1
|
Shilenok V, Kobzeva K, Bushueva O. "SERBP1 (Hero45) is a Novel Link with Ischemic Heart Disease Risk: Associations with Coronary Arteries Occlusion, Blood Coagulation and Lipid Profile". Cell Biochem Biophys 2025:10.1007/s12013-025-01736-z. [PMID: 40175693 DOI: 10.1007/s12013-025-01736-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2025] [Indexed: 04/04/2025]
Abstract
Ischemic heart disease (IHD), stemming from coronary atherosclerosis, involves pathological processes in which chaperone proteins play an essential role. SERBP1 (Hero45), an RNA-binding protein, has recently been ascribed to the newly discovered class of Hero proteins with chaperone-like activity, making it particularly relevant in atherosclerosis-related diseases. In this study, 2164 subjects (836 IHD patients and 1328 controls) were genotyped for five common single nucleotide polymorphisms (SNPs) of SERBP1 using probe-based PCR. Here, we report that SNPs of SERBP1 are associated with reduced risk of left coronary artery atherosclerosis: rs4655707 (effect allele [EA] T, OR = 0.63, 95% CI 0.43-0.93, p = 0.02), (EA C, OR = 0.63, 95% CI 0.42-0.95, p = 0.02), rs12561767 (EA G, OR = 0.65, 95% CI 0.45-0.96, p = 0.03), rs6702742 (EA A, OR = 0.63, 95% CI 0.43-0.94, p = 0.02). Additionally, SERBP1 loci are linked to lower coronary artery stenosis (rs1058074), improved blood lipid profiles (rs1058074), and favorable blood coagulation parameters (rs4655707, rs6702742, rs1058074, rs12561767). Together, our study is the first to provide evidence that SERBP1 is involved in lipid metabolism and coagulation regulation, modulating IHD risk.
Collapse
Affiliation(s)
- Vladislav Shilenok
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
- Cardiology Department with the intensive care unit, Kursk Emergency Hospital, Kursk, Russia
| | - Ksenia Kobzeva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia.
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russia.
| |
Collapse
|
2
|
Heo KS, Phan LP, Le NTT, Jin Y. Mechanistic insights and emerging therapeutic strategies targeting endothelial dysfunction in cardiovascular diseases. Arch Pharm Res 2025; 48:305-332. [PMID: 40301174 DOI: 10.1007/s12272-025-01542-4] [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: 02/19/2025] [Accepted: 04/08/2025] [Indexed: 05/01/2025]
Abstract
Endothelial dysfunction plays a pivotal role in the pathogenesis of various cardiovascular diseases (CVDs), including atherosclerosis, hypertension, heart failure, stroke, and peripheral artery disease. It disrupts vascular homeostasis, leading to reduced nitric oxide (NO) bioavailability, increased oxidative stress, and chronic inflammation, all of which collectively drive vascular damage, atherosclerotic plaque formation, and thrombosis. Additionally, shear stress-induced alterations in blood flow patterns, particularly disturbed flow (d-flow), aggravate endothelial dysfunction. Furthermore, the endothelial-to-mesenchymal transition (EndMT), a process in which endothelial cells acquire mesenchymal-like properties, contributes to vascular remodeling and accelerates CVD progression.This review explores the significant role of epigenetic mechanisms, such as DNA methylation, histone modifications, and noncoding RNAs (ncRNAs), which serve as critical regulators of endothelial function in response to shear stress in endothelial dysfunction and the development of atherosclerosis. Furthermore, we discuss the pivotal role of endothelial dysfunction in cardiovascular and metabolic diseases, emphasizing the need for innovative therapeutic strategies beyond conventional treatments. In particular, we highlight the endothelial-protective mechanisms of emerging pharmacological agents, including proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitors, glucagon-like peptide-1 receptor agonists (GLP-1RAs), and sodium-glucose cotransporter 2 (SGLT2) inhibitors, along with supporting clinical evidence demonstrating their efficacy in improving endothelial function and reducing cardiovascular risk.
Collapse
Affiliation(s)
- Kyung-Sun Heo
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| | - Lan Phuong Phan
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Nhi Thi Thao Le
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Yujin Jin
- Department of Pharmacology, Chungnam National University, College of Pharmacy, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| |
Collapse
|
3
|
Cohen-Hagai K, Kuchuk E, Matalon ST, Benchetrit S, Zitman-Gal T. Exosomes Derived from Dialysis Patients' Serum Enhance Endothelial-Mesenchymal Transition and Calcification in Endothelial Cells. KIDNEY360 2025; 6:351-360. [PMID: 39899380 PMCID: PMC11970854 DOI: 10.34067/kid.0000000678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 12/06/2024] [Indexed: 02/05/2025]
Abstract
Key Points Vascular calcification (VC) is an independent risk factor of cardiovascular disease in patients with ESKD. We found that exosomes derived from serum of dialysis patients with VC induced endothelial–mesenchymal transition and thus contribute to calcification. The complex interplay between exosomes from dialysis patients with VC and endothelial cells highlights the critical need for therapeutic strategies. Background Vascular calcification (VC) is prevalent among patients with ESKD. Exosomes, small extracellular vesicles actively secreted by cells, contain proteins, nucleic acids, lipids, and other bioactive substances and are considered major mediators of cell–cell interactions. Endothelial–mesenchymal transition (EndMT) has been observed in a variety of pathological conditions, such as abnormal shear stress, vascular damage, and chronic inflammation. The aim of this research was to assess the effects of serum-derived exosomes from ESKD patients with VC on the induction of EndMT in endothelial cells (ECs) and their potential role in accelerating VC. Methods Twenty patients on hemodialysis with VC and ten healthy volunteers were recruited. Cardiac and brain VCs were assessed among patients with ESKD treated with dialysis. Serum samples were obtained at dialysis initiation for exosome isolation. Human umbilical vein ECs were treated with 100 µ g/ml exosomes for 24–96 hours. At the end of incubation, cells were collected for mRNA and protein analysis. Results Exosomes isolated from dialysis patients with VC induced EndMT in human umbilical vein ECs. After 24 hours, endothelial markers CD31 and vascular endothelial-cadherin were decreased (31% and 51%, respectively; P < 0.001) and the mesenchymal proteins Vimentin and N -cadherin were increased (283% and 156%, respectively; P < 0.001), compared with healthy exosomes. After 96 hours of incubation, expression of genes essential for osteoblast differentiation, including the bone morphogenetic genes (BMP2, BMPR2, BMP4, and BMP9), and the transcription factor RUNX2 were significantly elevated. Conclusions Exosomes derived from the serum of dialysis patients with VC induced EndMT and contributed to calcification. The vicious cycle highlighted the intricate interplay between exosomes, ECs, and VC, emphasizing the critical necessity for therapeutic strategies to disrupt this pathway and mitigate calcification advancement.
Collapse
Affiliation(s)
- Keren Cohen-Hagai
- Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Eran Kuchuk
- Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
| | - Shelly Tartakover Matalon
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
- Autoimmune Research Laboratory, Meir Medical Center, Kfar Saba, Israel
| | - Sydney Benchetrit
- Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tali Zitman-Gal
- Department of Nephrology and Hypertension, Meir Medical Center, Kfar Saba, Israel
- Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Moldovan D. The Severity of Carotid Calcifications, but Not Fibroblast Growth Factor 23, Is Associated with Mortality in Hemodialysis: A Single Center Experience. Diseases 2025; 13:73. [PMID: 40136613 PMCID: PMC11941726 DOI: 10.3390/diseases13030073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 02/21/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND The study goal was to assess the mortality effect of carotid vascular calcifications (VC), of fibroblast growth factor 23 (FGF-23), mineral markers, and comorbidities in hemodialysis (HD) patients. METHODS The influence of carotid VC severity, FGF-23, laboratory markers, clinical features, and comorbidities on mortality was analyzed in a cohort of 88 HD patients. The follow-up period lasted 8 years. The cut-off value for carotid VC was 4 for all-cause and cardiovascular mortality. RESULTS Carotid VC, diabetes, low serum albumin, high serum C-reactive protein (CRP), and the presence of cardiovascular diseases are associated with all-cause and cardiovascular mortality. Carotid VC score over 4 was an independent predictor of all-cause and cardiovascular mortality, along with diabetes, low albumin, and high CRP. FGF-23 was not found to be predictable for the study outcomes. CONCLUSIONS The study documented in a cohort of patients prevalent in chronic HD that carotid VC predicts all-cause and cardiovascular mortality at 8 years and improves risk stratification, but FGF-23 is not associated with mortality. Other risk factors for all-cause and cardiovascular mortality were diabetes, inflammation, and malnutrition. However, future efforts are needed to assess whether a risk-based approach, including VC screening, improves survival.
Collapse
Affiliation(s)
- Diana Moldovan
- Department of Nephrology, "Iuliu Hatieganu" University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| |
Collapse
|
5
|
Pan L, Xia W, Song J, Zhang S. Association between oxidative balance scores and severe abdominal aortic calcification in American adults: National health and nutrition examination survey. Nutr Metab Cardiovasc Dis 2025; 35:103697. [PMID: 39174433 DOI: 10.1016/j.numecd.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND AND AIM Abdominal aortic calcification (AAC) is a key predictor of cardiovascular diseases (CVDs). The Oxidative Balance Score (OBS) served as a tool to evaluate the systemic status of oxidative stress. However, evidence on the link between OBS and severe abdominal aortic calcification (SAAC) is currently inadequate. This study aims to establish this correlation in the US adult population, contributing valuable insights to the understanding of cardiovascular health. METHODS AND RESULTS In our study with 2745 participants from the 2013-2014 National Health and Nutrition Examination Survey (NHANES), we analyzed both OBS and AAC score data. Logistic regression and smooth curve fitting were used to investigate the relationship between OBS and SAAC. The overall prevalence of severe abdominal aortic calcification disease was 9.1%. Multivariable logistic regression revealed that higher oxidative balance scores were associated with a lower risk of SAAC. After adjusting for potential confounders (model III), for every 1-point increase in oxidative balance scores, the odds of SAAC decreased by 3% [OR = 0.97, 95% CI= (0.95,0.99), P = 0.03]. The dose-response relationship demonstrated a negative correlation between oxidative balance scores and SAAC (p for nonlinear = 0.368). CONCLUSIONS This study reveals a negative association between oxidative balance scores and severe abdominal aortic calcification in US adults. The implications of these findings merit careful consideration and should be taken into account in the formulation of clinical guidelines and updates.
Collapse
Affiliation(s)
- Liya Pan
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Wujie Xia
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Jing Song
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Suqin Zhang
- Department of Cardiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
6
|
Qian C, Dong G, Yang C, Zheng W, Zhong C, Shen Q, Lu Y, Zhao Y. Broadening horizons: molecular mechanisms and disease implications of endothelial-to-mesenchymal transition. Cell Commun Signal 2025; 23:16. [PMID: 39789529 PMCID: PMC11720945 DOI: 10.1186/s12964-025-02028-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/03/2025] [Indexed: 01/12/2025] Open
Abstract
Endothelial-mesenchymal transition (EndMT) is defined as an important process of cellular differentiation by which endothelial cells (ECs) are prone to lose their characteristics and transform into mesenchymal cells. During EndMT, reduced expression of endothelial adhesion molecules disrupts intercellular adhesion, triggering cytoskeletal reorganization and mesenchymal transition. Numerous studies have proved that EndMT is a multifaceted biological event driven primarily by cytokines such as TGF-β, TNF-α, and IL-1β, alongside signaling pathways like WNT, Smad, MEK-ERK, and Notch. Nevertheless, the exact roles of EndMT in complicated diseases have not been comprehensively reviewed. In this review, we summarize the predominant molecular regulatory mechanisms and signaling pathways that contribute to the development of EndMT, as well as highlight the contributions of a series of imperative non-coding RNAs in curbing the initiation of EndMT. Furthermore, we discuss the significant impact of EndMT on worsening vasculature-related diseases, including cancer, cardiovascular diseases, atherosclerosis, pulmonary vascular diseases, diabetes-associated fibrotic conditions, and cerebral cavernous malformation, providing the implications that targeting EndMT holds promise as a therapeutic strategy to mitigate disease progression.
Collapse
Affiliation(s)
- Cheng Qian
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Guanglu Dong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chunmei Yang
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Weiwei Zheng
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Chongjin Zhong
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Qiuhong Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yang Zhao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
7
|
You N, Liu G, Yu M, Chen W, Fei X, Sun T, Han M, Qin Z, Wei Z, Wang D. Reconceptualizing Endothelial-to-mesenchymal transition in atherosclerosis: Signaling pathways and prospective targeting strategies. J Adv Res 2025:S2090-1232(24)00627-1. [PMID: 39756576 DOI: 10.1016/j.jare.2024.12.049] [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: 09/15/2024] [Revised: 12/27/2024] [Accepted: 12/28/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND The modification of endothelial cells (ECs) biological function under pathogenic conditions leads to the expression of mesenchymal stromal cells (MSCs) markers, defined as endothelial-to-mesenchymal transition (EndMT). Invisible in onset and slow in progression, atherosclerosis (AS) is a potential contributor to various atherosclerotic cardiovascular diseases (ASCVD). By triggering AS, EndMT, the "initiator" of AS, induces the progression of ASCVD such as coronary atherosclerotic heart disease (CHD) and ischemic cerebrovascular disease (ICD), with serious clinical complications such as myocardial infarction (MI) and stroke. In-depth research of the pathomechanisms of EndMT and identification of potential targeted therapeutic strategies hold considerable research value for the prevention and treatment of ASCVD-associated with delayed EndMT. Although previous studies have progressively unraveled the complexity of EndMT and its pathogenicity triggered by alterations in vascular microenvironmental factors, systematic descriptions of the most recent pathogenic roles of EndMT in the progression of AS, targeted therapeutic strategies, and their future research directions are scarce. AIM OF REVIEW We aim to provide new researchers with comprehensive knowledge of EndMT in AS. We exhaustively review the latest research advancements in the field and provide a theoretical basis for investigating EndMT, a biological process with sophisticated mechanisms. KEY SCIENTIFIC CONCEPTS OF REVIEW This review summarized that altered hemodynamics with microenvironmental crosstalk consisting of inflammatory responses or glycolysis, oxidative stress, lactate or acetyl-CoA (Ac-CoA), fatty acid oxidation (FAO), intracellular iron overload, and transcription factors, including ELK1 and STAT3, modulate the EndMT and affect AS progression. In addition, we provide new paradigms for the development of promising therapeutic agents against these disease-causing processes and indicate promising directions and challenges that need to be addressed to elucidate the EndMT process.
