Z. Pycard deficiency inhibits microRNA maturation and prevents neointima formation by promoting chaperone-mediated autophagic degradation of AGO2/argonaute 2 in adipose tissue. Autophagy 2024; 20:629-644. [PMID: 37963060 PMCID: PMC10936599 DOI: 10.1080/15548627.2023.2277610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/10/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023] Open
Abstract
PYCARD (PYD and CARD domain containing), a pivotal adaptor protein in inflammasome assembly and activation, contributes to innate immunity, and plays an essential role in the pathogenesis of atherosclerosis and restenosis. However, its roles in microRNA biogenesis remain unknown. Therefore, this study aimed to investigate the roles of PYCARD in miRNA biogenesis and neointima formation using pycard knockout (pycard-/-) mice. Deficiency of Pycard reduced circulating miRNA profile and inhibited Mir17 seed family maturation. The systemic pycard knockout also selectively reduced the expression of AGO2 (argonaute RISC catalytic subunit 2), an important enzyme in regulating miRNA biogenesis, by promoting chaperone-mediated autophagy (CMA)-mediated degradation of AGO2, specifically in adipose tissue. Mechanistically, pycard knockout increased PRMT8 (protein arginine N-methyltransferase 8) expression in adipose tissue, which enhanced AGO2 methylation, and subsequently promoted its binding to HSPA8 (heat shock protein family A (Hsp70) member 8) that targeted AGO2 for lysosome degradation through chaperone-mediated autophagy. Finally, the reduction of AGO2 and Mir17 family expression prevented vascular injury-induced neointima formation in Pycard-deficient conditions. Overexpression of AGO2 or administration of mimic of Mir106b (a major member of the Mir17 family) prevented Pycard deficiency-mediated inhibition of neointima formation in response to vascular injury. These data demonstrate that PYCARD inhibits CMA-mediated degradation of AGO2, which promotes microRNA maturation, thereby playing a critical role in regulating neointima formation in response to vascular injury independently of inflammasome activity and suggest that modulating PYCARD expression and function may represent a powerful therapeutic strategy for neointima formation.Abbreviations: 6-AN: 6-aminonicotinamide; ACTB: actin, beta; aDMA: asymmetric dimethylarginine; AGO2: argonaute RISC catalytic subunit 2; CAL: carotid artery ligation; CALCOCO2: calcium binding and coiled-coil domain 2; CMA: chaperone-mediated autophagy; CTSB: cathepsin B; CTSD: cathepsin D; DGCR8: DGCR8 microprocessor complex subunit; DOCK2: dedicator of cyto-kinesis 2; EpiAdi: epididymal adipose tissue; HSPA8: heat shock protein family A (Hsp70) member 8; IHC: immunohistochemical; ISR: in-stent restenosis; KO: knockout; LAMP2: lysosomal-associated membrane protein 2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; miRNA: microRNA; NLRP3: NLR family pyrin domain containing 3; N/L: ammonium chloride combined with leupeptin; PRMT: protein arginine methyltransferase; PVAT: peri-vascular adipose tissues; PYCARD: PYD and CARD domain containing; sDMA: symmetric dimethylarginine; ULK1: unc-51 like kinase 1; VSMCs: vascular smooth muscle cells; WT: wild-type.
Collapse
Affiliation(s)
- Jian Li
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Hongmin Yao
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Fujie Zhao
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Junqing An
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Qilong Wang
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Jing Mu
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Zhixue Liu
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Ming-Hui Zou
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| | - Zhonglin Xie
- Center of Molecular and Translational Medicine, Georgia State University, Atlanta, Georgia
| |
Collapse
|
30
|
Solanki K, Bezsonov E, Orekhov A, Parihar SP, Vaja S, White FA, Obukhov AG, Baig MS. Effect of reactive oxygen, nitrogen, and sulfur species on signaling pathways in atherosclerosis. Vascul Pharmacol 2024; 154:107282. [PMID: 38325566 DOI: 10.1016/j.vph.2024.107282] [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: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease in which fats, lipids, cholesterol, calcium, proliferating smooth muscle cells, and immune cells accumulate in the intima of the large arteries, forming atherosclerotic plaques. A complex interplay of various vascular and immune cells takes place during the initiation and progression of atherosclerosis. Multiple reports indicate that tight control of reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) production is critical for maintaining vascular health. Unrestricted ROS and RNS generation may lead to activation of various inflammatory signaling pathways, facilitating atherosclerosis. Given these deleterious consequences, it is important to understand how ROS and RNS affect the signaling processes involved in atherogenesis. Conversely, RSS appears to exhibit an atheroprotective potential and can alleviate the deleterious effects of ROS and RNS. Herein, we review the literature describing the effects of ROS, RNS, and RSS on vascular smooth muscle cells, endothelial cells, and macrophages and focus on how changes in their production affect the initiation and progression of atherosclerosis. This review also discusses the contribution of ROS, RNS, and RSS in mediating various post-translational modifications, such as oxidation, nitrosylation, and sulfation, of the molecules involved in inflammatory signaling.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia; Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; The Cell Physiology and Pathology Laboratory, Turgenev State University of Orel, Orel, Russia
| | - Alexander Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Suraj P Parihar
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Shivani Vaja
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Fletcher A White
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India.
| |
Collapse
|
31
|
Liang R, Qi X, Cai Q, Niu L, Huang X, Zhang D, Ling J, Wu Y, Chen Y, Yang P, Liu J, Zhang J, Yu P. The role of NLRP3 inflammasome in aging and age-related diseases. Immun Ageing 2024; 21:14. [PMID: 38317229 PMCID: PMC10840156 DOI: 10.1186/s12979-023-00395-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/15/2023] [Indexed: 02/07/2024]
Abstract
The gradual aging of the global population has led to a surge in age-related diseases, which seriously threaten human health. Researchers are dedicated to understanding and coping with the complexities of aging, constantly uncovering the substances and mechanism related to aging like chronic low-grade inflammation. The NOD-like receptor protein 3 (NLRP3), a key regulator of the innate immune response, recognizes molecular patterns associated with pathogens and injury, initiating an intrinsic inflammatory immune response. Dysfunctional NLRP3 is linked to the onset of related diseases, particularly in the context of aging. Therefore, a profound comprehension of the regulatory mechanisms of the NLRP3 inflammasome in aging-related diseases holds the potential to enhance treatment strategies for these conditions. In this article, we review the significance of the NLRP3 inflammasome in the initiation and progression of diverse aging-related diseases. Furthermore, we explore preventive and therapeutic strategies for aging and related diseases by manipulating the NLRP3 inflammasome, along with its upstream and downstream mechanisms.