Collapse
Affiliation(s)
- Nanlin You
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China.
| | - Guohao Liu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengchen Yu
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Wenbo Chen
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Xiaoyao Fei
- Key Laboratory for Experimental Teratology of Ministry of Education, Shandong Key Laboratory of Infection and Immunology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Tao Sun
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Mengtao Han
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhen Qin
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Zhaosheng Wei
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Donghai Wang
- Department of Neurosurgery, Qilu Hospital of Shandong University, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Institute of Brain and Brain-Inspired Science, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China; Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, Shandong 253032, China.
| |
Collapse
|
8
|
Cabiati M, Vozzi F, Ceccherini E, Guiducci L, Persiani E, Gisone I, Sgalippa A, Cecchettini A, Del Ry S. Exploring Bone Morphogenetic Protein-2 and -4 mRNA Expression and Their Receptor Assessment in a Dynamic In Vitro Model of Vascular Calcification. Cells 2024; 13:2091. [PMID: 39768183 PMCID: PMC11674890 DOI: 10.3390/cells13242091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/10/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Vascular calcification (VC) is a dynamic, tightly regulated process driven by cellular activity and resembling the mechanisms of bone formation, with specific molecules playing pivotal roles in its progression. We aimed to investigate the involvement of the bone morphogenic proteins (BMP-2, BMP-4, BMPR-1a/1b, and BMPR-2) system in this process. Our study used an advanced in vitro model that simulates the biological environment of the vascular wall, assessing the ability of a phosphate mixture to induce the osteoblastic switch in human coronary artery smooth muscle cells (HCASMCs). METHODS HCASMCs were grown in mono- and co-culture with human coronary artery endothelial cells (HCAECs) in a double-flow bioreactor (LiveBox2 and IVTech), allowing static and dynamic conditions through a peristaltic pump. The VC was stimulated by incubation in a calcifying medium for 7 days. A BMP system Real-Time PCR was performed at the end of each experiment. RESULTS In monocultures, BMP-2 expression increased in calcified HCASMCs in static (p = 0.01) and dynamic conditions. BMP-4 and the biological receptors were expressed in all the experimental settings, increasing mainly in dynamic flow conditions. In co-cultures, we observed a marked increase in BMP-2 and BMP-4, BMPR-1a (p = 0.04 and p = 0.01, respectively), and BMPR-2 (p = 0.001) in the calcifying setting mostly in dynamic conditions. CONCLUSIONS The increase in BMP-2/4 in co-culture suggests that these genes might promote the switch towards an osteogenic-like phenotype, data also supported by the rise of both BMPR-1a and BMPR-2. Thus, our findings provide insights into the mechanisms by which dynamic co-culture modulates the BMP system activation in an environment mimicking in vivo VC's cellular and mechanical characteristics.
Collapse
Affiliation(s)
- Manuela Cabiati
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Federico Vozzi
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Elisa Ceccherini
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Letizia Guiducci
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Elisa Persiani
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Ilaria Gisone
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
| | - Agnese Sgalippa
- Health Science Interdisciplinary Center, Sant’Anna School of Advanced Studies, 56100 Pisa, Italy;
| | - Antonella Cecchettini
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy
| | - Silvia Del Ry
- Institute of Clinical Physiology IFC-CNR, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy; (F.V.); (E.C.); (L.G.); (E.P.); (I.G.); (S.D.R.)
- Department of Clinical and Experimental Medicine, University of Pisa, 56100 Pisa, Italy
| |
Collapse
|
9
|
Wits M, Haarmans N, Sanchez-Duffhues G, Goumans MJ. TGF-β receptor-specific NanoBRET Target Engagement in living cells for high-throughput kinase inhibitor screens. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100196. [PMID: 39542424 DOI: 10.1016/j.slasd.2024.100196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/17/2024]
Abstract
Targeting transforming growth factor-β (TGF-β) receptors is a promising pharmacological approach to normalize aberrant signaling in genetic and non-genetic TGF-β associated diseases including fibrosis, cancer, cardiovascular and musculoskeletal disorders. To identify novel TGF-β receptor kinase inhibitors, methods like in vitro kinase assays, western blot or transcriptional reporter assays are often used for screening purposes. While these methods may have certain advantages, the lack of integration of key features such as receptor specificity, high-throughput capability, and cellular context resemblance remains a major disadvantage. This deficiency could ultimately hinder the translation of study outcomes into later (clinical) stages of drug development. In this study, we introduce an adjusted and optimized live cell NanoBRET Target Engagement (TE)-based method to identify TGF-β receptor specific kinase inhibitors. This comprehensive toolkit contains various TGF-β type I and type II receptors, with corresponding nanoBRET tracers, and disease-related cell lines, including novel non-commercially available materials. The nanoBRET capacity and kinase inhibitory window can be significantly enhanced for functional measurements when stable expression cell lines and substantially low tracer concentrations are used. In addition, this system can be tailored to study TGF-β associated genetic disorders and possibly be used to screen for disease-specific therapeutics. Therefore, the use of this optimized, live cell, antibody-independent nanoBRET Target Engagement assay is highly encouraged for future high-throughput compound screens targeting TGF-β/BMP receptors.
Collapse
Affiliation(s)
- Marius Wits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, the Netherlands
| | - Nicole Haarmans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, the Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, the Netherlands; Nanomaterials and Nanotechnology Research Center (CINN), Spanish National Research Council (CSIC), Health Research Institute of Asturias (ISPA), 33011 Oviedo, Asturias, Spain.
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC, Leiden, the Netherlands.
| |
Collapse
|
10
|
Lin A, Miano JM, Fisher EA, Misra A. Chronic inflammation and vascular cell plasticity in atherosclerosis. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1408-1423. [PMID: 39653823 DOI: 10.1038/s44161-024-00569-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 10/10/2024] [Indexed: 12/13/2024]
Abstract
Vascular smooth muscle cells, endothelial cells and macrophages undergo phenotypic conversions throughout atherosclerosis progression, both as a consequence of chronic inflammation and as subsequent drivers of it. The inflammatory hypothesis of atherosclerosis has been catapulted to the forefront of cardiovascular research as clinical trials have shown that anti-inflammatory therapy reduces adverse cardiovascular events. However, no current therapies have been specifically designed to target the phenotype of plaque cells. Fate mapping has revealed that plaque cells convert to detrimental and beneficial cell phenotypes during atherosclerosis, with cumulative evidence highlighting that vascular cell plasticity is intimately linked with plaque inflammation, ultimately impacting lesion stability. Here we review vascular cell plasticity during atherosclerosis in the context of the chronic inflammatory plaque microenvironment. We highlight the need to better understand how plaque cells behave during therapeutic intervention. We then propose modulating plaque cell phenotype as an unexplored therapeutic paradigm in the clinical setting.
Collapse
Affiliation(s)
- Alexander Lin
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia
- School of Biomedical Engineering, Faculty of Engineering, The University of Sydney, Sydney, New South Wales, Australia
| | - Joseph M Miano
- Vascular Biology Center, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Edward A Fisher
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY, USA
- Cardiovascular Research Center, New York University Grossman School of Medicine, New York, NY, USA
| | - Ashish Misra
- Atherosclerosis and Vascular Remodelling Group, Heart Research Institute, Sydney, New South Wales, Australia.
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia.
| |
Collapse
|
11
|
Liu B, Cai Z, Wang Y, Liu X, Zhang B, Zheng Q, Li J, Li C, Cui Y, Lv P, Yang D. Transglutaminase 2 regulates endothelial cell calcification via IL-6-mediated autophagy. Front Pharmacol 2024; 15:1393534. [PMID: 39654623 PMCID: PMC11625581 DOI: 10.3389/fphar.2024.1393534] [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: 02/29/2024] [Accepted: 10/31/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction Endothelial cell (EC) calcification is an important marker of atherosclerotic calcification. ECs play a critical role not only in atherogenesis but also in intimal calcification, as they have been postulated to serve as a source of osteoprogenitor cells that initiate this process. While the role of transglutaminase 2 (TG2) in cellular differentiation, survival, apoptosis, autophagy, and cell adhesion is well established, the mechanism underlying the TG2-mediated regulation of EC calcification is yet to be fully elucidated. Methods The TG2 gene was overexpressed or silenced by using siRNA and recombinant adenovirus. RT-PCR and WB were used to analyze the relative expression of target genes and proteins. 5-BP method analyzed TG2 activity. mCherry-eGFP-LC3 adenovirus and transmission electron microscopy analyzed EC autophagy level. Calcium concentrations were measured by using a calcium colorimetric assay kit. Alizarin red S staining assay analyzed EC calcification level. Elisa analyzed IL-6 level. Establishing EC calcification model by using a calcification medium (CM). Results Our findings demonstrated that CM increased TG2 activity and expression, which activated the NF-κB signaling pathway, and induced IL-6 autocrine signaling in ECs. Furthermore, IL-6 activated the JAK2/STAT3 signaling pathway to suppress cell autophagy and promoted ECs calcification. Discussion ECs are not only critical for atherogenesis but also believed to be a source of osteoprogenitor cells that initiate intimal calcification. Previous research has shown that TG2 plays an important role in the development of VC, but the mechanism by which it exerts this effect is not yet fully understood. Our results demonstrated that TG2 forms complexes with NF-κB components inhibition of autophagy promoted endothelial cell calcification through EndMT. Therefore, our research investigated the molecular mechanism of EC calcification, which can provide new insights into the pathogenesis of atherosclerosis.
Collapse
Affiliation(s)
- Bo Liu
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Zhiyuan Cai
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yan Wang
- The First Department of Ocular Fundus Diseases, Zhengzhou Second Hospital, Zhengzhou, Henan, China
| | - Xinye Liu
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Zhang
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Qian Zheng
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Jingye Li
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Cien Li
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuanbo Cui
- Translational Medical Center, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Pengju Lv
- Department of clinical laboratory, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| | - Dongwei Yang
- Department of Cardiology, Zhengzhou Central Hospital, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
12
|
Hamczyk MR, Nevado RM, Gonzalo P, Andrés-Manzano MJ, Nogales P, Quesada V, Rosado A, Torroja C, Sánchez-Cabo F, Dopazo A, Bentzon JF, López-Otín C, Andrés V. Endothelial-to-Mesenchymal Transition Contributes to Accelerated Atherosclerosis in Hutchinson-Gilford Progeria Syndrome. Circulation 2024; 150:1612-1630. [PMID: 39206565 DOI: 10.1161/circulationaha.123.065768] [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: 05/29/2023] [Accepted: 07/26/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Atherosclerosis is the main medical problem in Hutchinson-Gilford progeria syndrome, a rare premature aging disorder caused by the mutant lamin-A protein progerin. Recently, we found that limiting progerin expression to vascular smooth muscle cells (VSMCs) is sufficient to hasten atherosclerosis and death in Apoe-deficient mice. However, the impact of progerin-driven VSMC defects on endothelial cells (ECs) remained unclear. METHODS Apoe- or Ldlr-deficient C57BL/6J mice with ubiquitous, VSMC-, EC- or myeloid-specific progerin expression fed a normal or high-fat diet were used to study endothelial phenotype during Hutchinson-Gilford progeria syndrome-associated atherosclerosis. Endothelial permeability to low-density lipoproteins was assessed by intravenous injection of fluorescently labeled human low-density lipoprotein and confocal microscopy analysis of the aorta. Leukocyte recruitment to the aortic wall was evaluated by en face immunofluorescence. Endothelial-to-mesenchymal transition (EndMT) was assessed by quantitative polymerase chain reaction and RNA sequencing in the aortic intima and by immunofluorescence in aortic root sections. TGFβ (transforming growth factor β) signaling was analyzed by multiplex immunoassay in serum, by Western blot in the aorta, and by immunofluorescence in aortic root sections. The therapeutic benefit of TGFβ1/SMAD3 pathway inhibition was evaluated in mice by intraperitoneal injection of SIS3 (specific inhibitor of SMAD3), and vascular phenotype was assessed by Oil Red O staining, histology, and immunofluorescence in the aorta and the aortic root. RESULTS Both ubiquitous and VSMC-specific progerin expression in Apoe-null mice provoked alterations in aortic ECs, including increased permeability to low-density lipoprotein and leukocyte recruitment. Atherosclerotic lesions in these progeroid mouse models, but not in EC- and myeloid-specific progeria models, contained abundant cells combining endothelial and mesenchymal features, indicating extensive EndMT triggered by dysfunctional VSMCs. Accordingly, the intima of ubiquitous and VSMC-specific progeroid models at the onset of atherosclerosis presented increased expression of EndMT-linked genes, especially those specific to fibroblasts and extracellular matrix. Aorta in both models showed activation of the TGFβ1/SMAD3 pathway, a major trigger of EndMT, and treatment of VSMC-specific progeroid mice with SIS3 alleviated the aortic phenotype. CONCLUSIONS Progerin-induced VSMC alterations promote EC dysfunction and EndMT through TGFβ1/SMAD3, identifying this process as a candidate target for Hutchinson-Gilford progeria syndrome treatment. These findings also provide insight into the complex role of EndMT during atherogenesis.
Collapse
Affiliation(s)
- Magda R Hamczyk
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
| | - Rosa M Nevado
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Pilar Gonzalo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - María J Andrés-Manzano
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Paula Nogales
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Víctor Quesada
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
- Centro de Investigación Biomédica en Red de Cáncer, Spain (V.Q.)
| | - Aránzazu Rosado
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Carlos Torroja
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Ana Dopazo
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| | - Jacob F Bentzon
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
- Department of Clinical Medicine, Aarhus University, Denmark (J.F.B.)
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Instituto Universitario de Oncología, Universidad de Oviedo, Spain (M.R.H., V.Q., C.L.-O.)
- Facultad de Ciencias de la Vida y la Naturaleza, Universidad Nebrija, Madrid, Spain (C.L.-O.)
- Centre de Recherche des Cordeliers, Université de Paris Cité, Sorbonne Université, INSERM U1138, France (C.L.-O.)
| | - Vicente Andrés
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares, Spain (M.R.H., R.M.N., P.G., M.J.A.-M., A.D., V.A.)
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain (R.M.N., P.G., M.J.A.-M., P.N., A.R., C.T., F.S.-C., A.D., J.F.B., V.A.)
| |
Collapse
|
13
|
Tobal R, Potjewijd J, de Vries F, van Doorn DPC, Jaminon A, Bittner R, Akbulut C, van Empel V, Heeringa P, Damoiseaux J, Schurgers L, van Paassen P. Dephosphorylated uncarboxylated Matrix-Gla-Protein as candidate biomarker for immune-mediated vascular remodeling and prognosis in pulmonary hypertension. Sci Rep 2024; 14:26633. [PMID: 39496657 PMCID: PMC11535036 DOI: 10.1038/s41598-024-77000-w] [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: 04/26/2024] [Accepted: 10/18/2024] [Indexed: 11/06/2024] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease characterized by pulmonary vascular remodeling. Since dephosphorylated-uncarboxylated Matrix Gla-Protein (dp-ucMGP) is associated with cardiovascular mortality in systemic sclerosis, a disease associated with PAH, and immune-system involvement in PAH is increasingly recognized, we investigated the relationship between dp-ucMGP, vascular remodeling and soluble immune-checkpoint proteins in PAH. This prospective cohort study included patients with idiopathic (I)PAH, connective tissue disease (CTD)-PAH, chronic thrombo-embolic PH (CTEPH) and CTD patients without PAH. Patients with IPAH and CTD-PAH were stratified by clinical signs of immune-mediated inflammatory disease (IMID). We measured dp-ucMGP plasma levels, soluble immune-checkpoint proteins (sICPs), and vascular smooth muscle cell (iVSMC) calcification. We found elevated dp-ucMGP levels in all PAH subtypes and CTD patients compared to healthy controls. PAH patients showed increased iVSMC calcification, but no direct correlation was found with dp-ucMGP. IMID-PAH patients had higher dp-ucMGP levels than non-IMID PAH patients. dp-ucMGP correlated with several sICPs in both IPAH and CTD patients; multiple sICPs were elevated in IMID PAH patients. High dp-ucMGP levels in IPAH patients were associated with worse survival. Our findings suggest dp-ucMGP as a potential biomarker of immune-mediated vascular remodeling in PAH. Hence, dp-ucMGP, could help identify PAH patients who might benefit from immunosuppressive therapies.
Collapse
Affiliation(s)
- R Tobal
- Department of Internal Medicine, Division of Clinical and Experimental Immunology, Maastricht University Medical Center, Maastricht, The Netherlands.
| | - J Potjewijd
- Department of Internal Medicine, Division of Clinical and Experimental Immunology, Maastricht University Medical Center, Maastricht, The Netherlands.
| | - F de Vries
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - D P C van Doorn
- Department of Internal Medicine, Division of Clinical and Experimental Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - A Jaminon
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - R Bittner
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - C Akbulut
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - V van Empel
- Department of Cardiology, Maastricht University Medical Center, Maastricht, The Netherlands
| | - P Heeringa
- Department of Pathology and Medical Biology, University Medical Center Groningen, Groningen, The Netherlands
| | - J Damoiseaux
- Central Diagnostic Laboratory, Maastricht University Medical Center, Maastricht, The Netherlands
| | - L Schurgers
- Department of Biochemistry, CARIM, Maastricht University, Maastricht, The Netherlands
| | - P van Paassen
- Department of Internal Medicine, Division of Clinical and Experimental Immunology, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
14
|
Tóth A, Balogh E, Jeney V. In Vitro Models of Cardiovascular Calcification. Biomedicines 2024; 12:2155. [PMID: 39335668 PMCID: PMC11429067 DOI: 10.3390/biomedicines12092155] [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: 08/30/2024] [Revised: 09/15/2024] [Accepted: 09/21/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular calcification, characterized by hydroxyapatite deposition in the arterial wall and heart valves, is associated with high cardiovascular morbidity and mortality. Cardiovascular calcification is a hallmark of aging but is frequently seen in association with chronic diseases, such as chronic kidney disease (CKD), diabetes, dyslipidemia, and hypertension in the younger population as well. Currently, there is no therapeutic approach to prevent or cure cardiovascular calcification. The pathophysiology of cardiovascular calcification is highly complex and involves osteogenic differentiation of various cell types of the cardiovascular system, such as vascular smooth muscle cells and valve interstitial cells. In vitro cellular and ex vivo tissue culture models are simple and useful tools in cardiovascular calcification research. These models contributed largely to the discoveries of the numerous calcification inducers, inhibitors, and molecular mechanisms. In this review, we provide an overview of the in vitro cell culture and the ex vivo tissue culture models applied in the research of cardiovascular calcification.
Collapse
Affiliation(s)
- Andrea Tóth
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Enikő Balogh
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| | - Viktória Jeney
- MTA-DE Lendület Vascular Pathophysiology Research Group, Research Centre for Molecular Medicine, Faculty of Medicine, University of Debrecen, 4032 Debrecen, Hungary
| |
Collapse
|
15
|
Tian J, You H, Ding J, Shi D, Long C, li Y, Luo Z, He X. Platelets could be key regulators of epithelial/endothelial-to- mesenchymal transition in atherosclerosis and wound healing. Med Hypotheses 2024; 189:111397. [DOI: 10.1016/j.mehy.2024.111397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
|
16
|
Ning L, Zanella S, Tomov ML, Amoli MS, Jin L, Hwang B, Saadeh M, Chen H, Neelakantan S, Dasi LP, Avazmohammadi R, Mahmoudi M, Bauser‐Heaton HD, Serpooshan V. Targeted Rapamycin Delivery via Magnetic Nanoparticles to Address Stenosis in a 3D Bioprinted in Vitro Model of Pulmonary Veins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400476. [PMID: 38696618 PMCID: PMC11234432 DOI: 10.1002/advs.202400476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/09/2024] [Indexed: 05/04/2024]
Abstract
Vascular cell overgrowth and lumen size reduction in pulmonary vein stenosis (PVS) can result in elevated PV pressure, pulmonary hypertension, cardiac failure, and death. Administration of chemotherapies such as rapamycin have shown promise by inhibiting the vascular cell proliferation; yet clinical success is limited due to complications such as restenosis and off-target effects. The lack of in vitro models to recapitulate the complex pathophysiology of PVS has hindered the identification of disease mechanisms and therapies. This study integrated 3D bioprinting, functional nanoparticles, and perfusion bioreactors to develop a novel in vitro model of PVS. Bioprinted bifurcated PV constructs are seeded with endothelial cells (ECs) and perfused, demonstrating the formation of a uniform and viable endothelium. Computational modeling identified the bifurcation point at high risk of EC overgrowth. Application of an external magnetic field enabled targeting of the rapamycin-loaded superparamagnetic iron oxide nanoparticles at the bifurcation site, leading to a significant reduction in EC proliferation with no adverse side effects. These results establish a 3D bioprinted in vitro model to study PV homeostasis and diseases, offering the potential for increased throughput, tunability, and patient specificity, to test new or more effective therapies for PVS and other vascular diseases.
Collapse
Affiliation(s)
- Liqun Ning
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of Mechanical EngineeringCleveland State UniversityClevelandOH44115USA
| | - Stefano Zanella
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Martin L. Tomov
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Mehdi Salar Amoli
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Linqi Jin
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Boeun Hwang
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Maher Saadeh
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Huang Chen
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Sunder Neelakantan
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lakshmi Prasad Dasi
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
| | - Reza Avazmohammadi
- Department of Biomedical EngineeringTexas A&M UniversityCollege StationTX77843USA
- J. Mike Walker ’66 Department of Mechanical EngineeringTexas A&M UniversityCollege StationTX77840USA
| | - Morteza Mahmoudi
- Department of Radiology and Precision Health ProgramMichigan State UniversityEast LandingMI48824USA
| | - Holly D. Bauser‐Heaton
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
- Sibley Heart Center at Children's Healthcare of AtlantaAtlantaGA30322USA
| | - Vahid Serpooshan
- Wallace H. Coulter Department of Biomedical EngineeringEmory University School of Medicine and Georgia Institute of TechnologyAtlantaGA30322USA
- Department of PediatricsEmory University School of MedicineAtlantaGA30322USA
- Children's Healthcare of AtlantaAtlantaGA30322USA
| |
Collapse
|
17
|
Donovan MK, Abdel-Rahman AA. Ethanol-induced lung and cardiac right ventricular inflammation and remodeling underlie progression to pulmonary arterial hypertension. ALCOHOL, CLINICAL & EXPERIMENTAL RESEARCH 2024; 48:1250-1260. [PMID: 38710650 PMCID: PMC11236493 DOI: 10.1111/acer.15341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/05/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Current research on ethanol-induced cardiovascular anomalies has focused on left ventricular (LV) function and blood pressure. To extend this area of research, we sought to determine whether ethanol-induced alterations in the structure and function of the right cardiac ventricle (RV) and pulmonary artery (PA) lead to pulmonary arterial hypertension (PAH). METHODS Two groups of male Sprague-Dawley rats received a balanced liquid diet containing 5% ethanol (w/v) or a pair-fed isocaloric liquid diet for 8 weeks. Weekly echocardiography was conducted to evaluate cardiopulmonary function, and lung and RV tissues were collected for ex vivo histological and molecular studies. RESULTS The ethanol-treated rats exhibited: (1) Elevated mean pulmonary arterial pressure and decreased pulmonary artery acceleration time/ejection time; (2) Pulmonary vascular remodeling comprising intrapulmonary artery medial layer thickening; and (3) RV hypertrophy along with increased RV/LV + septum, RV diameter, RV cardiomyocyte cross-sectional area, and LV mass/body weight ratio. These responses were associated with increased lung and RV pro-inflammatory markers, endothelin-1 (ET-1), TNF-α, and IL-6 levels and higher ET-1, ET-1 type A/B receptor ratio, and downregulation of the cytoprotective protein, bone morphogenetic protein receptor 2 (BMPR2), in the lungs. CONCLUSION These findings show that moderate ethanol-induced cardiopulmonary changes underlie progression to PAH via an upregulated proinflammatory ET1-TNFα-IL6 pathway and suppression of the anti-inflammatory BMPR2.
Collapse
Affiliation(s)
- Mary Katherine Donovan
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| | - Abdel A Abdel-Rahman
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, North Carolina, USA
| |
Collapse
|
18
|
Horvat Mercnik M, Schliefsteiner C, Sanchez-Duffhues G, Wadsack C. TGFβ signalling: a nexus between inflammation, placental health and preeclampsia throughout pregnancy. Hum Reprod Update 2024; 30:442-471. [PMID: 38519450 PMCID: PMC11215164 DOI: 10.1093/humupd/dmae007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/16/2024] [Indexed: 03/25/2024] Open
Abstract
BACKGROUND The placenta is a unique and pivotal organ in reproduction, controlling crucial growth and cell differentiation processes that ensure a successful pregnancy. Placental development is a tightly regulated and dynamic process, in which the transforming growth factor beta (TGFβ) superfamily plays a central role. This family of pleiotropic growth factors is heavily involved in regulating various aspects of reproductive biology, particularly in trophoblast differentiation during the first trimester of pregnancy. TGFβ signalling precisely regulates trophoblast invasion and the cell transition from cytotrophoblasts to extravillous trophoblasts, which is an epithelial-to-mesenchymal transition-like process. Later in pregnancy, TGFβ signalling ensures proper vascularization and angiogenesis in placental endothelial cells. Beyond its role in trophoblasts and endothelial cells, TGFβ signalling contributes to the polarization and function of placental and decidual macrophages by promoting maternal tolerance of the semi-allogeneic foetus. Disturbances in early placental development have been associated with several pregnancy complications, including preeclampsia (PE) which is one of the severe complications. Emerging evidence suggests that TGFβ is involved in the pathogenesis of PE, thereby offering a potential target for intervention in the human placenta. OBJECTIVE AND RATIONALE This comprehensive review aims to explore and elucidate the roles of the major members of the TGFβ superfamily, including TGFβs, bone morphogenetic proteins (BMPs), activins, inhibins, nodals, and growth differentiation factors (GDFs), in the context of placental development and function. The review focusses on their interactions within the major cell types of the placenta, namely trophoblasts, endothelial cells, and immune cells, in both normal pregnancies and pregnancies complicated by PE throughout pregnancy. SEARCH METHODS A literature search was carried out using PubMed and Google Scholar, searching terms: 'TGF signalling preeclampsia', 'pregnancy TGF signalling', 'preeclampsia tgfβ', 'preeclampsia bmp', 'preeclampsia gdf', 'preeclampsia activin', 'endoglin preeclampsia', 'endoglin pregnancy', 'tgfβ signalling pregnancy', 'bmp signalling pregnancy', 'gdf signalling pregnancy', 'activin signalling pregnancy', 'Hofbauer cell tgfβ signalling', 'placental macrophages tgfβ', 'endothelial cells tgfβ', 'endothelium tgfβ signalling', 'trophoblast invasion tgfβ signalling', 'trophoblast invasion Smad', 'trophoblast invasion bmp', 'trophoblast invasion tgfβ', 'tgfβ preeclampsia', 'tgfβ placental development', 'TGFβ placental function', 'endothelial dysfunction preeclampsia tgfβ signalling', 'vascular remodelling placenta TGFβ', 'inflammation pregnancy tgfβ', 'immune response pregnancy tgfβ', 'immune tolerance pregnancy tgfβ', 'TGFβ pregnancy NK cells', 'bmp pregnancy NK cells', 'bmp pregnancy tregs', 'tgfβ pregnancy tregs', 'TGFβ placenta NK cells', 'TGFβ placenta tregs', 'NK cells preeclampsia', 'Tregs preeclampsia'. Only articles published in English until 2023 were used. OUTCOMES A comprehensive understanding of TGFβ signalling and its role in regulating interconnected cell functions of the main placental cell types provides valuable insights into the processes essential for successful placental development and growth of the foetus during pregnancy. By orchestrating trophoblast invasion, vascularization, immune tolerance, and tissue remodelling, TGFβ ligands contribute to the proper functioning of a healthy maternal-foetal interface. However, dysregulation of TGFβ signalling has been implicated in the pathogenesis of PE, where the shallow trophoblast invasion, defective vascular remodelling, decreased uteroplacental perfusion, and endothelial cell and immune dysfunction observed in PE, are all affected by an altered TGFβ signalling. WIDER IMPLICATIONS The dysregulation of TGFβ signalling in PE has important implications for research and clinical practice. Further investigation is required to understand the underlying mechanisms, including the role of different ligands and their regulation under pathophysiological conditions, in order to discover new therapeutic targets. Distinguishing between clinically manifested subtypes of PE and studying TGFβ signalling in different placental cell types holistically is an important first step. To put this knowledge into practice, pre-clinical animal models combined with new technologies are needed. This may also lead to improved human research models and identify potential therapeutic targets, ultimately improving outcomes for affected pregnancies and reducing the burden of PE.