Collapse
Affiliation(s)
- Ruikai Liang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Xinrui Qi
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
- Queen Mary School, Nanchang University, Nanchang, China
| | - Qi Cai
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Liyan Niu
- Huan Kui College of Nanchang University, Nanchang, China
| | - Xi Huang
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Hong Kong, China
| | - Jitao Ling
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Yuting Wu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Yixuan Chen
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Pingping Yang
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Jianping Liu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China
| | - Jing Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China.
| | - Peng Yu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Nanchang University, Jiangxi, Nanchang, China.
| |
Collapse
|
32
|
Sun R, Zhou Y, Liang J, Yang L, Fan Z, Wang H. Interference of MDM2 attenuates vascular endothelial dysfunction in hypertension partly through blocking Notch1/NLRP3 inflammasome pathway. Ann Anat 2024; 252:152183. [PMID: 37926401 DOI: 10.1016/j.aanat.2023.152183] [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: 11/23/2022] [Revised: 10/22/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND Hypertension is a life-threatening disease mainly featured as vascular endothelial dysfunction. This study aims to explore the regulatory role of murine double minute 2 (MDM2) in hypertension and vascular damage. METHODS Mice were infused with angiotensin II (AngII) to establish a hypertension mouse model in vivo and AngII-stimulated HUVECs were constructed to simulate the damage of vascular endothelial cells in hypertension in vitro. The plasmids targeting to MDM2 was injected to mice or transfected to HUVECs. qRT-PCR and western blot were performed to detect corresponding gene expression in mice aorta. Blood pressure was measured. H&E and Masson staining were conducted to evaluate histological changes of aorta. Responses to the acetylcholine (ACh) and sodium nitroprusside (SNP) were assessed in aorta. ZO-1 expression and cell apoptosis were detected by immunofluorescence and TUNEL, respectively. Network formation ability was determined employing a tube formation. RESULTS MDM2 was upregulated in hypertensive mice. Knockdown of MDM2 inhibited AngII-induced high BP, histological damage, vascular relaxation to Ach, and promoted the levels of p-eNOS and ZO-1 in the aorta in hypertensive mice. MDM2 knockdown inactivated Notch1 signaling and NLRP3 inflammasome, while the inhibitory effect of MDM2 knockdown on NLRP3 inflammasome activation was partly restored by the activation of Notch1. Furthermore, knockdown of MDM2 relieved AngII-induced endothelial dysfunction in HUVECs, as well as suppressing AngII-promoted cell apoptosis. Whereas, the impacts generated by MDM2 knockdown were partly weakened by the activation of Notch1 signaling or NLRP3 inflammasome. CONCLUSION In summary, knockdown of MDM2 can attenuate vascular endothelial dysfunction in hypertension, which may be achieved through inhibiting the activation of Notch1 and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Rongyan Sun
- Department of General Practice, The First People's Hospital of Qujing City, Qujing, Yunnan 655000, China
| | - Yubo Zhou
- Department of breast surgery, The First People's Hospital of Qujing City, Qujing, Yunnan 655000, China
| | - Jiao Liang
- Department of General Practice, The First People's Hospital of Qujing City, Qujing, Yunnan 655000, China
| | - Lihong Yang
- Department of General Practice, The First People's Hospital of Qujing City, Qujing, Yunnan 655000, China
| | - Zhengjun Fan
- Department of Ultrasound, The First People's Hospital of Qujing City, Qujing, Yunnan 655000, China
| | - Huali Wang
- Department of Geriatric Medicine, The First People's Hospital of Qujing City, Qujing, Yunnan 655000, China.
| |
Collapse
|
33
|
Liu N, Gong Z, Li Y, Xu Y, Guo Y, Chen W, Sun X, Yin X, Liu W. CTRP3 inhibits myocardial fibrosis through the P2X7R-NLRP3 inflammasome pathway in SHR rats. J Hypertens 2024; 42:315-328. [PMID: 37850974 DOI: 10.1097/hjh.0000000000003591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
BACKGROUND AND PURPOSE Reducing hypertensive myocardial fibrosis is the fundamental approach to preventing hypertensive ventricular remodelling. C1q/TNF-related protein-3 (CTRP3) is closely associated with hypertension. However, the role and mechanism of CTRP3 in hypertensive myocardial fibrosis are unclear. In this study, we aimed to explore the effect of CTRP3 on hypertensive myocardial fibrosis and the potential mechanism. METHODS AND RESULTS WKY and SHR rats were employed, blood pressure, body weight, heart weight, H/BW were measured, and fibrotic-related proteins, CTRP3 and Collagen I were tested in myocardium at 12 and 20 weeks by immunohistochemical staining and Western blotting, respectively. The results showed that compared with the WKY, SBP, DBP, mean arterial pressure and heart rate (HR) were all significantly increased in SHR at 12 and 20 weeks, while heart weight and H/BW were only increased at 20 weeks. Meanwhile, CTRP3 decreased, while Collagen I increased significantly in the SHR rat myocardium at 20 weeks, which compared to the WKY. Moreover, the expression of α-SMA increased from 12 weeks, Collagen I/III and MMP2/9 increased and TIMP-2 decreased until 20 weeks. In order to explore the function and mechanism of CTRP3 in hypertensive fibrosis, Angiotensin II (Ang II) was used to induce hypertension in primary neonatal rat cardiac fibroblasts in vitro . CTRP3 significantly inhibited the Ang II induced activation of fibrotic proteins, purinergic 2X7 receptor (P2X7R)-NLRP3 inflammasome pathway. The P2X7R agonist BzATP significantly exacerbated Ang II-induced NLRP3 inflammasome activation, which was decreased by the P2X7R antagonists A43079, CTRP3 and MCC950. CONCLUSION CTRP3 expression was decreased in the myocardium of SHR rats, and exogenous CTRP3 inhibited Ang II-induced fibrosis in cardiac fibroblasts by regulating the P2X7R-NLRP3 inflammasome pathway, suggesting that CTRP3 is a potential drug for alleviating myocardial fibrosis in hypertensive conditions.
Collapse
Affiliation(s)
- Na Liu
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
| | - Zhaowei Gong
- Department of Cardiology, the Fourth Affiliated Hospital of Harbin Medical University
| | - Yang Li
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
| | - Yang Xu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
| | - Yutong Guo
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
| | - Wenjia Chen
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
| | - Xue Sun
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
| | - Xinhua Yin
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
- Department of Cardiology, Shenzhen University General Hospital, Shenzhen, Guangdong, China
| | - Wenxiu Liu
- Department of Cardiology, the First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang
| |
Collapse
|
34
|
Ray A, Stelloh C, Liu Y, Meyer A, Geurts AM, Cowley AW, Greene AS, Liang M, Rao S. Histone Modifications and Their Contributions to Hypertension. Hypertension 2024; 81:229-239. [PMID: 38031837 PMCID: PMC11229175 DOI: 10.1161/hypertensionaha.123.21755] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
Essential hypertension, a multifaceted disorder, is a worldwide health problem. A complex network of genetic, epigenetic, physiological, and environmental components regulates blood pressure (BP), and any dysregulation of this network may result in hypertension. Growing evidence suggests a role for epigenetic factors in BP regulation. Any alterations in the expression or functions of these epigenetic regulators may dysregulate various determinants of BP, thereby promoting the development of hypertension. Histone posttranslational modifications are critical epigenetic regulators that have been implicated in hypertension. Several studies have demonstrated a clear association between the increased expression of some histone-modifying enzymes, especially HDACs (histone deacetylases), and hypertension. In addition, treatment with HDAC inhibitors lowers BP in hypertensive animal models, providing an excellent opportunity to design new drugs to treat hypertension. In this review, we discuss the potential contribution of different histone modifications to the regulation of BP.