Collapse
Affiliation(s)
| | | | - Gonzalo Sanchez-Duffhues
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Tissue-Specific BMP Signalling ISPA-HUCA, Oviedo, Spain
| | - Christian Wadsack
- Department of Obstetrics and Gynaecology, Medical University of Graz, Graz, Austria
- BioTechMed-Graz, Graz, Austria
| |
Collapse
|
19
|
Laan SNJ, de Boer S, Dirven RJ, van Moort I, Kuipers TB, Mei H, Bierings R, Eikenboom J. Transcriptional and functional profiling identifies inflammation and endothelial-to-mesenchymal transition as potential drivers for phenotypic heterogeneity within a cohort of endothelial colony forming cells. J Thromb Haemost 2024; 22:2027-2038. [PMID: 38574861 DOI: 10.1016/j.jtha.2024.03.018] [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: 10/19/2023] [Revised: 03/14/2024] [Accepted: 03/24/2024] [Indexed: 04/06/2024]
Abstract
BACKGROUND Endothelial colony-forming cells (ECFCs) derived from patients can be used to investigate pathogenic mechanisms of vascular diseases like von Willebrand disease. Considerable phenotypic heterogeneity has been observed between ECFC clones derived from healthy donors. This heterogeneity needs to be well understood in order to use ECFCs as endothelial models for disease. OBJECTIVES Therefore, we aimed to determine phenotypic and gene expression differences between control ECFCs. METHODS A total of 34 ECFC clones derived from 16 healthy controls were analyzed. The transcriptome of a selection of ECFC clones (n = 15) was analyzed by bulk RNA sequencing and gene set enrichment analysis. Gene expression was measured in all ECFC clones by quantitative polymerase chain reaction. Phenotypic profiling was performed and migration speed of the ECFCs was measured using confocal microscopy, followed by automated quantification of cell morphometrics and migration speed. RESULTS Through hierarchical clustering of RNA expression profiles, we could distinguish 2 major clusters within the ECFC cohort. Major differences were associated with proliferation and migration in cluster 1 and inflammation and endothelial-to-mesenchymal transition in cluster 2. Phenotypic profiling showed significantly more and smaller ECFCs in cluster 1, which contained more and longer Weibel-Palade bodies. Migration speed in cluster 1 was also significantly higher. CONCLUSION We observed a range of different RNA expression patterns between ECFC clones, mostly associated with inflammation and clear differences in Weibel-Palade body count and structure. We developed a quantitative polymerase chain reaction panel that can be used for the characterization of ECFC clones, which is essential for the correct analysis of pathogenic mechanisms in vascular disorders.
Collapse
Affiliation(s)
- Sebastiaan N J Laan
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands; Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands. https://twitter.com/laan_bas
| | - Suzan de Boer
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Richard J Dirven
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands
| | - Iris van Moort
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Thomas B Kuipers
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Hailiang Mei
- Sequencing Analysis Support Core, Department of Biomedical Data Sciences, Leiden University Medical Centre, Leiden, the Netherlands
| | - Ruben Bierings
- Department of Hematology, Erasmus University Medical Centre, Rotterdam, the Netherlands
| | - Jeroen Eikenboom
- Department of Internal Medicine, Division of Thrombosis and Hemostasis, Leiden University Medical Centre, Leiden, the Netherlands.
| |
Collapse
|
20
|
Chen W, Kim SY, Lee A, Kim YJ, Chang C, Ton-That H, Kim R, Kim S, Park NH. hTERT Peptide Fragment GV1001 Prevents the Development of Porphyromonas gingivalis-Induced Periodontal Disease and Systemic Disorders in ApoE-Deficient Mice. Int J Mol Sci 2024; 25:6126. [PMID: 38892314 PMCID: PMC11172542 DOI: 10.3390/ijms25116126] [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: 04/09/2024] [Revised: 05/23/2024] [Accepted: 05/28/2024] [Indexed: 06/21/2024] Open
Abstract
GV1001, an anticancer vaccine, exhibits other biological functions, including anti-inflammatory and antioxidant activity. It also suppresses the development of ligature-induced periodontitis in mice. Porphyromonas gingivalis (Pg), a major human oral bacterium implicated in the development of periodontitis, is associated with various systemic disorders, such as atherosclerosis and Alzheimer's disease (AD). This study aimed to explore the protective effects of GV1001 against Pg-induced periodontal disease, atherosclerosis, and AD-like conditions in Apolipoprotein (ApoE)-deficient mice. GV1001 effectively mitigated the development of Pg-induced periodontal disease, atherosclerosis, and AD-like conditions by counteracting Pg-induced local and systemic inflammation, partly by inhibiting the accumulation of Pg DNA aggregates, Pg lipopolysaccharides (LPS), and gingipains in the gingival tissue, arterial wall, and brain. GV1001 attenuated the development of atherosclerosis by inhibiting vascular inflammation, lipid deposition in the arterial wall, endothelial to mesenchymal cell transition (EndMT), the expression of Cluster of Differentiation 47 (CD47) from arterial smooth muscle cells, and the formation of foam cells in mice with Pg-induced periodontal disease. GV1001 also suppressed the accumulation of AD biomarkers in the brains of mice with periodontal disease. Overall, these findings suggest that GV1001 holds promise as a preventive agent in the development of atherosclerosis and AD-like conditions associated with periodontal disease.
Collapse
Affiliation(s)
- Wei Chen
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 714 Tiverton Ave., Los Angeles, CA 90095, USA; (W.C.); (S.Y.K.); (A.L.); (Y.-J.K.); (R.K.)
| | - Sharon Y. Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 714 Tiverton Ave., Los Angeles, CA 90095, USA; (W.C.); (S.Y.K.); (A.L.); (Y.-J.K.); (R.K.)
| | - Alicia Lee
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 714 Tiverton Ave., Los Angeles, CA 90095, USA; (W.C.); (S.Y.K.); (A.L.); (Y.-J.K.); (R.K.)
| | - Yun-Jeong Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 714 Tiverton Ave., Los Angeles, CA 90095, USA; (W.C.); (S.Y.K.); (A.L.); (Y.-J.K.); (R.K.)
| | - Chungyu Chang
- Section of Oral Biology, UCLA School of Dentistry, 714 Tiverton Avenue, Los Angeles, CA 90095, USA; (C.C.); (H.T.-T.)
| | - Hung Ton-That
- Section of Oral Biology, UCLA School of Dentistry, 714 Tiverton Avenue, Los Angeles, CA 90095, USA; (C.C.); (H.T.-T.)
| | - Reuben Kim
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 714 Tiverton Ave., Los Angeles, CA 90095, USA; (W.C.); (S.Y.K.); (A.L.); (Y.-J.K.); (R.K.)
- UCLA Jonsson Comprehensive Cancer Center, 10833 Le Conte Ave., Los Angeles, CA 90095, USA
| | - Sangjae Kim
- Teloid Inc., 920 Westholme Avenue, Los Angeles, CA 90024, USA;
| | - No-Hee Park
- The Shapiro Family Laboratory of Viral Oncology and Aging Research, UCLA School of Dentistry, 714 Tiverton Ave., Los Angeles, CA 90095, USA; (W.C.); (S.Y.K.); (A.L.); (Y.-J.K.); (R.K.)
- Teloid Inc., 920 Westholme Avenue, Los Angeles, CA 90024, USA;
- Department of Medicine, David Geffen School of Medicine at UCLA, 10833 Le Conte Ave., Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Wang Q, Peng F, Yang J, Chen X, Peng Z, Zhang M, Tang D, Liu J, Zhao H. MicroRNAs regulate the vicious cycle of vascular calcification-osteoporosis in postmenopausal women. Mol Biol Rep 2024; 51:622. [PMID: 38709309 DOI: 10.1007/s11033-024-09550-1] [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: 12/29/2023] [Accepted: 04/12/2024] [Indexed: 05/07/2024]
Abstract
Menopause is a normal physiological process accompanied by changes in various physiological states. The incidence of vascular calcification (VC) increases each year after menopause and is closely related to osteoporosis (OP). Although many studies have investigated the links between VC and OP, the interaction mechanism of the two under conditions of estrogen loss remains unclear. MicroRNAs (miRNAs), which are involved in epigenetic modification, play a critical role in estrogen-mediated mineralization. In the past several decades, miRNAs have been identified as biomarkers or therapeutic targets in diseases. Thus, we hypothesize that these small molecules can provide new diagnostic and therapeutic approaches. In this review, we summarize the close interactions between VC and OP and the role of miRNAs in their interplay.
Collapse
Affiliation(s)
- Qian Wang
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Fei Peng
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Jing Yang
- Changsha Central Hospital Affiliated to University of South China, Changsha, Hunan, China
| | - Xiaolong Chen
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Zhaojie Peng
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Minyi Zhang
- The University of South China, Hengyang, Hunan, China
| | - Deqiu Tang
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China
| | - Jianghua Liu
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China.
| | - Heng Zhao
- Department of Radiology, The First Affiliated Hospital of The University of South China, Hengyang, Hunan, China.
| |
Collapse
|
22
|
He L, Li X, Shen E, He YM. Association between Abdominal Aortic Calcification and Coronary Heart Disease in Essential Hypertension: A Cross-Sectional Study from the 2013-2014 National Health and Nutrition Examination Survey. J Cardiovasc Dev Dis 2024; 11:143. [PMID: 38786965 PMCID: PMC11122146 DOI: 10.3390/jcdd11050143] [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: 03/24/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND This study aimed to investigate the association between abdominal aortic calcification (AAC) and coronary heart disease (CHD) in essential hypertension (EH). METHODS This study included patients diagnosed with EH during the 2013-2014 NHANES survey cycle. The study cohort was categorized into the following four groups based on their AAC-24 score: no AAC (0); mild AAC (1-4); moderate AAC (5-15); and severe AAC (16-24). Logistic regression models were used to assess the association between AAC and CHD. Restricted cubic spline curves (RCS) were used to explore possible nonlinear relationships between AAC and CHD. RESULTS The prevalence of CHD was found to be higher in the moderate AAC and severe AAC groups than in the group without AAC (40.1% versus 30.9%, 47.7% versus 30.9%). On a continuous scale, the fully adjusted model showed a 7% increase in the risk of CHD prevalence per score increase in AAC [OR (95% CI) = 1.07 (1.03-1.11)]. On a categorical scale, the fully adjusted model showed the risk of CHD prevalence in EH patients with moderate AAC and severe AAC was 2.06 (95%CI, 1.23-3.45) and 2.18 (1.09-5.25) times higher than that in patients without AAC, respectively. The RCS curve suggested a dose-response linear relationship between AAC and CHD. CONCLUSION These findings highlight that in patients with EH, a higher severity of AAC is associated with a higher risk of CHD prevalence.
Collapse
Affiliation(s)
- Lan He
- Division of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Ave., Gusu District, Suzhou 215006, China; (L.H.); (X.L.)
| | - Xu Li
- Division of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Ave., Gusu District, Suzhou 215006, China; (L.H.); (X.L.)
| | - E Shen
- Department of Ultrasound, Shanghai Chest Hospital, School of Medicine, Shanghai Jiao Tong University, 241 Huaihai West Road, Xuhui District, Shanghai 200030, China
| | - Yong-Ming He
- Division of Cardiology, The First Affiliated Hospital of Soochow University, 188 Shizi Ave., Gusu District, Suzhou 215006, China; (L.H.); (X.L.)
| |
Collapse
|
23
|
Yao J, Chen Y, Huang Y, Sun X, Shi X. The role of cardiac microenvironment in cardiovascular diseases: implications for therapy. Hum Cell 2024; 37:607-624. [PMID: 38498133 DOI: 10.1007/s13577-024-01052-3] [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: 11/29/2023] [Accepted: 02/25/2024] [Indexed: 03/20/2024]
Abstract
Due to aging populations and changes in lifestyle, cardiovascular diseases including cardiomyopathy, hypertension, and atherosclerosis, are the leading causes of death worldwide. The heart is a complicated organ composed of multicellular types, including cardiomyocytes, fibroblasts, endothelial cells, vascular smooth muscle cells, and immune cells. Cellular specialization and complex interplay between different cell types are crucial for the cardiac tissue homeostasis and coordinated function of the heart. Mounting studies have demonstrated that dysfunctional cells and disordered cardiac microenvironment are closely associated with the pathogenesis of various cardiovascular diseases. In this paper, we discuss the composition and the homeostasis of cardiac tissues, and focus on the role of cardiac environment and underlying molecular mechanisms in various cardiovascular diseases. Besides, we elucidate the novel treatment for cardiovascular diseases, including stem cell therapy and targeted therapy. Clarification of these issues may provide novel insights into the prevention and potential targets for cardiovascular diseases.
Collapse
Affiliation(s)
- Jiayu Yao
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuejun Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yuqing Huang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| | - Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| |
Collapse
|
24
|
He Q, Li J, Tao C, Zeng C, Liu C, Zheng Z, Mou S, Liu W, Zhang B, Yu X, Zhai Y, Wang J, Zhang Q, Zhang Y, Zhang D, Zhao J, Ge P. High glutamine increases stroke risk by inducing the endothelial-to-mesenchymal transition in moyamoya disease. MedComm (Beijing) 2024; 5:e525. [PMID: 38628905 PMCID: PMC11018113 DOI: 10.1002/mco2.525] [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: 07/09/2023] [Revised: 02/04/2024] [Accepted: 02/26/2024] [Indexed: 04/19/2024] Open
Abstract
At present, there is limited research on the mechanisms underlying moyamoya disease (MMD). Herein, we aimed to determine the role of glutamine in MMD pathogenesis, and 360 adult patients were prospectively enrolled. Human brain microvascular endothelial cells (HBMECs) were subjected to Integrin Subunit Beta 4 (ITGB4) overexpression or knockdown and atorvastatin. We assessed factors associated with various signaling pathways in the context of the endothelial-to-mesenchymal transition (EndMT), and the expression level of related proteins was validated in the superficial temporal arteries of patients. We found glutamine levels were positively associated with a greater risk of stroke (OR = 1.599, p = 0.022). After treatment with glutamine, HBMECs exhibited enhanced proliferation, migration, and EndMT, all reversed by ITGB4 knockdown. In ITGB4-transfected HBMECs, the MAPK-ERK-TGF-β/BMP pathway was activated, with Smad4 knockdown reversing the EndMT. Furthermore, atorvastatin suppressed the EndMT by inhibiting Smad1/5 phosphorylation and promoting Smad4 ubiquitination in ITGB4-transfected HBMECs. We also found the protein level of ITGB4 was upregulated in the superficial temporal arteries of patients with MMD. In conclusion, our study suggests that glutamine may be an independent risk factor for hemorrhage or infarction in patients with MMD and targeting ITGB4 could potentially be therapeutic approaches for MMD.
Collapse
Affiliation(s)
- Qiheng He
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Junsheng Li
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chuming Tao
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chaofan Zeng
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Chenglong Liu
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Zhiyao Zheng
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- Research Unit of Accurate Diagnosis, Treatment, and Translational Medicine of Brain TumorsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of NeurosurgeryPeking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Siqi Mou
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Wei Liu
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Bojian Zhang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Xiaofan Yu
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Yuanren Zhai
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Jia Wang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- 3D Printing Center in Clinical NeuroscienceChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Qian Zhang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Yan Zhang
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| | - Dong Zhang
- Department of NeurosurgeryBeijing HospitalBeijingChina
| | - Jizong Zhao
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
- 3D Printing Center in Clinical NeuroscienceChina National Clinical Research Center for Neurological DiseasesBeijingChina
| | - Peicong Ge
- Department of NeurosurgeryBeijing Tiantan Hospital, Capital Medical UniversityBeijingChina
| |
Collapse
|
25
|
Adelus ML, Ding J, Tran BT, Conklin AC, Golebiewski AK, Stolze LK, Whalen MB, Cusanovich DA, Romanoski CE. Single-cell 'omic profiles of human aortic endothelial cells in vitro and human atherosclerotic lesions ex vivo reveal heterogeneity of endothelial subtype and response to activating perturbations. eLife 2024; 12:RP91729. [PMID: 38578680 PMCID: PMC10997331 DOI: 10.7554/elife.91729] [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: 04/06/2024] Open
Abstract
Heterogeneity in endothelial cell (EC) sub-phenotypes is becoming increasingly appreciated in atherosclerosis progression. Still, studies quantifying EC heterogeneity across whole transcriptomes and epigenomes in both in vitro and in vivo models are lacking. Multiomic profiling concurrently measuring transcriptomes and accessible chromatin in the same single cells was performed on six distinct primary cultures of human aortic ECs (HAECs) exposed to activating environments characteristic of the atherosclerotic microenvironment in vitro. Meta-analysis of single-cell transcriptomes across 17 human ex vivo arterial specimens was performed and two computational approaches quantitatively evaluated the similarity in molecular profiles between heterogeneous in vitro and ex vivo cell profiles. HAEC cultures were reproducibly populated by four major clusters with distinct pathway enrichment profiles and modest heterogeneous responses: EC1-angiogenic, EC2-proliferative, EC3-activated/mesenchymal-like, and EC4-mesenchymal. Quantitative comparisons between in vitro and ex vivo transcriptomes confirmed EC1 and EC2 as most canonically EC-like, and EC4 as most mesenchymal with minimal effects elicited by siERG and IL1B. Lastly, accessible chromatin regions unique to EC2 and EC4 were most enriched for coronary artery disease (CAD)-associated single-nucleotide polymorphisms from Genome Wide Association Studies (GWAS), suggesting that these cell phenotypes harbor CAD-modulating mechanisms. Primary EC cultures contain markedly heterogeneous cell subtypes defined by their molecular profiles. Surprisingly, the perturbations used here only modestly shifted cells between subpopulations, suggesting relatively stable molecular phenotypes in culture. Identifying consistently heterogeneous EC subpopulations between in vitro and ex vivo models should pave the way for improving in vitro systems while enabling the mechanisms governing heterogeneous cell state decisions.
Collapse
Affiliation(s)
- Maria L Adelus
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
- The Clinical Translational Sciences Graduate Program, The University of ArizonaTucsonUnited States
| | - Jiacheng Ding
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Binh T Tran
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Austin C Conklin
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Anna K Golebiewski
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Lindsey K Stolze
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Michael B Whalen
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
| | - Darren A Cusanovich
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
- Asthma and Airway Disease Research Center, The University of ArizonaTucsonUnited States
| | - Casey E Romanoski
- The Department of Cellular and Molecular Medicine, The University of ArizonaTucsonUnited States
- The Clinical Translational Sciences Graduate Program, The University of ArizonaTucsonUnited States
- Asthma and Airway Disease Research Center, The University of ArizonaTucsonUnited States
| |
Collapse
|
26
|
Fan Y, Pei J, Qin Y, Du H, Qu X, Li W, Huang B, Tan J, Liu Y, Li G, Ke M, Xu Y, Zhu C. Construction of tissue-engineered vascular grafts with enhanced patency by integrating heparin, cell-adhesive peptide, and carbon monoxide nanogenerators into acellular blood vessels. Bioact Mater 2024; 34:221-236. [PMID: 38235307 PMCID: PMC10792202 DOI: 10.1016/j.bioactmat.2023.12.015] [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: 10/18/2023] [Revised: 12/11/2023] [Accepted: 12/21/2023] [Indexed: 01/19/2024] Open
Abstract
Small-diameter tissue-engineered vascular grafts (sdTEVGs) have garnered significant attention as a potential treatment modality for vascular bypass grafting and replacement therapy. However, the intimal hyperplasia and thrombosis are two major complications that impair graft patency during transplantation. To address this issue, we fabricated the covalent-organic framework (COF)-based carbon monoxide (CO) nanogenerator-and co-immobilized with LXW-7 peptide and heparin to establish a multifunctional surface on TEVGs constructed from acellular blood vessels for preventing thrombosis and stenosis. The cell-adhesive peptide LXW-7 could capture endothelial-forming cells (EFCs) to promote endothelialization, while the antithrombotic molecule heparin prevented thrombus formation. The reactive oxygen species (ROS)-triggered CO release suppressed the adhesion and activation of macrophages, leading to the reduction of ROS and inflammatory factors. As a result, the endothelial-to-mesenchymal transition (EndMT) triggered by inflammation was restricted, facilitating the maintenance of the homeostasis of the neo-endothelium and preventing pathological remodeling in TEVGs. When transplanted in vivo, these vascular grafts exhibited negligible intimal hyperplasia and remained patent for 3 months. This achievement provided a novel approach for constructing antithrombotic and anti-hyperplastic TEVGs.
Collapse
Affiliation(s)
- Yonghong Fan
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Laboratory of Basic Medicine, The General Hospital of Western Theater Command, Chengdu, 610083, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
| | - Juan Pei
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Yinhua Qin
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Huifang Du
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Xiaohang Qu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Wenya Li
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Boyue Huang
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Ju Tan
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Yong Liu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Gang Li
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
| | - Ming Ke
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
| | - Youqian Xu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
| | - Chuhong Zhu
- Department of Anatomy, Engineering Research Center for Organ Intelligent Biological Manufacturing of Chongqing, Key Lab for Biomechanics and Tissue Engineering of Chongqing, Third Military Medical University, Chongqing, 400038, China
- Engineering Research Center of Tissue and Organ Regeneration and Manufacturing, Ministry of Education, Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Chongqing, 400038, China
- Department of Plastic and Aesthetic Surgery, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| |
Collapse
|
27
|
Hall IF, Kishta F, Xu Y, Baker AH, Kovacic JC. Endothelial to mesenchymal transition: at the axis of cardiovascular health and disease. Cardiovasc Res 2024; 120:223-236. [PMID: 38385523 PMCID: PMC10939465 DOI: 10.1093/cvr/cvae021] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 08/01/2023] [Accepted: 08/25/2023] [Indexed: 02/23/2024] Open
Abstract
Endothelial cells (ECs) line the luminal surface of blood vessels and play a major role in vascular (patho)-physiology by acting as a barrier, sensing circulating factors and intrinsic/extrinsic signals. ECs have the capacity to undergo endothelial-to-mesenchymal transition (EndMT), a complex differentiation process with key roles both during embryonic development and in adulthood. EndMT can contribute to EC activation and dysfunctional alterations associated with maladaptive tissue responses in human disease. During EndMT, ECs progressively undergo changes leading to expression of mesenchymal markers while repressing EC lineage-specific traits. This phenotypic and functional switch is considered to largely exist in a continuum, being characterized by a gradation of transitioning stages. In this report, we discuss process plasticity and potential reversibility and the hypothesis that different EndMT-derived cell populations may play a different role in disease progression or resolution. In addition, we review advancements in the EndMT field, current technical challenges, as well as therapeutic options and opportunities in the context of cardiovascular biology.
Collapse
Affiliation(s)
- Ignacio Fernando Hall
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Franceska Kishta
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
| | - Yang Xu
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
| | - Andrew H Baker
- Centre for Cardiovascular Science, Queens Medical Research Institute, University of Edinburgh, Edinburgh EH16 4TJ, UK
- CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht 6229ER, The Netherlands
| | - Jason C Kovacic
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Place, New York, NY 10029, USA
- Victor Chang Cardiac Research Institute, Lowy Packer Building, 405 Liverpool Street, Darlinghurst, NSW 2010, Australia
- St. Vincent’s Clinical School and University of New South Wales, 390 Victoria St, Darlinghurst, NSW 2010, Australia
| |
Collapse
|
28
|
Lin HL, Wang S, Sato K, Zhang YQ, He BT, Xu J, Nakazawa T, Qin YJ, Zhang HY. Uric acid-driven NLRP3 inflammasome activation triggers lens epithelial cell senescence and cataract formation. Cell Death Discov 2024; 10:126. [PMID: 38461179 PMCID: PMC10925029 DOI: 10.1038/s41420-024-01900-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: 09/12/2023] [Revised: 02/24/2024] [Accepted: 02/29/2024] [Indexed: 03/11/2024] Open
Abstract
Excessive uric acid (UA) is associated with age-related cataract. A previous study showed that a high UA level in the aqueous humor stimulated the senescence of lens epithelial cells (LECs), leading to cataract progression. To better understand the underlying mechanisms, we investigated UA-driven senescence in human lens tissue samples obtained during surgery, rat lens organ cultures, and in vivo experiments, using senescence-associated β-galactosidase (SA-β-gal) staining, electronic microscopy, Western blotting, and histological analyses. Initially, we identified markedly higher expressions of NLRP3 and caspase-1 in the lens capsules of hyper-uricemic patients compared to normo-uricemic patients. This increase was accompanied by a significant rise in the SA-β-gal positive rate. We next built a cataract model in which rat lenses in an organ culture system were treated with an increasing dosage of UA. Notably, opacification was apparent in the lenses treated with 800 μM of UA starting on the fifth day. Mechanistically, UA treatment not only significantly induced the expression of NLRP3, caspase-1, and IL-1β, but also upregulated the levels of SA-β-gal and the senescence regulators p53 and p21. These effects were fully reversed, and lens opacification was ameliorated by the addition of MCC950, a selective NLRP3 antagonist. Moreover, an in vivo model showed that intravitreal UA injection rapidly induced cataract phenotypes within 21 days, an effect significantly mitigated by co-injection with MCC950. Together, our findings suggest that targeting the UA-induced NLRP3 inflammasome with MCC950 could be a promising strategy for preventing cataract formation associated with inflammageing.
Collapse
Affiliation(s)
- Hong Liang Lin
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Sheng Wang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Kota Sato
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yu Qiao Zhang
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Department of Ophthalmology & Visual Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Bei Ting He
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- School of Medicine, South China University of Technology, Guangzhou, China
| | - Jing Xu
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Toru Nakazawa
- Department of Ophthalmology, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Ophthalmic Imaging and Information Analytics, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Retinal Disease Control, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Advanced Ophthalmic Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
- Department of Collaborative Program for Ophthalmic Drug Discovery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yong Jie Qin
- Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Hong Yang Zhang
- Department of Ophthalmology, Nanfang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
29
|
Zou G, Que L, Liu Y, Lu Q. Interplay of endothelial-mesenchymal transition, inflammation, and autophagy in proliferative diabetic retinopathy pathogenesis. Heliyon 2024; 10:e25166. [PMID: 38327444 PMCID: PMC10847601 DOI: 10.1016/j.heliyon.2024.e25166] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 01/13/2024] [Accepted: 01/22/2024] [Indexed: 02/09/2024] Open
Abstract
Background Assessment and validation of endothelial-mesenchymal transition (EndoMT) in the retinal endothelium of patients with proliferative diabetic retinopathy (PDR) at the level of retinal and vitreous specimens, and preliminary discussion of its regulatory mechanisms. Methods Transcriptome sequencing profiles of CD31+ cells from 9 retinal fibrovascular mem-branes (FVMs) and 4 postmortem retinas were downloaded from GEO databases to analyze EndoMT-related differentially expressed genes (DEGs). Then, 42 PDR patients and 34 idiopathic macular holes (IMH) patients were enrolled as the PDR and control groups, respectively. Vitreous humor (VH) samples were collected, and the expression of EndoMT-related proteins was quantified by enzyme-linked immunosorbent assay. Results A total of 5845 DEGs were identified, and we subsequently focused on the analysis of 24 EndoMT-related marker genes, including the trigger of EndoMT, endothelial genes, mesenchymal genes, transcription factors, inflammatory factors, and autophagy markers. Six of these genes were selected for protein assay validation in VH, showing increased mesenchymal marker (type I collagen α 2 chain, COL1A2) and decreased endothelial marker (VE-cadherin, CDH5) accompanied by increased TGFβ, IL-1β, LC3B and P62 in PDR patients. In addition, anti-VEGF therapy could enhance EndoMT-related phenotypes. Conclusions EndoMT may underlie the pathogenesis of PDR, and the induction and regulation correlate with autophagy defects and the inflammatory response.