Collapse
Affiliation(s)
- Atrayee Ray
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
- Department of Physiology, Center of Systems Molecular Medicine (A.R., A.M.G., A.C.), Medical College of Wisconsin, Milwaukee
| | - Cary Stelloh
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
| | - Yong Liu
- Department of Physiology, The University of Arizona, Tucson (Y.L., M.L.)
| | - Alison Meyer
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
| | - Aron M Geurts
- Department of Physiology, Center of Systems Molecular Medicine (A.R., A.M.G., A.C.), Medical College of Wisconsin, Milwaukee
| | - Allen W Cowley
- Department of Physiology, Center of Systems Molecular Medicine (A.R., A.M.G., A.C.), Medical College of Wisconsin, Milwaukee
| | | | - Mingyu Liang
- Department of Physiology, The University of Arizona, Tucson (Y.L., M.L.)
| | - Sridhar Rao
- Versiti Blood Research Institute, Milwaukee, WI (A.R., C.S., A.M., S.R.)
- Department of Pediatrics, Section of Hematology/Oncology/Transplantation (S.R.), Medical College of Wisconsin, Milwaukee
| |
Collapse
|
35
|
Zeng X, Yang Y. Molecular Mechanisms Underlying Vascular Remodeling in Hypertension. Rev Cardiovasc Med 2024; 25:72. [PMID: 39077331 PMCID: PMC11263180 DOI: 10.31083/j.rcm2502072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/15/2023] [Accepted: 10/25/2023] [Indexed: 07/31/2024] Open
Abstract
Hypertension, a common cardiovascular disease, is primarily characterized by vascular remodeling. Recent extensive research has led to significant progress in understanding its mechanisms. Traditionally, vascular remodeling has been described as a unidirectional process in which blood vessels undergo adaptive remodeling or maladaptive remodeling. Adaptive remodeling involves an increase in vessel diameter in response to increased blood flow, while maladaptive remodeling refers to the narrowing or thickening of blood vessels in response to pathological conditions. However, recent research has revealed that vascular remodeling is much more complex. It is now understood that vascular remodeling is a dynamic interplay between various cellular and molecular events. This interplay process involves different cell types, including endothelial cells, smooth muscle cells, and immune cells, as well as their interactions with the extracellular matrix. Through these interactions, blood vessels undergo intricate and dynamic changes in structure and function in response to various stimuli. Moreover, vascular remodeling involves various factors and mechanisms such as the renin-angiotensin-aldosterone system (RAS), oxidative stress, inflammation, the extracellular matrix (ECM), sympathetic nervous system (SNS) and mechanical stress that impact the arterial wall. These factors may lead to vascular and circulatory system diseases and are primary causes of long-term increases in systemic vascular resistance in hypertensive patients. Additionally, the presence of stem cells in adventitia, media, and intima of blood vessels plays a crucial role in vascular remodeling and disease development. In the future, research will focus on examining the underlying mechanisms contributing to hypertensive vascular remodeling to develop potential solutions for hypertension treatment. This review provides us with a fresh perspective on hypertension and vascular remodeling, undoubtedly sparking further research efforts aimed at uncovering more potent treatments and enhanced preventive and control measures for this disease.
Collapse
Affiliation(s)
- Xinyi Zeng
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| | - Yan Yang
- Key Lab of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Lab of Sichuan Province, Collaborative Innovation Center for Prevention and Treatment of Cardiovascular Disease, Institute of Cardiovascular Research, Southwest Medical University, 646000 Luzhou, Sichuan, China
| |
Collapse
|
36
|
Yao J, Sterling K, Wang Z, Zhang Y, Song W. The role of inflammasomes in human diseases and their potential as therapeutic targets. Signal Transduct Target Ther 2024; 9:10. [PMID: 38177104 PMCID: PMC10766654 DOI: 10.1038/s41392-023-01687-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 09/18/2023] [Accepted: 10/13/2023] [Indexed: 01/06/2024] Open
Abstract
Inflammasomes are large protein complexes that play a major role in sensing inflammatory signals and triggering the innate immune response. Each inflammasome complex has three major components: an upstream sensor molecule that is connected to a downstream effector protein such as caspase-1 through the adapter protein ASC. Inflammasome formation typically occurs in response to infectious agents or cellular damage. The active inflammasome then triggers caspase-1 activation, followed by the secretion of pro-inflammatory cytokines and pyroptotic cell death. Aberrant inflammasome activation and activity contribute to the development of diabetes, cancer, and several cardiovascular and neurodegenerative disorders. As a result, recent research has increasingly focused on investigating the mechanisms that regulate inflammasome assembly and activation, as well as the potential of targeting inflammasomes to treat various diseases. Multiple clinical trials are currently underway to evaluate the therapeutic potential of several distinct inflammasome-targeting therapies. Therefore, understanding how different inflammasomes contribute to disease pathology may have significant implications for developing novel therapeutic strategies. In this article, we provide a summary of the biological and pathological roles of inflammasomes in health and disease. We also highlight key evidence that suggests targeting inflammasomes could be a novel strategy for developing new disease-modifying therapies that may be effective in several conditions.
Collapse
Affiliation(s)
- Jing Yao
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Keenan Sterling
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada
| | - Zhe Wang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yun Zhang
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing, P.R. China.
| | - Weihong Song
- The National Clinical Research Center for Geriatric Disease, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Townsend Family Laboratories, Department of Psychiatry, Brain Research Center, The University of British Columbia, 2255 Wesbrook Mall, Vancouver, BC, V6T 1Z3, Canada.
- Zhejiang Clinical Research Center for Mental Disorders, Key Laboratory of Alzheimer's Disease of Zhejiang Province, School of Mental Health and The Affiliated Kangning Hospital, Institute of Aging, Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
37
|
Li RL, Zhuo CL, Yan X, Li H, Lin L, Li LY, Jiang Q, Zhang D, Wang XM, Liu LL, Huang WJ, Wang YL, Li XY, Mao Y, Chen Y, Liu X, Xu QC, Cai YY, Yang XJ, Chen HY, Wu SS, Jiang W. Irisin attenuates vascular remodeling in hypertensive mice induced by Ang II by suppressing Ca 2+-dependent endoplasmic reticulum stress in VSMCs. Int J Biol Sci 2024; 20:680-700. [PMID: 38169582 PMCID: PMC10758105 DOI: 10.7150/ijbs.84153] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 12/01/2023] [Indexed: 01/05/2024] Open
Abstract
Vascular remodeling plays a vital role in hypertensive diseases and is an important target for hypertension treatment. Irisin, a newly discovered myokine and adipokine, has been found to have beneficial effects on various cardiovascular diseases. However, the pharmacological effect of irisin in antagonizing hypertension-induced vascular remodeling is not well understood. In the present study, we investigated the protection and mechanisms of irisin against hypertension and vascular remodeling induced by angiotensin II (Ang II). Adult male mice of wild-type, FNDC5 (irisin-precursor) knockout, and FNDC5 overexpression were used to develop hypertension by challenging them with Ang II subcutaneously in the back using a microosmotic pump for 4 weeks. Similar to the attenuation of irisin on Ang II-induced VSMCs remodeling, endogenous FNDC5 ablation exacerbated, and exogenous FNDC5 overexpression alleviated Ang II-induced hypertension and vascular remodeling. Aortic RNA sequencing showed that irisin deficiency exacerbated intracellular calcium imbalance and increased vasoconstriction, which was parallel to the deterioration in both ER calcium dysmetabolism and ER stress. FNDC5 overexpression/exogenous irisin supplementation protected VSMCs from Ang II-induced remodeling by improving endoplasmic reticulum (ER) homeostasis. This improvement includes inhibiting Ca2+ release from the ER and promoting the re-absorption of Ca2+ into the ER, thus relieving Ca2+-dependent ER stress. Furthermore, irisin was confirmed to bind to its receptors, αV/β5 integrins, to further activate the AMPK pathway and inhibit the p38 pathway, leading to vasoprotection in Ang II-insulted VSMCs. These results indicate that irisin protects against hypertension and vascular remodeling in Ang II-challenged mice by restoring calcium homeostasis and attenuating ER stress in VSMCs via activating AMPK and suppressing p38 signaling.