Collapse
Affiliation(s)
- Gaocheng Zou
- Department of Ophthalmology, The First Affiliated Hospital of Anhui University of Traditional Chinese Medicine, Hefei, China
| | - Lijuan Que
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yaping Liu
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guang-dong Provincial Key Laboratory of Ophthalmology and Visual Science, Guangzhou, China
| | - Qianyi Lu
- Department of Ophthalmology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
30
|
Zhang K, Han Y, Gu F, Gu Z, Zhao J, Chen J, Chen B, Gao M, Hou Z, Yu X, Cai T, Gao Y, Xie J, Liu T, Liu K. Association between dietary total choline and abdominal aorta calcification among older US adults: A cross-sectional study of the National Health and Nutrition Examination Survey. JPEN J Parenter Enteral Nutr 2024; 48:155-164. [PMID: 37932919 DOI: 10.1002/jpen.2577] [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: 04/12/2023] [Revised: 10/23/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023]
Abstract
BACKGROUND Numerous studies indicate a potential bidirectional association between dietary choline intake and its derivative, betaine, and subclinical atherosclerosis. However, little research has been conducted on the relationship between dietary choline and severe abdominal aortic calcification (SAAC). METHODS This cross-sectional study analyzed population-based data from the National Health and Nutrition Examination Survey (2013-2014). Choline intake and food sources were measured using two 24-h dietary-recall interviews. The abdominal aortic calcification score was measured using a dual-emission x-ray absorptiometry scan. To assess the relationship between choline intake and SAAC, the study utilized restricted cubic spline and a multivariable logistic regression model. RESULTS Among the 2640 individuals included in the study, 10.9% had SAAC. After adjusting for all selected covariates, compared with the lowest quartile of dietary choline, the odds ratios of SAAC for the second-quartile, third-quartile, and fourth-quartile dietary choline intake were 0.63 (95% confidence interval [CI], 0.43-0.93), 0.63 (95% CI, 0.42-0.94), and 0.77 (95% CI, 0.5-1.16), respectively. The study found an L-shaped relationship between dietary choline and SAAC in the dose-response analysis. Subgroup analyses did not demonstrate any statistically significant interaction effects for any subgroup. CONCLUSION The study found that a higher intake of dietary choline is associated with a lower prevalence of SAAC. The dose-response analysis revealed an L-shaped relationship between dietary choline and SAAC. However, further studies are warranted to investigate the direct role of choline in the development of SAAC.
Collapse
Affiliation(s)
- Kai Zhang
- Cardiovascular Surgery Department of Jilin University Second Hospital, Changchun, China
| | - Yu Han
- Department of Ophthalmology, First Hospital of Jilin University, Changchun, China
| | - Fangmin Gu
- Cardiovascular Surgery Department of Jilin University Second Hospital, Changchun, China
| | - Zhaoxuan Gu
- Cardiovascular Surgery Department of Jilin University Second Hospital, Changchun, China
| | - JiaYu Zhao
- Cardiovascular Surgery Department of Jilin University Second Hospital, Changchun, China
| | - Jianguo Chen
- Bethune First College of Clinical Medicine, Jilin University, Changchun, China
| | - Bowen Chen
- Bethune First College of Clinical Medicine, Jilin University, Changchun, China
| | - Min Gao
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, China
| | - Zhengyan Hou
- Bethune Second School of Clinical Medicine, Jilin University, Changchun, China
| | - Xiaoqi Yu
- Bethune Second School of Clinical Medicine, Jilin University, Changchun, China
| | - Tianyi Cai
- Bethune Second School of Clinical Medicine, Jilin University, Changchun, China
| | - Yafang Gao
- Bethune Second School of Clinical Medicine, Jilin University, Changchun, China
| | - Jinyu Xie
- Cardiovascular Surgery Department of Jilin University Second Hospital, Changchun, China
| | - Tianzhou Liu
- Department of Gastrointestinal Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Kexiang Liu
- Cardiovascular Surgery Department of Jilin University Second Hospital, Changchun, China
| |
Collapse
|
31
|
Zaslow SJ, Oliveira-Paula GH, Chen W. Magnesium and Vascular Calcification in Chronic Kidney Disease: Current Insights. Int J Mol Sci 2024; 25:1155. [PMID: 38256228 PMCID: PMC10816532 DOI: 10.3390/ijms25021155] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/12/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
Magnesium (Mg) plays crucial roles in multiple essential biological processes. As the kidneys are the primary organ responsible for maintaining the blood concentration of Mg, people with chronic kidney disease (CKD) may develop disturbances in Mg. While both hyper- and hypomagnesemia may lead to adverse effects, the consequences associated with hypomagnesemia are often more severe and lasting. Importantly, observational studies have shown that CKD patients with hypomagnesemia have greater vascular calcification. Vascular calcification is accelerated and contributes to a high mortality rate in the CKD population. Both in vitro and animal studies have demonstrated that Mg protects against vascular calcification via several potential mechanisms, such as inhibiting the formation of both hydroxyapatite and pathogenic calciprotein particles as well as limiting osteogenic differentiation, a process in which vascular smooth muscle cells in the media layer of the arteries transform into bone-like cells. These preclinical findings have led to several important clinical trials that have investigated the effects of Mg supplementation on vascular calcification in people with CKD. Interestingly, two major clinical studies produced contradictory findings, resulting in a state of equipoise. This narrative review provides an overview of our current knowledge in the renal handling of Mg in health and CKD and the underlying mechanisms by which Mg may protect against vascular calcification. Lastly, we evaluate the strength of evidence from clinical studies on the efficacy of Mg supplementation and discuss future research directions.
Collapse
Affiliation(s)
- Shari J. Zaslow
- Department of Medicine, Nephrology Division, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- The Robert Larner, M.D. College of Medicine, University of Vermont, Burlington, VT 05405, USA
| | - Gustavo H. Oliveira-Paula
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Wei Chen
- Department of Medicine, Nephrology Division, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
32
|
Adelus ML, Ding J, Tran BT, Conklin AC, Golebiewski AK, Stolze LK, Whalen MB, Cusanovich DA, Romanoski CE. Single cell 'omic profiles of human aortic endothelial cells in vitro and human atherosclerotic lesions ex vivo reveals heterogeneity of endothelial subtype and response to activating perturbations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.04.03.535495. [PMID: 37066416 PMCID: PMC10104082 DOI: 10.1101/2023.04.03.535495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Objective Endothelial cells (ECs), macrophages, and vascular smooth muscle cells (VSMCs) are major cell types in atherosclerosis progression, and heterogeneity in EC sub-phenotypes are becoming increasingly appreciated. Still, studies quantifying EC heterogeneity across whole transcriptomes and epigenomes in both in vitro and in vivo models are lacking. Approach and Results To create an in vitro dataset to study human EC heterogeneity, multiomic profiling concurrently measuring transcriptomes and accessible chromatin in the same single cells was performed on six distinct primary cultures of human aortic ECs (HAECs). To model pro-inflammatory and activating environments characteristic of the atherosclerotic microenvironment in vitro, HAECs from at least three donors were exposed to three distinct perturbations with their respective controls: transforming growth factor beta-2 (TGFB2), interleukin-1 beta (IL1B), and siRNA-mediated knock-down of the endothelial transcription factor ERG (siERG). To form a comprehensive in vivo/ex vivo dataset of human atherosclerotic cell types, meta-analysis of single cell transcriptomes across 17 human arterial specimens was performed. Two computational approaches quantitatively evaluated the similarity in molecular profiles between heterogeneous in vitro and in vivo cell profiles. HAEC cultures were reproducibly populated by 4 major clusters with distinct pathway enrichment profiles: EC1-angiogenic, EC2-proliferative, EC3-activated/mesenchymal-like, and EC4-mesenchymal. Exposure to siERG, IL1B or TGFB2 elicited mostly distinct transcriptional and accessible chromatin responses. EC1 and EC2, the most canonically 'healthy' EC populations, were affected predominantly by siERG; the activated cluster EC3 was most responsive to IL1B; and the mesenchymal population EC4 was most affected by TGFB2. Quantitative comparisons between in vitro and in vivo transcriptomes confirmed EC1 and EC2 as most canonically EC-like, and EC4 as most mesenchymal with minimal effects elicited by siERG and IL1B. Lastly, accessible chromatin regions unique to EC2 and EC4 were most enriched for coronary artery disease (CAD)-associated SNPs from GWAS, suggesting these cell phenotypes harbor CAD-modulating mechanisms. Conclusion Primary EC cultures contain markedly heterogeneous cell subtypes defined by their molecular profiles. Surprisingly, the perturbations used here, which have been reported by others to be involved in the pathogenesis of atherosclerosis as well as induce endothelial-to-mesenchymal transition (EndMT), only modestly shifted cells between subpopulations, suggesting relatively stable molecular phenotypes in culture. Identifying consistently heterogeneous EC subpopulations between in vitro and in vivo models should pave the way for improving in vitro systems while enabling the mechanisms governing heterogeneous cell state decisions.
Collapse
Affiliation(s)
- Maria L. Adelus
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
- The Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, 85721, USA
| | - Jiacheng Ding
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Binh T. Tran
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Austin C. Conklin
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Anna K. Golebiewski
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Lindsey K. Stolze
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Michael B. Whalen
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
| | - Darren A. Cusanovich
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
- Asthma and Airway Disease Research Center, The University of Arizona, Tucson, AZ, 85721, USA
| | - Casey E. Romanoski
- The Department of Cellular and Molecular Medicine, The University of Arizona, Tucson, AZ 85721, USA
- The Clinical Translational Sciences Graduate Program, The University of Arizona, Tucson, AZ, 85721, USA
- Asthma and Airway Disease Research Center, The University of Arizona, Tucson, AZ, 85721, USA
| |
Collapse
|
33
|
Wang X, Wang Z, He J. Similarities and Differences of Vascular Calcification in Diabetes and Chronic Kidney Disease. Diabetes Metab Syndr Obes 2024; 17:165-192. [PMID: 38222032 PMCID: PMC10788067 DOI: 10.2147/dmso.s438618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 12/21/2023] [Indexed: 01/16/2024] Open
Abstract
Presently, the mechanism of occurrence and development of vascular calcification (VC) is not fully understood; a range of evidence suggests a positive association between diabetes mellitus (DM) and VC. Furthermore, the increasing burden of central vascular disease in patients with chronic kidney disease (CKD) may be due, at least in part, to VC. In this review, we will review recent advances in the mechanisms of VC in the context of CKD and diabetes. The study further unveiled that VC is induced through the stimulation of pro-inflammatory factors, which in turn impairs endothelial function and triggers similar mechanisms in both disease contexts. Notably, hyperglycemia was identified as the distinctive mechanism driving calcification in DM. Conversely, in CKD, calcification is facilitated by mechanisms including mineral metabolism imbalance and the presence of uremic toxins. Additionally, we underscore the significance of investigating vascular alterations and newly identified molecular pathways as potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Xiabo Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| | - Jianqiang He
- Department of Nephrology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, People’s Republic of China
| |
Collapse
|
34
|
Diallo LH, Mariette J, Laugero N, Touriol C, Morfoisse F, Prats AC, Garmy-Susini B, Lacazette E. Specific Circular RNA Signature of Endothelial Cells: Potential Implications in Vascular Pathophysiology. Int J Mol Sci 2024; 25:680. [PMID: 38203852 PMCID: PMC10779679 DOI: 10.3390/ijms25010680] [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: 11/21/2023] [Revised: 12/23/2023] [Accepted: 12/28/2023] [Indexed: 01/12/2024] Open
Abstract
Circular RNAs (circRNAs) are a recently characterized family of gene transcripts forming a covalently closed loop of single-stranded RNA. The extent of their potential for fine-tuning gene expression is still being discovered. Several studies have implicated certain circular RNAs in pathophysiological processes within vascular endothelial cells and cancer cells independently. However, to date, no comparative study of circular RNA expression in different types of endothelial cells has been performed and analysed through the lens of their central role in vascular physiology and pathology. In this work, we analysed publicly available and original RNA sequencing datasets from arterial, veinous, and lymphatic endothelial cells to identify common and distinct circRNA expression profiles. We identified 4713 distinct circRNAs in the compared endothelial cell types, 95% of which originated from exons. Interestingly, the results show that the expression profile of circular RNAs is much more specific to each cell type than linear RNAs, and therefore appears to be more suitable for distinguishing between them. As a result, we have discovered a specific circRNA signature for each given endothelial cell type. Furthermore, we identified a specific endothelial cell circRNA signature that is composed four circRNAs: circCARD6, circPLXNA2, circCASC15 and circEPHB4. These circular RNAs are produced by genes that are related to endothelial cell migration pathways and cancer progression. More detailed studies of their functions could lead to a better understanding of the mechanisms involved in physiological and pathological (lymph)angiogenesis and might open new ways to tackle tumour spread through the vascular system.
Collapse
Affiliation(s)
- Leïla Halidou Diallo
- U1297-I2MC, INSERM, University of Toulouse, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse, France; (L.H.D.); (N.L.); (F.M.); (A.-C.P.); (B.G.-S.)
| | - Jérôme Mariette
- MIAT, University of Toulouse, INRAE, 31326 Castanet-Tolosan, France;
| | - Nathalie Laugero
- U1297-I2MC, INSERM, University of Toulouse, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse, France; (L.H.D.); (N.L.); (F.M.); (A.-C.P.); (B.G.-S.)
| | - Christian Touriol
- UMR1037 INSERM, University of Toulouse, 2 Avenue Hubert Curien, 31100 Toulouse, France;
| | - Florent Morfoisse
- U1297-I2MC, INSERM, University of Toulouse, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse, France; (L.H.D.); (N.L.); (F.M.); (A.-C.P.); (B.G.-S.)
| | - Anne-Catherine Prats
- U1297-I2MC, INSERM, University of Toulouse, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse, France; (L.H.D.); (N.L.); (F.M.); (A.-C.P.); (B.G.-S.)
| | - Barbara Garmy-Susini
- U1297-I2MC, INSERM, University of Toulouse, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse, France; (L.H.D.); (N.L.); (F.M.); (A.-C.P.); (B.G.-S.)
| | - Eric Lacazette
- U1297-I2MC, INSERM, University of Toulouse, 1 Avenue Jean Poulhes, BP 84225, 31432 Toulouse, France; (L.H.D.); (N.L.); (F.M.); (A.-C.P.); (B.G.-S.)
| |
Collapse
|
35
|
Guan J, Gong S, He Q, Wang X, Shen S, Wu X, Shan J, Gong T, Yang Y, Xie H. Soluble urokinase plasminogen activator receptor is associated with cardiovascular calcification in peritoneal dialysis patients. Int Urol Nephrol 2024; 56:191-198. [PMID: 37195572 DOI: 10.1007/s11255-023-03623-z] [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: 04/01/2023] [Accepted: 05/01/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Cardiovascular disease (CVD) is an important cause of morbidity and mortality in peritoneal dialysis (PD) patients. Cardiovascular calcification (CVC) is highly prevalent in PD patients and could predict their cardiovascular mortality. Soluble urokinase plasminogen activator receptor (suPAR) is closely associated with coronary artery calcification in hemodialysis patients and is an important predictor of CVD. However, the role of suPAR in PD patients is poorly understood. We investigated the relationship between serum suPAR and CVC in PD patients. METHODS Abdominal aortic calcification (AAC) was assessed by lateral lumbar radiography, coronary artery calcification (CAC) by multi-slice computed tomography, and cardiac valvular calcification (ValvC) by echocardiography. CVC was defined as confirmed presence of calcification in one site (AAC, CAC, or ValvC). Patients were divided into CVC group and non-CVC group. Demographic characteristics, biochemical variables, comorbidities, PD regimen, serum suPAR, and medication were compared between the two groups. Logistic regression was conducted to determine association between serum suPAR and presence of CVC. The receiver-operator curve (ROC) was plotted to calculate the area under the curve (AUC) for suPAR to identify CVC and ValvC. RESULTS Of 226 PD patients, 111 (49.1%) had AAC, 155 (68.6%) had CAC, and 26 (11.5%) had ValvC. There were significant differences in age, BMI, diabetes, white blood cell, phosphorus, hs-CRP, suPAR, time on dialysis, total volume of dialysate, ultrafiltration, volume of urine, and Kt/V between CVC and non-CVC group. Serum suPAR was associated with CVC by multivariate logistic regression analysis in PD patients, especially in elderly patients. The levels of serum suPAR were closely related to the degree of AAC, CAC, and ValvC in PD patients. The incidence of CVC was higher in patients with higher levels of suPAR. The ROC curve showed that serum suPAR had a predictive value for CVC (AUC = 0.651), especially for ValvC (AUC = 0.828). CONCLUSION Cardiovascular calcification is prevalent in PD patients. High levels of serum suPAR are associated with cardiovascular calcification in PD patients, especially in elderly patients.
Collapse
Affiliation(s)
- Jichao Guan
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Shuwen Gong
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Qiuli He
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Xiaoling Wang
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Shuijuan Shen
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Xiujuan Wu
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Juanping Shan
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Tujian Gong
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Yanru Yang
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China
| | - Haiying Xie
- Shaoxing People's Hospital, Shaoxing, Zhejiang, China.
| |
Collapse
|
36
|
Zhang K, Gu F, Han Y, Cai T, Gu Z, Chen J, Chen B, Gao M, Hou Z, Yu X, Zhao J, Gao Y, Xie J, Hu R, Liu T, Li B. Association between dietary calcium intake and severe abdominal aorta calcification among American adults: a cross-sectional analysis of the National Health and Nutrition Examination Survey. Ther Adv Cardiovasc Dis 2024; 18:17539447241232774. [PMID: 38415471 PMCID: PMC10903221 DOI: 10.1177/17539447241232774] [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: 07/06/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Evidence regarding the relationship between dietary calcium intake and severe abdominal aortic calcification (AAC) is limited. Therefore, this study aimed to investigate the association between dietary calcium intake and severe AAC in American adults based on data from the National Health and Nutrition Examination Survey (NHANES). METHODS The present cross-sectional study utilized data from the NHANES 2013-2014, a population-based dataset. Dietary calcium intake was assessed using two 24-h dietary recall interviews. Quantification of the AAC scores was accomplished utilizing the Kauppila score system, whereby severe AAC was defined as having an AAC score greater than 6. We used multivariable logistic regression models, a restricted cubic spline analysis, and a two-piecewise linear regression model to show the effect of calcium intake on severe AAC. RESULTS Out of the 2640 individuals examined, 10.9% had severe AAC. Following the adjustment for confounding variables, an independent association was discovered between an augmented intake of dietary calcium and the incidence of severe AAC. When comparing individuals in the second quartile (Q2) of dietary calcium intake with those in the lowest quartile (Q1), a decrease in the occurrence of severe AAC was observed (odds ratio: 0.66; 95% confidence interval: 0.44-0.99). Furthermore, the relationship between dietary calcium intake and severe AAC demonstrated an L-shaped pattern, with an inflection point observed at 907.259 mg/day. Subgroup analyses revealed no significant interaction effects. CONCLUSION The study revealed that the relationship between dietary calcium intake and severe AAC in American adults is L-shaped, with an inflection point of 907.259 mg/day. Further research is required to confirm this association.
Collapse
Affiliation(s)
- Kai Zhang
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Fangming Gu
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Han
- Department of Ophthalmology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Tianyi Cai
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Zhaoxuan Gu
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jianguo Chen
- Bethune First College of Clinical Medicine, Jilin University, Changchun, Jilin, China
| | - Bowen Chen
- Bethune First College of Clinical Medicine, Jilin University, Changchun, Jilin, China
| | - Min Gao
- Department of Cancer Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Zhengyan Hou
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoqi Yu
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - JiaYu Zhao
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yafang Gao
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jinyu Xie
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Rui Hu
- Bethune First College of Clinical Medicine, Jilin University, Changchun, Jilin, China
| | - Tianzhou Liu
- The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Bo Li
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Jilin University, No. 218, Ziqiang Street, Changchun, Jilin, 130000, China
| |
Collapse
|
37
|
Mimouni M, Lajoix AD, Desmetz C. Experimental Models to Study Endothelial to Mesenchymal Transition in Myocardial Fibrosis and Cardiovascular Diseases. Int J Mol Sci 2023; 25:382. [PMID: 38203553 PMCID: PMC10779210 DOI: 10.3390/ijms25010382] [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: 12/01/2023] [Revised: 12/21/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Fibrosis is a common feature of cardiovascular diseases and targets multiple organs, such as the heart and vessels. Endothelial to mesenchymal transition is a complex, vital process that occurs during embryonic formation and plays a crucial role in cardiac development. It is also a fundamental process implicated in cardiac fibrosis and repair, but also in other organs. Indeed, in numerous cardiovascular diseases, the endothelial-to-mesenchymal transition has been shown to be involved in the generation of fibroblasts that are able to produce extracellular matrix proteins such as type I collagen. This massive deposition results in tissue stiffening and organ dysfunction. To advance our understanding of this process for the development of new specific diagnostic and therapeutic strategies, it is essential to develop relevant cellular and animal models of this process. In this review, our aim was to gain an in-depth insight into existing in vitro and in vivo models of endothelial to mesenchymal transition in cardiovascular diseases with a focus on cardiac fibrosis. We discuss important parameters impacting endothelial to mesenchymal transition, and we give perspectives for the development of relevant models to decipher the underlying mechanisms and ultimately find new treatments specific to fibrosis happening in cardiovascular diseases.
Collapse
Affiliation(s)
- Mohammed Mimouni
- Biocommunication in Cardio-Metabolism (BC2M), University of Montpellier, 34000 Montpellier, France
| | - Anne-Dominique Lajoix
- Biocommunication in Cardio-Metabolism (BC2M), University of Montpellier, 34000 Montpellier, France
| | - Caroline Desmetz
- Biocommunication in Cardio-Metabolism (BC2M), University of Montpellier, 34000 Montpellier, France
| |
Collapse
|
38
|
Cooke JP, Lai L. Transflammation in tissue regeneration and response to injury: How cell-autonomous inflammatory signaling mediates cell plasticity. Adv Drug Deliv Rev 2023; 203:115118. [PMID: 37884127 PMCID: PMC10842620 DOI: 10.1016/j.addr.2023.115118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 08/01/2023] [Accepted: 10/23/2023] [Indexed: 10/28/2023]
Abstract
Inflammation is a first responder against injury and infection and is also critical for the regeneration and repair of tissue after injury. The role of professional immune cells in tissue healing is well characterized. Professional immune cells respond to pathogens with humoral and cytotoxic responses; remove cellular debris through efferocytosis; secrete angiogenic cytokines and growth factors to repair the microvasculature and parenchyma. However, non-immune cells are also capable of responding to damage or pathogens. Non-immune somatic cells express pattern recognition receptors (PRRs) to detect pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs). The PRRs activation leads to the release of inflammatory cytokines required for tissue defense and repair. Notably, the activation of PRRs also triggers epigenetic changes that promote DNA accessibility and cellular plasticity. Thus, non-immune cells directly respond to the local inflammatory cues and can undergo phenotypic modifications or even cell lineage transitions to facilitate tissue regeneration. This review will focus on the novel role of cell-autonomous inflammatory signaling in mediating cell plasticity, a process which is termed transflammation. We will discuss the regulation of this process by changes in the functions and expression levels of epigenetic modifiers, as well as metabolic and ROS/RNS-mediated epigenetic modulation of DNA accessibility during cell fate transition. We will highlight the recent technological developments in detecting cell plasticity and potential therapeutic applications of transflammation in tissue regeneration.
Collapse
Affiliation(s)
- John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Li Lai
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States.
| |
Collapse
|
39
|
刘 颖, 马 良, 付 平. [Ketone Body Metabolism and Renal Diseases]. SICHUAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF SICHUAN UNIVERSITY. MEDICAL SCIENCE EDITION 2023; 54:1091-1096. [PMID: 38162055 PMCID: PMC10752776 DOI: 10.12182/20231160202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Indexed: 01/03/2024]
Abstract
A ketogenic diet limits energy supply from glucose and stimulates lipolysis, lipid oxidation, and ketogenesis, resulting in elevated levels of ketone bodies in the bloodstream. Ketone bodies are synthesized in the mitochondrial matrix of liver cells and β-hydroxybutyric acid (BHB) is the most abundant type of ketone body. Herein, we reviewed published findings on the metabolism of ketone bodies and the role of BHB in renal diseases. Through blood circulation, ketone bodies reach metabolically active tissues and provides an alternative source of energy. BHB, being a signaling molecule, mediates various types of cellular signal transduction and participates in the development and progression of many diseases. BHB also has protective and therapeutic effects on a variety of renal diseases. BHB improves the prognosis of renal diseases, such as diabetic kidney disease, chronic kidney disease, acute kidney injury, and polycystic kidney disease, through its antioxidant, anti-inflammatory, and stress response mechanisms. Previous studies have focused on the role of ketone bodies in regulating inflammation and oxidative stress in immune cells. Investigations into the effect of elevated levels of ketone bodies on the metabolism of renal podocytes and tubular cells remain inconclusive. Further research is needed to investigate the effect of BHB on podocyte damage and podocyte senescence in renal diseases.
Collapse
Affiliation(s)
- 颖 刘
- 四川大学华西医院 肾脏内科 (成都 610041)Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 肾脏病研究所 (成都 610041)Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 良 马
- 四川大学华西医院 肾脏内科 (成都 610041)Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 肾脏病研究所 (成都 610041)Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| | - 平 付
- 四川大学华西医院 肾脏内科 (成都 610041)Department of Nephrology, West China Hospital, Sichuan University, Chengdu 610041, China
- 四川大学华西医院 肾脏病研究所 (成都 610041)Kidney Research Institute, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
40
|
Song JH, Liu MY, Ma YX, Wan QQ, Li J, Diao XO, Niu LN. Inflammation-associated ectopic mineralization. FUNDAMENTAL RESEARCH 2023; 3:1025-1038. [PMID: 38933004 PMCID: PMC11197766 DOI: 10.1016/j.fmre.2022.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/06/2022] [Accepted: 04/21/2022] [Indexed: 10/18/2022] Open
Abstract
Ectopic mineralization refers to the deposition of mineralized complexes in the extracellular matrix of soft tissues. Calcific aortic valve disease, vascular calcification, gallstones, kidney stones, and abnormal mineralization in arthritis are common examples of ectopic mineralization. They are debilitating diseases and exhibit excess mortality, disability, and morbidity, which impose on patients with limited social or financial resources. Recent recognition that inflammation plays an important role in ectopic mineralization has attracted the attention of scientists from different research fields. In the present review, we summarize the origin of inflammation in ectopic mineralization and different channels whereby inflammation drives the initiation and progression of ectopic mineralization. The current knowledge of inflammatory milieu in pathological mineralization is reviewed, including how immune cells, pro-inflammatory mediators, and osteogenic signaling pathways induce the osteogenic transition of connective tissue cells, providing nucleating sites and assembly of aberrant minerals. Advances in the understanding of the underlying mechanisms involved in inflammatory-mediated ectopic mineralization enable novel strategies to be developed that may lead to the resolution of these enervating conditions.
Collapse
Affiliation(s)
| | | | | | - Qian-Qian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jing Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xiao-Ou Diao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Li-Na Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Centre for Oral Diseases & Shaanxi Key Laboratory of Stomatology, Department of Prosthodontics, School of Stomatology, The Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
41
|
Wits M, Becher C, de Man F, Sanchez-Duffhues G, Goumans MJ. Sex-biased TGFβ signalling in pulmonary arterial hypertension. Cardiovasc Res 2023; 119:2262-2277. [PMID: 37595264 PMCID: PMC10597641 DOI: 10.1093/cvr/cvad129] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 06/21/2023] [Accepted: 07/04/2023] [Indexed: 08/20/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a rare cardiovascular disorder leading to pulmonary hypertension and, often fatal, right heart failure. Sex differences in PAH are evident, which primarily presents with a female predominance and increased male severity. Disturbed signalling of the transforming growth factor-β (TGFβ) family and gene mutations in the bone morphogenetic protein receptor 2 (BMPR2) are risk factors for PAH development, but how sex-specific cues affect the TGFβ family signalling in PAH remains poorly understood. In this review, we aim to explore the sex bias in PAH by examining sex differences in the TGFβ signalling family through mechanistical and translational evidence. Sex hormones including oestrogens, progestogens, and androgens, can determine the expression of receptors (including BMPR2), ligands, and soluble antagonists within the TGFβ family in a tissue-specific manner. Furthermore, sex-related genetic processes, i.e. Y-chromosome expression and X-chromosome inactivation, can influence the TGFβ signalling family at multiple levels. Given the clinical and mechanistical similarities, we expect that the conclusions arising from this review may apply also to hereditary haemorrhagic telangiectasia (HHT), a rare vascular disorder affecting the TGFβ signalling family pathway. In summary, we anticipate that investigating the TGFβ signalling family in a sex-specific manner will contribute to further understand the underlying processes leading to PAH and likely HHT.