Collapse
Affiliation(s)
- Ru-li Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
- Sichuan Key Laboratory of TCM Regulating Metabolic Diseases, Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610072, PR China
| | - Cai-li Zhuo
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin Yan
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - He Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lan Lin
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ling-yu Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Qiying Jiang
- West China College of Preclinical Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Die Zhang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xue-mei Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Lin-ling Liu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
- West China College of Preclinical Medicine and Forensic Medicine, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wen-jing Huang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Ying-ling Wang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xin-yue Li
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yan Mao
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yixin Chen
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiao Liu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Quan-chen Xu
- Department of Biomedical Engineering, Southern University of Science and Technology, Guangdong 518055, PR. China
| | - Yu-yan Cai
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xi-jing Yang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Hong-ying Chen
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Si-si Wu
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Wei Jiang
- Molecular Medicine Research Center, State Key Laboratory of Biotherapy,West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
38
|
Zhang X, Li B, Yan Y, Sun F, Zhang S, Wang M, Liu H. AT1R autoantibody promotes phenotypic transition of smooth muscle cells by activating AT1R-OAS2. Biochem Pharmacol 2024; 219:115977. [PMID: 38092283 DOI: 10.1016/j.bcp.2023.115977] [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: 10/05/2023] [Revised: 12/04/2023] [Accepted: 12/06/2023] [Indexed: 12/26/2023]
Abstract
Phenotypic transition of vascular smooth muscle cells (VSMCs) is an early event in the onset and progression of several cardiovascular diseases. As an important mediator of the renin-angiotensin system (RAS), activation of the angiotensin II type 1 receptor (AT1R) induces phenotypic transition of VSMCs. AT1R autoantibodies (AT1-AAs), which are agonistic autoantibodies of AT1R, have been detected in the sera of patients with a variety of cardiovascular diseases associated with phenotypic transition. However, the effect of AT1-AA on phenotypic transition is currently unknown. In this study, AT1-AA-positive rat model was established by active immunization to detect markers of VSMCs phenotypic transition. The results showed that AT1-AA-positive rats showed phenotypic transition of VSMCs, which was evidenced by the decrease of contractile markers, while the increase of synthetic markers in the thoracic aorta. However, in AT1-AA-positive AT1R knockout rats, the phenotypic transition-related proteins were not altered. In vitro, after stimulating human aortic smooth muscle cells with AT1-AA for 48 h, 2'-5' oligoadenylate synthase 2 (OAS2) was identified as the key differentially expressed gene by RNA sequencing and bioinformatics analysis. Furthermore, high expression of OAS2 was found in aorta of AT1-AA-positive rats; knockdown of OAS2 by siRNA can reverse the phenotypic transition of VSMCs induced by AT1-AA. In summary, this study suggests that AT1-AA can promote phenotypic transition of VSMCs through AT1R-OAS2 pathway, and OAS2 might serve as a potential therapeutic target to prevent pathological phenotypic transition of smooth muscle cells.
Collapse
Affiliation(s)
- Xi Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Bingjie Li
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Yingshuo Yan
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Fei Sun
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China
| | - Suli Zhang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Laboratory for Clinical Medicine, Capital Medical University, Beijing 100069, PR China
| | - Meili Wang
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| | - Huirong Liu
- Department of Physiology & Pathophysiology, School of Basic Medical Sciences, Capital Medical University, Beijing 100069, PR China; Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing 100069, PR China.
| |
Collapse
|
39
|
Sun J, Zhu Q, Yu X, Liang X, Guan H, Zhao H, Yao W. RhoGDI3 at the trans-Golgi network participates in NLRP3 inflammasome activation, VSMC phenotypic modulation, and neointima formation. Atherosclerosis 2023; 387:117391. [PMID: 38029612 DOI: 10.1016/j.atherosclerosis.2023.117391] [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: 03/30/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND AND AIMS The pathological roles and mechanisms of Rho-specific guanine nucleotide dissociation inhibitor 3 (RhoGDI3) in vascular smooth muscle cell (VSMC) phenotypic modulation and neointima formation are currently unknown. This study aimed to investigate how RhoGDI3 regulates the Nod-like receptor protein 3 (NLRP3) inflammasome in platelet-derived growth factor-BB (PDGF-BB)-induced neointima formation. METHODS For in vitro assays, human aortic VSMCs (HA-VSMCs) were transfected with pcDNA3.1-GDI3 and RhoGDI3 siRNA to overexpress and knockdown RhoGDI3, respectively. HA-VSMCs were also treated with an NLRP3 inhibitor (CY-09) or agonist (NSS). Protein transcription and expression, cell proliferation and migration, Golgi morphology, and protein binding and colocalization were measured. For the in vivo assays, balloon injury (BI) rats were injected with recombinant adenovirus carrying RhoGDI3 shRNA. Carotid arterial morphology, protein expression and colocalization, and activation of the NLRP3 inflammasome were measured. RESULTS PDGF-BB treatment induced transcription and expression of RhoGDI3 through PDGF receptor αβ (PDGFRαβ) rather than PDGFRαα or PDGFRββ in HA-VSMCs. RhoGDI3 suppression blocked PDGF-BB-induced VSMC phenotypic transformation. In contrast, RhoGDI3 overexpression further promoted PDGF-BB-induced VSMC dedifferentiation. The in vivo results also confirmed that RhoGDI3 expressed in VSMCs participated in neointima formation and muscle fiber and collagen deposition caused by balloon injury. In addition, PDGF-BB increased binding of RhoGDI3 to NLRP3 and apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) at the trans-Golgi membrane, which depended on the normal Golgi network. However, recruitment of NLRP3 and ASC to the trans-Golgi network after PDGF-BB treatment was independent of RhoGDI3. Moreover, RhoGDI3 knockdown significantly inhibited ASC expression and NLRP3 inflammasome assembly and activation and reduced NLRP3 protein stability in PDGF-BB-treated HA-VSMCs. Inhibiting NLRP3 effectively prevented PDGF-BB-induced VSMC phenotypic modulation, and an NLRP3 agonist reversed the decline in VSMC phenotypic transformation caused by RhoGDI3 knockdown. Furthermore, RhoGDI3 suppression reduced the protein levels and assembly of NLRP3 and ASC, and the activation of the NLRP3 inflammasome in VSMCs in a rat balloon injury model. CONCLUSIONS The results of this study reveal a novel mechanism through which RhoGDI3 regulates VSMC phenotypic modulation and neointima formation by activating the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Jingwen Sun
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong, 226001, China
| | - Qingyu Zhu
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong, 226001, China
| | - Xiaoqiang Yu
- Department of Vascular Surgery, The First People's Hospital of Nantong, Nantong, 226001, China
| | - Xiuying Liang
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong, 226001, China
| | - Haijing Guan
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong, 226001, China
| | - Heyan Zhao
- Medical School, Nantong University, 19 QiXiu Road, Nantong, 226001, China.
| | - Wenjuan Yao
- School of Pharmacy, Nantong University, 19 QiXiu Road, Nantong, 226001, China.
| |
Collapse
|
40
|
Liao X, Han Y, Shen C, Liu J, Wang Y. Targeting the NLRP3 inflammasome for the treatment of hypertensive target organ damage: Role of natural products and formulations. Phytother Res 2023; 37:5622-5638. [PMID: 37690983 DOI: 10.1002/ptr.8009] [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: 04/27/2023] [Revised: 08/10/2023] [Accepted: 08/25/2023] [Indexed: 09/12/2023]
Abstract
BACKGROUND AND AIM Hypertension is a major global health problem that causes target organ damage (TOD) in the heart, brain, kidney, and blood vessels. The mechanisms of hypertensive TOD are not fully understood, and its treatment is challenging. This review provides an overview of the current knowledge on the role of Nod-like receptor pyrin domain containing 3 (NLRP3) inflammasome in hypertensive TOD and the natural products and formulations that inhibit it. METHODS We searched PubMed, Web of Science, Google Scholar, and CNKI for relevant articles using the keywords "hypertension," "target organ damage," "NLRP3 inflammasome," "natural products," and "formulations." We reviewed the effects of the NLRP3 inflammasome on hypertensive TOD in different organs and discussed the natural products and formulations that modulate it. KEY RESULTS In hypertensive TOD, the NLRP3 inflammasome is activated by various stimuli such as oxidative stress and inflammation. Activation of NLRP3 inflammasome leads to the production of pro-inflammatory cytokines that exacerbate tissue damage and dysfunction. Natural products and formulations, including curcumin, resveratrol, triptolide, and allicin, have shown protective effects against hypertensive TOD by inhibiting the NLRP3 inflammasome. CONCLUSIONS AND IMPLICATIONS The NLRP3 inflammasome is a promising therapeutic target in hypertensive TOD. Natural products and formulations that inhibit the NLRP3 inflammasome may provide novel drug candidates or therapies for hypertensive TOD. Further studies are needed to elucidate the molecular mechanisms and optimize the dosages of these natural products and formulations and evaluate their clinical efficacy and safety.
Collapse
Affiliation(s)
- Xiaolin Liao
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yuanshan Han
- Scientific Research Department, The First Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Chuanpu Shen
- Anhui Province Key Laboratory of Major Autoimmune Diseases, Anhui Institute of Innovative Drugs, School of Pharmacy, Anhui Medical University, Hefei, China
- The Key laboratory of Anti-inflammatory and Immune medicines, Ministry of Education, Institute for Liver Diseases of Anhui Medical University Hefei, Hefei, China
| | - Jianjun Liu
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Yuhong Wang
- Institute of Innovation and Applied Research in Chinese Medicine, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
41
|
He L, Zhou W, Wang L, Tang N, Wang Y, Zhong H, Tang Y, Xi D, He F. Murine Cytomegalovirus Infection Induced miR-1929-3p Down-Regulation Promotes the Proliferation and Apoptosis of Vascular Smooth Muscle Cells in Mice by Targeting Endothelin A Receptor and Downstream NLRP3 Activation Pathway. Mol Biotechnol 2023; 65:1954-1967. [PMID: 37022597 DOI: 10.1007/s12033-023-00720-3] [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: 01/06/2023] [Accepted: 03/02/2023] [Indexed: 04/07/2023]
Abstract
Our previous study demonstrated in vivo that mouse cytomegalovirus (MCMV) infection promoted vascular remodeling after downregulation of miR-1929-3p. This study aimed to investigate the role of miR-1929-3p/ETAR/NLRP3 pathway in mouse vascular smooth muscle cells (MOVAS) after MCMV infection. First, PCR was used to detect the success of the infection. Second, MOVAS were transfected with the miR-1929-3p mimic, inhibitor, and ETAR overexpressed adenovirus vector. Cell proliferation was detected using EdU, whereas apoptosis was detected using flow cytometry. The expression of miR-1929-3p and ETAR were detected using qRT-PCR. Western blot detected proteins of cell proliferation, apoptosis, and the NLRP3 inflammasome. Interleukin-1β and interleukin-18 were determined using ELISA. The results revealed that after 48 h, MCMV infection promoted the proliferation of MOVAS when the MOI was 0.01. MCMV infection increased ETAR by downregulating miR-1929-3p. The miR-1929-3p mimic reversed the proliferation and apoptosis, whereas the miR-1929-3p inhibitor promoted this effect. ETAR overexpression further promoted MCMV infection by downregulating miR-1929-3p-mediated proliferation and apoptosis. MCMV infection mediates the downregulation of miR-1929-3p and the upregulation of ETAR, which activates NLRP3 inflammasome. In conclusion, MCMV infection promoted the proliferation of MOVAS, possibly by downregulating miR-1929-3p, promoting the upregulation of the target gene ETAR and activating NLRP3 inflammasome.
Collapse
Affiliation(s)
- Lijuan He
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases and Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, People's Republic of China
| | - Wei Zhou
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases and Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, People's Republic of China
| | - Lamei Wang
- Centre of Medical Functional Experiments, Medical College of Shihezi University, Shihezi, 832002, People's Republic of China
| | - Na Tang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases and Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, People's Republic of China
| | - Yongjia Wang
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases and Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, People's Republic of China
| | - Hua Zhong
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases and Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, People's Republic of China
| | - Yan Tang
- Department of Geriatrics, The First Affiliated Hospital of Medical College of Shihezi University, Shihezi, 832002, People's Republic of China
| | - Dongmei Xi
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases and Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, People's Republic of China.
| | - Fang He
- NHC Key Laboratory of Prevention and Treatment of Central Asia High Incidence Diseases and Key Laboratory of Education Ministry of Xinjiang Endemic and Ethnic Diseases, Medical College of Shihezi University, 59 North 2Nd Road, Shihezi, 832002, Xinjiang, People's Republic of China.
| |
Collapse
|
42
|
Luo Y, Tan Z, Ye Y, Ma X, Yue G. Qiqilian ameliorates vascular endothelial dysfunction by inhibiting NLRP3-ASC inflammasome activation in vivo and in vitro. PHARMACEUTICAL BIOLOGY 2023; 61:815-824. [PMID: 37194678 PMCID: PMC10599261 DOI: 10.1080/13880209.2023.2208617] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 03/25/2023] [Accepted: 04/25/2023] [Indexed: 05/18/2023]
Abstract
CONTEXT Previous studies have highlighted significant therapeutic effects of Qiqilian (QQL) capsule on hypertension in spontaneously hypertensive rats (SHRs); however, its underlying molecular mechanism remains unclear. OBEJECTIVE We investigated the potential mechanism by which QQL improves hypertension-induced vascular endothelial dysfunction (VED). MATERIALS AND METHODS In vivo, SHRs were divided into four groups (20 per group) and were administered gradient doses of QQL (0, 0.3, 0.6, and 1.2 g/kg) for 8 weeks, while Wistar Kyoto rats were used as normal control. The vascular injury extent, IL-1β and IL-18 levels, NLRP3, ASC and caspase-1 contents were examined. In vitro, the effects of QQL-medicated serum on angiotensin II (AngII)-induced inflammatory and autophagy in human umbilical vein endothelial cells (HUVECs) were assessed. RESULT Compared with the SHR group, QQL significantly decreased thickness (125.50 to 105.45 μm) and collagen density (8.61 to 3.20%) of arterial vessels, and reduced serum IL-1β (96.25 to 46.13 pg/mL) and IL-18 (345.01 to 162.63 pg/mL) levels. The NLRP3 and ACS expression in arterial vessels were downregulated (0.21- and 0.16-fold, respectively) in the QQL-HD group compared with the SHR group. In vitro, QQL treatment restored NLRP3 and ASC expression, which was downregulated approximately 2-fold compared with that of AngII-induced HUVECs. Furthermore, QQL decreased LC3II and increased p62 contents (p < 0.05), indicating a reduction in autophagosome accumulation. These effects were inhibited by the autophagy agonist rapamycin and enhanced by the autophagy inhibitor chloroquine. CONCLUSION QQL effectively attenuated endothelial injury and inflammation by inhibiting AngII-induced excessive autophagy, which serves as a potential therapeutic strategy for hypertension.
Collapse
Affiliation(s)
- Yuan Luo
- The First Affiliated Hospital of Guangxi University of Chinese Medicine, Nanning, P.R. China
| | | | - Yun Ye
- No. 923 Hospital of the PLA Joint Logistics Support Force, Nanning, P.R. China
| | - Xiaocong Ma
- Guangxi University of Chinese Medicine, Nanning, P.R. China
| | - Guihua Yue
- Guangxi Internation Zhuang Medicine Hospital to Guangxi University of Chinese Medicine, Nanning, P.R. China
- Guangxi Internation Zhuang Medicine Hospital, Nanning, P.R. China
| |
Collapse
|
43
|
He KY, Lei XY, Wu DH, Zhang L, Li JQ, Li QT, Yin WT, Zhao ZL, Liu H, Xiang XY, Zhu LJ, Cui CY, Wang KK, Wang JH, Lv L, Sun QH, Liu GL, Xu ZX, Jian YP. Akkermansia muciniphila protects the intestine from irradiation-induced injury by secretion of propionic acid. Gut Microbes 2023; 15:2293312. [PMID: 38087436 PMCID: PMC10730217 DOI: 10.1080/19490976.2023.2293312] [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: 02/20/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
Intestinal dysbiosis frequently occurs in abdominal radiotherapy and contributes to irradiation (IR)-induced intestinal damage and inflammation. Akkermansia muciniphila (A. muciniphila) is a recently characterized probiotic, which is critical for maintaining the dynamics of the intestinal mucus layer and preserving intestinal microbiota homeostasis. However, the role of A. muciniphila in the alleviation of radiation enteritis remains unknown. In this study, we reported that the abundance of A. muciniphila was markedly reduced in the intestines of mice exposed to abdominal IR and in the feces of patients who received abdominal radiotherapy. Abundance of A. muciniphila in feces of radiotherapy patients was negatively correlated with the duration of diarrhea in patients. Administration of A. muciniphila substantially mitigated IR-induced intestinal damage and prevented mouse death. Analyzing the metabolic products of A. muciniphila revealed that propionic acid, a short-chain fatty acid secreted by the microbe, mediated the radioprotective effect. We further demonstrated that propionic acid bound to G-protein coupled receptor 43 (GRP43) on the surface of intestinal epithelia and increased histone acetylation and hence enhanced the expression of tight junction proteins occludin and ZO-1 and elevated the level of mucins, leading to enhanced integrity of intestinal epithelial barrier and reduced radiation-induced intestinal damage. Metformin, a first-line agent for the treatment of type II diabetes, promoted intestinal epithelial barrier integrity and reduced radiation intestinal damage through increasing the abundance of A. muciniphila. Together, our results demonstrated that A. muciniphila plays a critical role in the reduction of abdominal IR-induced intestinal damage. Application of probiotics or their regulators, such as metformin, could be an effective treatment for the protection of radiation exposure-damaged intestine.
Collapse
Affiliation(s)
- Kai-Yue He
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Xin-Yuan Lei
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Dan-Hui Wu
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Lei Zhang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Jun-Qi Li
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Qiu-Tong Li
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Wei-Tao Yin
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Zi-Long Zhao
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Huai Liu
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Xiong-Yan Xiang
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Ling-Jun Zhu
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Cui-Yun Cui
- Department of Blood Transfusion, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Ke-Ke Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jin-Hua Wang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, and The Affiliated Cancer Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lin Lv
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Qian-Hui Sun
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Guo-Long Liu
- Department of Medical Oncology, Guangzhou First People’s Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhi-Xiang Xu
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| | - Yong-Ping Jian
- School of Life Sciences, Henan University, Kaifeng, Henan, China
| |
Collapse
|
44
|
Ho LC, Chen YH, Wu TY, Kao LZ, Hung SY, Liou HH, Chen PC, Tsai PJ, Lin HK, Lee YC, Wang HH, Tsai YS. Phosphate burden induces vascular calcification through a NLRP3-caspase-1-mediated pyroptotic pathway. Life Sci 2023; 332:122123. [PMID: 37742736 DOI: 10.1016/j.lfs.2023.122123] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/01/2023] [Accepted: 09/21/2023] [Indexed: 09/26/2023]
Abstract
AIMS The aim of this study is to clarify the role of NLRP3 inflammasome in phosphate burden-induced vascular smooth muscle cell (VSMC) calcification. MAIN METHODS VSMC calcification was induced using a high concentration of inorganic phosphate. After pharmacological inhibition or genetic silencing of the NLRP3 inflammasome, pyroptosis, or potassium efflux, the cells were examined by RT-qPCR, immunofluorescence, and western blotting to identify the NLRP3-mediated pathway for VSMC calcification. KEY FINDINGS Calcified VSMCs with α-smooth muscle actin (α-SMA) disarray presented features of pyroptosis, including caspase-1 maturation, cleaved gasdermin D (GSDMD), and a high supernatant level of lactate dehydrogenase A. Pharmacological inhibitions of caspase-1 and pyroptosis attenuated VSMC calcification, whereas interleukin-1β receptor antagonism did not. Unlike canonical NLRP3 activation, osteogenic VSMCs did not upregulate NLRP3 expression. However, NLRP3 genetic silencing or inhibitions, which targets different domains of the NLRP3 protein, could ameliorate VSMC calcification by aborting caspase-1 and GSDMD activation. Furthermore, potassium efflux through the inward-rectifier potassium channel, and not through the P2X7 receptor, triggered NLRP3 inflammasome activation and VSMC calcification. SIGNIFICANCE In the present study, we identified a potassium efflux-triggered NLRP3-caspase-1-mediated pyroptotic pathway for VSMC calcification that is unique and different from the canonical NLRP3 inflammasome activation. Therefore, targeting this pathway may serve as a novel therapeutic strategy for vascular calcification.
Collapse
Affiliation(s)
- Li-Chun Ho
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan; Division of General Medicine, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan.
| | - Yu-Hsin Chen
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ting-Yun Wu
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Ling-Zhen Kao
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shih-Yuan Hung
- Division of Nephrology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Hung-Hsiang Liou
- Division of Nephrology, Department of Internal Medicine, Hsin-Jen Hospital, New Taipei City, Taiwan
| | - Pei-Chun Chen
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Hui-Kuan Lin
- Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA
| | - Yi-Che Lee
- School of Medicine, College of Medicine, I-Shou University, Kaohsiung, Taiwan
| | - Hsi-Hao Wang
- Division of Nephrology, Department of Internal Medicine, E-Da Hospital, I-Shou University, Kaohsiung, Taiwan
| | - Yau-Sheng Tsai
- Institute of Clinical Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan; Department of Cancer Biology, Wake Forest Baptist Medical Center, Wake Forest University, Winston Salem, NC, USA; Clinical Medicine Research Center, National Cheng Kung University Hospital, Tainan, Taiwan.
| |
Collapse
|
45
|
Zheng X, Zhao D, Jin Y, Liu Y, Liu D. Role of the NLRP3 inflammasome in gynecological disease. Biomed Pharmacother 2023; 166:115393. [PMID: 37660654 DOI: 10.1016/j.biopha.2023.115393] [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: 07/04/2023] [Revised: 08/20/2023] [Accepted: 08/26/2023] [Indexed: 09/05/2023] Open
Abstract
The NLR family pyrin domain containing 3 (NLRP3) inflammasome is involved in the innate immune system and is a three-part macromolecular complex comprising the NLRP3 protein, apoptosis-associated speck-like protein containing a CARD (ASC) and the cysteine protease pro-caspase-1. When the NLRP3 inflammasome is activated, it can produce interleukin (IL)- 1β and IL-18 and eventually lead to inflammatory cell pyroptosis. Related studies have demonstrated that the NLRP3 inflammasome can induce an immune response and is related to the occurrence and development of gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer. NLRP3 inflammasome inhibitors are beneficial for maintaining cellular homeostasis and tissue health and have been found effective in targeting some gynecological diseases. However, excessive inhibitor concentrations have been found to cause adverse effects. Therefore, proper control of NLRP3 inflammasome activity is critical. This paper summarizes the structure and function of the NLRP3 inflammasome and highlights the therapeutic potential of targeting it in gynecological diseases, such as endometriosis, polycystic ovary syndrome and breast cancer The application of NLRP3 inflammasome inhibitors is also discussed.
Collapse
Affiliation(s)
- Xu Zheng
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Dan Zhao
- College of Acupuncture and Massage, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China
| | - Ye Jin
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Yang Liu
- Acupuncture department,Affiliated Hospital of Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| | - Da Liu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, Jilin, China.
| |
Collapse
|
46
|
Zheng F, Ye C, Ge R, Wang Y, Tian XL, Chen Q, Li YH, Zhu GQ, Zhou B. MiR-21-3p in extracellular vesicles from vascular fibroblasts of spontaneously hypertensive rat promotes proliferation and migration of vascular smooth muscle cells. Life Sci 2023; 330:122023. [PMID: 37579834 DOI: 10.1016/j.lfs.2023.122023] [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: 04/27/2023] [Revised: 08/07/2023] [Accepted: 08/10/2023] [Indexed: 08/16/2023]
Abstract
Enhanced proliferation and migration of vascular smooth muscle cells (VSMCs) contributes to vascular remodeling in hypertension. Adventitial fibroblasts (AFs)-derived extracellular vesicles (EVs) modulate vascular remodeling in spontaneously hypertensive rat (SHR). This study shows the important roles of EVs-mediated miR-21-3p transfer in VSMC proliferation and migration and underlying mechanisms in SHR. AFs and VSMCs were obtained from aorta of Wistar-Kyoto rat (WKY) and SHR. EVs were separated from AFs culture with ultracentrifugation method. MiR-21-3p content in the EVs of SHR was increased compared with those of WKY. MiR-21-3p mimic promoted VSMC proliferation and migration of WKY and SHR, while miR-21-3p inhibitor attenuated proliferation and migration only in the VSMCs of SHR. EVs of SHR stimulated VSMC proliferation and migration, which were attenuated by miR-21-3p inhibitor. Sorbin and SH3 domain containing 2 (SORBS2) mRNA and protein levels were reduced in the VSMCs of SHR. MiR-21-3p mimic inhibited, while miR-21-3p inhibitor promoted SORBS2 expressions in the VSMCs of both WKY and SHR. EVs of SHR reduced SORBS2 expression, which was prevented by miR-21-3p inhibitor. EVs of WKY had no significant effect on SORBS2 expressions. SORBS2 overexpression attenuated the roles of miR-21-3p mimic and EVs of SHR in promoting VSMC proliferation and migration of SHR. Overexpression of miR-21-3p in vivo promotes vascular remodeling and hypertension. These results indicate that miR-21-3p in the EVs of SHR promotes VSMC proliferation and migration via negatively regulating SORBS2 expression.
Collapse
Affiliation(s)
- Fen Zheng
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Chao Ye
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Rui Ge
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yu Wang
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, China
| | - Xiao-Lei Tian
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, China
| | - Qi Chen
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Yue-Hua Li
- Department of Pathophysiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Guo-Qing Zhu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Department of Physiology, Nanjing Medical University, Nanjing, Jiangsu 211166, China.
| | - Bing Zhou
- Department of Pathology, Yijishan Hospital, The First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui 241001, China.
| |
Collapse
|
47
|
Jin BR, Lim CY, Kim HJ, Lee M, An HJ. Antioxidant mitoquinone suppresses benign prostatic hyperplasia by regulating the AR-NLRP3 pathway. Redox Biol 2023; 65:102816. [PMID: 37454529 PMCID: PMC10368918 DOI: 10.1016/j.redox.2023.102816] [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: 06/14/2023] [Revised: 06/30/2023] [Accepted: 07/10/2023] [Indexed: 07/18/2023] Open
Abstract
Mitoquinone (MitoQ), a mitochondria-targeted antioxidant, has been used to treat several diseases. The present study aimed to investigate the therapeutic effects of MitoQ in benign prostatic hyperplasia (BPH) models and their underlying molecular mechanisms. In this study, we determined that MitoQ inhibited dihydrotestosterone (DHT)-induced cell proliferation and mitochondrial ROS by inhibiting androgen receptor (AR) and NOD-like receptor family pyrin domain-containing 3 (NLRP3) signaling in prostate epithelial cells. Molecular modeling revealed that DHT may combine with AR and NLRP3, and that MitoQ inhibits both AR and NLRP3. AR and NLRP3 downregulation using siRNA showed the linkage among AR, NLRP3, and MitoQ. MitoQ administration alleviated pathological prostate enlargement and exerted anti-proliferative and antioxidant effects by suppressing the AR and NLRP3 signaling pathways in rats with BPH. Hence, our findings demonstrated that MitoQ is an inhibitor of NLPR3 and AR and a therapeutic agent for BPH treatment.
Collapse
Affiliation(s)
- Bo-Ram Jin
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Chae-Young Lim
- Department of Life Science, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, Republic of Korea
| | - Hyo-Jung Kim
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Minho Lee
- Department of Life Science, Dongguk University-Seoul, 32 Dongguk-ro, Ilsandong-gu, Goyang-si, Gyeonggi-do, 10326, Republic of Korea.
| | - Hyo-Jin An
- Department of Oriental Pharmaceutical Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea; Department of Integrated Drug Development and Natural Products, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
48
|
Lien CF, Lin CS, Shyue SK, Hsieh PS, Chen SJ, Lin YT, Chien S, Tsai MC. Peroxisome proliferator-activated receptor δ improves the features of atherosclerotic plaque vulnerability by regulating smooth muscle cell phenotypic switching. Br J Pharmacol 2023; 180:2085-2101. [PMID: 36942453 DOI: 10.1111/bph.16074] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 03/23/2023] Open
Abstract
BACKGROUND AND PURPOSE Vascular smooth muscle cells (SMCs) undergo phenotypic switching during sustained inflammation, contributing to an unfavourable atherosclerotic plaque phenotype. PPARδ plays an important role in regulating SMC functions; however, its role in atherosclerotic plaque vulnerability remains unclear. Here, we explored the pathological roles of PPARδ in atherosclerotic plaque vulnerability in severe atherosclerosis and elucidated the underlying mechanisms. EXPERIMENTAL APPROACH Plasma levels of PPARδ were measured in patients with acute coronary syndrome (ACS) and stable angina (SA). SMC contractile and synthetic phenotypic markers, endoplasmic reticulum (ER) stress, and features of atherosclerotic plaque vulnerability were analysed for the brachiocephalic artery of apolipoprotein E-knockout (ApoE-/- ) mice, fed a high-cholesterol diet (HCD) and treated with or without the PPARδ agonist GW501516. In vitro, the role of PPARδ was elucidated using human aortic SMCs (HASMCs). KEY RESULTS Patients with ACS had significantly lower plasma PPARδ levels than those with SA. GW501516 reduced atherosclerotic plaque vulnerability, a synthetic SMC phenotype, ER stress markers, and NLRP3 inflammasome expression in HCD-fed ApoE-/- mice. ER stress suppressed PPARδ expression in HASMCs. PPARδ activation inhibited ER stress-induced synthetic phenotype development, ER stress-NLRP3 inflammasome axis activation and matrix metalloproteinase 2 (MMP2) expression in HASMCs. PPARδ inhibited NFκB signalling and alleviated ER stress-induced SMC phenotypic switching. CONCLUSIONS AND IMPLICATIONS Low plasma PPARδ levels may be associated with atherosclerotic plaque vulnerability. Our findings provide new insights into the mechanisms underlying the protective effect of PPARδ on SMC phenotypic switching and improvement the features of atherosclerotic plaque vulnerability.
Collapse
Affiliation(s)
- Chih-Feng Lien
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Chin-Sheng Lin
- Division of Cardiology, Department of Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Song-Kun Shyue
- Cardiovascular Division, Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Po-Shiuan Hsieh
- Graduate Institute of Medical Science, National Defense Medical Center, Taipei, Taiwan
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Sy-Jou Chen
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Yi-Tan Lin
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| | - Shu Chien
- Department of Bioengineering and Medicine, Institute of Engineering in Medicine, University of California San Diego, La Jolla, California, USA
| | - Min-Chien Tsai
- Department of Physiology and Biophysics, Graduate Institute of Physiology, National Defense Medical Center, Taipei, Taiwan
| |
Collapse
|
49
|
Ali F, Wang D, Cheng Y, Wu M, Saleem MZ, Wei L, Xie Y, Yan M, Chu J, Yang Y, Shen A, Peng J. Quercetin attenuates angiotensin II-induced proliferation of vascular smooth muscle cells and p53 pathway activation in vitro and in vivo. Biofactors 2023; 49:956-970. [PMID: 37296538 DOI: 10.1002/biof.1959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 04/23/2023] [Indexed: 06/12/2023]
Abstract
Quercetin is an essential flavonoid mostly found in herbal plants, fruits, and vegetables, which exhibits anti-hypertension properties. However, its pharmacological impact on angiotensin II (Ang II) induced the increase of blood pressure along with in-depth mechanism needs further exploration. The present study pointed out the anti-hypertensive role of quercetin and its comprehensive fundamental mechanisms. Our data showed that quercetin treatment substantially reduced the increase in blood pressure, pulse wave velocity, and aortic thickness of abdominal aorta in Ang II-infused C57BL/6 mice. RNA sequencing revealed that quercetin treatment reversed 464 differentially expressed transcripts in the abdominal aorta of Ang II-infused mice. Moreover, overlapping KEGG-enriched signaling pathways identified multiple common pathways between the comparison of Ang II versus control and Ang II + quercetin versus Ang II. Likewise, these pathways included cell cycle as well as p53 pathways. Transcriptome was further validated by immunohistochemistry, indicating that quercetin treatment significantly decreased the Ang II-induced expression of proliferating cell nuclear antigen (PCNA), cyclin-dependent kinase-4 (CDK4), and cyclin D1, while increased protein expression of p53, and p21 in abdominal aortic tissues of mice. In vitro, quercetin treatment meaningfully decreased the cell viability, arrested cell cycle at G0/G1 phase, and up-regulated the p53 and p21 proteins expression, as well as down-regulated the protein expression of cell cycle-related markers, for example, CDK4, cyclin D1 in Ang II stimulated vascular smooth muscle cells (VSMCs). This study addresses pharmacologic and mechanistic perspectives of quercetin against Ang-II-induced vascular injury and the increase of blood pressure.
Collapse
Affiliation(s)
- Farman Ali
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Di Wang
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Ying Cheng
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Meizhu Wu
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Muhammad Zubair Saleem
- Fujian Key Laboratory of Natural Medicine Pharmacology, Fujian Medical University, Fuzhou, Fujian, China
| | - Lihui Wei
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- Center for Innovation and Transformation of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Yi Xie
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Mengchao Yan
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Jiangfeng Chu
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| | - Yanyan Yang
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- Center for Innovation and Transformation of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Aling Shen
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
- Center for Innovation and Transformation of Science and Technology, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
| | - Jun Peng
- Clinical Research Institute, the Second Affiliated Hospital and Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian, China
- Fujian Collaborative Innovation Center for Integrative Medicine in Prevention and Treatment of Major Chronic Cardiovascular Diseases, Fuzhou, Fujian, China
| |
Collapse
|
50
|
Aboukhater D, Morad B, Nasrallah N, Nasser SA, Sahebkar A, Kobeissy F, Boudaka A, Eid AH. Inflammation and hypertension: Underlying mechanisms and emerging understandings. J Cell Physiol 2023; 238:1148-1159. [PMID: 37039489 DOI: 10.1002/jcp.31019] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 03/24/2023] [Indexed: 04/12/2023]
Abstract
Hypertension remains a major contributor to cardiovascular disease (CVD), a leading cause of global death. One of the major insults that drive increased blood pressure is inflammation. While it is the body's defensive response against some homeostatic imbalances, inflammation, when dysregulated, can be very deleterious. In this review, we highlight and discuss the causative relationship between inflammation and hypertension. We critically discuss how the interplay between inflammation and reactive oxygen species evokes endothelial damage and dysfunction, ultimately leading to narrowing and stiffness of blood vessels. This, along with phenotypic switching of the vascular smooth muscle cells and the abnormal increase in extracellular matrix deposition further exacerbates arterial stiffness and noncompliance. We also discuss how hyperhomocysteinemia and microRNA act as links between inflammation and hypertension. The premises we discuss suggest that the blue-sky scenarios for targeting the underlying mechanisms of hypertension necessitate further research.
Collapse
Affiliation(s)
- Diana Aboukhater
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Bassel Morad
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nadim Nasrallah
- Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | | | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Firas Kobeissy
- Department of Neurobiology and Neuroscience, Morehouse School of Medicine, Atlanta, Georgia, USA
| | - Ammar Boudaka
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| | - Ali H Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
| |
Collapse
|