Collapse
Affiliation(s)
- Marius Wits
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Clarissa Becher
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| | - Frances de Man
- Department of Pulmonary Medicine, Amsterdam University Medical Center (UMC) (Vrije Universiteit), 1081 HV Amsterdam, The Netherlands
| | - Gonzalo Sanchez-Duffhues
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
- Nanomaterials and Nanotechnology Research Center (CINN-CSIC), Health Research Institute of Asturias (ISPA), 33011 Oviedo, Spain
| | - Marie-José Goumans
- Department of Cell and Chemical Biology, Leiden University Medical Center, Einthovenweg 20, 2333 ZC Leiden, The Netherlands
| |
Collapse
|
42
|
Yang S, Zeng Z, Yuan Q, Chen Q, Wang Z, Xie H, Liu J. Vascular calcification: from the perspective of crosstalk. MOLECULAR BIOMEDICINE 2023; 4:35. [PMID: 37851172 PMCID: PMC10584806 DOI: 10.1186/s43556-023-00146-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 09/20/2023] [Indexed: 10/19/2023] Open
Abstract
Vascular calcification (VC) is highly correlated with cardiovascular disease morbidity and mortality, but anti-VC treatment remains an area to be tackled due to the ill-defined molecular mechanisms. Regardless of the type of VC, it does not depend on a single cell but involves multi-cells/organs to form a complex cellular communication network through the vascular microenvironment to participate in the occurrence and development of VC. Therefore, focusing only on the direct effect of pathological factors on vascular smooth muscle cells (VSMCs) tends to overlook the combined effect of other cells and VSMCs, including VSMCs-VSMCs, ECs-VMSCs, Macrophages-VSMCs, etc. Extracellular vesicles (EVs) are a collective term for tiny vesicles with a membrane structure that are actively secreted by cells, and almost all cells secrete EVs. EVs docked on the surface of receptor cells can directly mediate signal transduction or transfer their contents into the cell to elicit a functional response from the receptor cells. They have been proven to participate in the VC process and have also shown attractive therapeutic prospects. Based on the advantages of EVs and the ability to be detected in body fluids, they may become a novel therapeutic agent, drug delivery vehicle, diagnostic and prognostic biomarker, and potential therapeutic target in the future. This review focuses on the new insight into VC molecular mechanisms from the perspective of crosstalk, summarizes how multi-cells/organs interactions communicate via EVs to regulate VC and the emerging potential of EVs as therapeutic methods in VC. We also summarize preclinical experiments on crosstalk-based and the current state of clinical studies on VC-related measures.
Collapse
Affiliation(s)
- Shiqi Yang
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zhaolin Zeng
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qing Yuan
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
- Department of Clinical Laboratory Medicine, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Qian Chen
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China
| | - Zuo Wang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hui Xie
- Department of Orthopaedics, Movement System Injury and Repair Research Centre, Xiangya Hospital, Central South University, Changsha, Hunan Province, China.
| | - Jianghua Liu
- Department of Metabolism and Endocrinology, Hengyang Medical School, The First Affiliated Hospital, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
43
|
Liu Z, Cheng P, Feng T, Xie Z, Yang M, Chen Z, Hu S, Han D, Chen W. Nrf2/HO-1 blocks TXNIP/NLRP3 interaction via elimination of ROS in oxygen-glucose deprivation-induced neuronal necroptosis. Brain Res 2023; 1817:148482. [PMID: 37442251 DOI: 10.1016/j.brainres.2023.148482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/20/2023] [Accepted: 07/08/2023] [Indexed: 07/15/2023]
Abstract
Acute ischemic stroke (AIS) is known to trigger a cascade of inflammatory events that induces secondary tissue damages. As a type of regulated inflammatory cell death, necroptosis is associated with AIS, whilst its regulation during neuroinflammation is not well understood. In particular, the actual function of NOD-like-receptor family pyrin domain-containing-3(NLRP3) inflammasome in cortical neuronal necroptosis still not clear. Herein, we explored the function of nuclear factor erythroid-2 related factor-2 (Nrf2)/heme oxygenase-1 (HO-1) in oxygen-glucose deprivation (OGD) induced neuronal necroptosis and its underlying mechanism. To establish an in vitro model of neuronal necrosis, we used OGD/caspase-8 inhibitors (Q-VD-OPh, QVD) to treat rat primary cortical neurons (PCNs) after reoxygenation, wherein we found that the model cause an elevated ROS levels by mediating TXNIP/NLRP3 interactions, which in turn activated the NLRP3 inflammasome. Also, we observed that regulation of nuclear factor erythroid-2 related factor-2 (Nrf2) promoted heme oxygenase-1 (HO-1) expression and decreased TXNIP (a protein that relate oxidative stress to activation of inflammasome) and ROS levels, which negatively regulated the expression of OGD-induced activation of NLRP3 inflammasomes. In addition, HO-1 weakened NLRP3 inflammation body activation, which suggests that Nrf2-regulated HO-1 could block the interaction between TXNIP and NLRP3 in OGD/R-treated cortical neurons by inhibiting ROS production. Our study has discovered the importance of Nrf2/HO-1 signaling cascade for inhibiting inflammasome of NLRP3, which negatively regulated necrosis. Therefore, NLRP3 is considered a potential target for a novel neuroprotective approach, which can expand the therapeutic windows of stroke drugs.
Collapse
Affiliation(s)
- Zhihan Liu
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Ping Cheng
- Bengbu Medical College, Bengbu, Anhui 233000, China
| | - Tao Feng
- Department of Rehabilitation Medicine, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zhiyuan Xie
- Department of Gastrointestinal Surgery, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, China
| | - Meifang Yang
- Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Zhiren Chen
- Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Shuqun Hu
- Institute of Emergency Rescue Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Dong Han
- Institute of Emergency Rescue Medicine, Xuzhou Medical University, Xuzhou, Jiangsu 221002, China
| | - Weiwei Chen
- Department of Neurology, Xuzhou Central Hospital/The Xuzhou School of Clinical Medicine of Nanjing Medical University/ XuZhou Clinical School of Xuzhou Medical University, Xuzhou, Jiangsu 221009, China.
| |
Collapse
|
44
|
Evenepoel P, Stenvinkel P, Shanahan C, Pacifici R. Inflammation and gut dysbiosis as drivers of CKD-MBD. Nat Rev Nephrol 2023; 19:646-657. [PMID: 37488276 DOI: 10.1038/s41581-023-00736-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/19/2023] [Indexed: 07/26/2023]
Abstract
Two decades ago, Kidney Disease: Improving Global Outcomes coined the term chronic kidney disease-mineral and bone disorder (CKD-MBD) to describe the syndrome of biochemical, bone and extra-skeletal calcification abnormalities that occur in patients with CKD. CKD-MBD is a prevalent complication and contributes to the excessively high burden of fractures and cardiovascular disease, loss of quality of life and premature mortality in patients with CKD. Thus far, therapy has focused primarily on phosphate retention, abnormal vitamin D metabolism and parathyroid hormone disturbances, but these strategies have largely proved unsuccessful, thus calling for paradigm-shifting concepts and innovative therapeutic approaches. Interorgan crosstalk is increasingly acknowledged to have an important role in health and disease. Accordingly, mounting evidence suggests a role for both the immune system and the gut microbiome in bone and vascular biology. Gut dysbiosis, compromised gut epithelial barrier and immune cell dysfunction are prominent features of the uraemic milieu. These alterations might contribute to the inflammatory state observed in CKD and could have a central role in the pathogenesis of CKD-MBD. The emerging fields of osteoimmunology and osteomicrobiology add another level of complexity to the pathogenesis of CKD-MBD, but also create novel therapeutic opportunities.
Collapse
Affiliation(s)
- Pieter Evenepoel
- Laboratory of Nephrology, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Herestraat, Leuven, Belgium.
| | - Peter Stenvinkel
- Department of Renal Medicine M99, Karolinska University Hospital, Stockholm, Sweden
| | - Catherine Shanahan
- British Heart Foundation Centre of Excellence, School of Cardiovascular and Metabolic Medicine and Sciences, King's College London, London, UK
| | - Roberto Pacifici
- Division of Endocrinology, Metabolism and Lipids, Department of Medicine, Emory Microbiome Research Center, and Immunology and Molecular Pathogenesis Program, Emory University, Atlanta, GA, USA
| |
Collapse
|
45
|
Huang M, Wu Z, Jia L, Wang Y, Gao S, Liu Y, Zhang Y, Li J. Bioinformatics and network pharmacology identify promotional effects and potential mechanisms of ethanol on esophageal squamous cell carcinoma and experimental validation. Toxicol Appl Pharmacol 2023; 474:116615. [PMID: 37406968 DOI: 10.1016/j.taap.2023.116615] [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: 04/06/2023] [Revised: 06/28/2023] [Accepted: 07/01/2023] [Indexed: 07/07/2023]
Abstract
Ethanol is an important risk factor for esophageal squamous cell carcinoma (ESCC); however, the molecular mechanisms behind how ethanol promotes ESCC development remain poorly understood. In this study, ethanol-ESCC-associated target genes were constructed and screened using network pharmacology and subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) and bioinformatics analysis. A mouse ethanol-exposed esophageal cancer model was constructed with 4-nitroquinoline-1-oxide (4-NQO) to assess its survival and tumor lesion status, and the mechanism of ethanol-promoted ESCC lesions was verified by qRT-PCR and Western blotting. The results showed that 126 ethanol-ESCC crossover genes were obtained, which were significantly enriched in the PI3K/AKT signaling pathway. Bioinformatics results showed that the target genes TNF, IL6, IL1β and JUN were highly expressed in esophageal tumor samples and positively correlated with tumor proliferation and apoptosis genes, and the genetic information of these genes was mutated to different degrees. Animal model experiments showed that ethanol decreased the survival rate and aggravated the occurrence of esophageal cancer in mice. qRT-PCR showed that ethanol promoted the expression of TNF, IL6, IL1β and JUN mRNA in mouse esophageal tumor tissues, and Western blotting showed that ethanol promoted p-PI3K and p-AKT protein expression in mouse esophageal tumor tissues. In conclusion, ethanol promotes esophageal carcinogenesis by increasing the expression of TNF, IL6, IL1β and JUN and activating the PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Ming Huang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Zhongbing Wu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Lei Jia
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yu Wang
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Shuang Gao
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Ying Liu
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China
| | - Yushuang Zhang
- The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| | - Jing Li
- College of Integrated Chinese and Western Medicine, Hebei Medical University, Shijiazhuang 050017, China; The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China.
| |
Collapse
|
46
|
Feng Y, Luo S, Fan D, Guo X, Ma S. The role of vascular endothelial cells in tumor metastasis. Acta Histochem 2023; 125:152070. [PMID: 37348328 DOI: 10.1016/j.acthis.2023.152070] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 06/11/2023] [Accepted: 06/12/2023] [Indexed: 06/24/2023]
Abstract
Vascular endothelial cells (VECs) are an integral component of the inner lining of blood vessels, and their functions are essential for the proper functioning of the vascular system. The tight junctions formed by VECs act as a significant barrier to the intravasation and extravasation of tumor cells (TCs). In addition to that, the proliferation, activation, and migration of VECs play a vital role in the growth of new blood vessels, a process known as tumor angiogenesis, which is closely related to the malignant progression of tumors. However, during tumor progression, VECs undergo endothelial-to-mesenchymal transition (EndMT), which further promotes tumor progression. Furthermore, VECs act as the first line of defense against effector immune cells and help prevent immune cells from infiltrating into tumor tissues. VECs also secrete various cytokines that can contribute to regulating the stemness of tumor stem cells. Thus, it has been increasingly recognized that dysfunction of VECs is one of the key driving forces behind tumor metastasis, and therapeutic strategies targeting VECs have the potential to be an effective means of antitumor therapy. This review aims to present a comprehensive overview of the role and mechanisms of VECs in regulating tumor progression and metastasis, providing insights into the possibilities for the development of novel antitumor therapies that target VECs.
Collapse
Affiliation(s)
- Ying Feng
- Department of Critical Care Medicine, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China
| | - Shan Luo
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Dandan Fan
- Hubei Key Laboratory of Embryonic Stem Cell Research, School of Biomedical Engineering, Hubei University of Medicine, Shiyan, Hubei 442000, China
| | - Xingrong Guo
- Department of Critical Care Medicine, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| | - Shinan Ma
- Department of Critical Care Medicine, Hubei Clinical Research Center for Umbilical Cord Blood Hematopoietic Stem Cells, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, Hubei, China; Hubei Key Laboratory of Embryonic Stem Cell Research, School of Basic Medical Sciences, Taihe Hospital, Hubei University of Medicine, Shiyan, Hubei 442000, China.
| |
Collapse
|
47
|
Bouhamida E, Morciano G, Pedriali G, Ramaccini D, Tremoli E, Giorgi C, Pinton P, Patergnani S. The Complex Relationship between Hypoxia Signaling, Mitochondrial Dysfunction and Inflammation in Calcific Aortic Valve Disease: Insights from the Molecular Mechanisms to Therapeutic Approaches. Int J Mol Sci 2023; 24:11105. [PMID: 37446282 DOI: 10.3390/ijms241311105] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 06/26/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS) is among the most common causes of cardiovascular mortality in an aging population worldwide. The pathomechanisms of CAVS are such a complex and multifactorial process that researchers are still making progress to understand its physiopathology as well as the complex players involved in CAVS pathogenesis. Currently, there is no successful and effective treatment to prevent or slow down the disease. Surgical and transcatheter valve replacement represents the only option available for treating CAVS. Insufficient oxygen availability (hypoxia) has a critical role in the pathogenesis of almost all CVDs. This process is orchestrated by the hallmark transcription factor, hypoxia-inducible factor 1 alpha subunit (HIF-1α), which plays a pivotal role in regulating various target hypoxic genes and metabolic adaptations. Recent studies have shown a great deal of interest in understanding the contribution of HIF-1α in the pathogenesis of CAVS. However, it is deeply intertwined with other major contributors, including sustained inflammation and mitochondrial impairments, which are attributed primarily to CAVS. The present review aims to cover the latest understanding of the complex interplay effect of hypoxia signaling pathways, mitochondrial dysfunction, and inflammation in CAVS. We propose further hypotheses and interconnections on the complexity of these impacts in a perspective of better understanding the pathophysiology. These interplays will be examined considering recent studies that shall help us better dissect the molecular mechanism to enable the design and development of potential future therapeutic approaches that can prevent or slow down CAVS processes.
Collapse
Affiliation(s)
- Esmaa Bouhamida
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Giampaolo Morciano
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Gaia Pedriali
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Daniela Ramaccini
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Elena Tremoli
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
| | - Carlotta Giorgi
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Paolo Pinton
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Simone Patergnani
- Translational Research Center, Maria Cecilia Hospital GVM Care & Research, 48033 Cotignola, Italy
- Department of Medical Sciences, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
48
|
Lu D, Jiang H, Zou T, Jia Y, Zhao Y, Wang Z. Endothelial-to-mesenchymal transition: New insights into vascular calcification. Biochem Pharmacol 2023; 213:115579. [PMID: 37589048 DOI: 10.1016/j.bcp.2023.115579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/23/2023] [Accepted: 04/25/2023] [Indexed: 08/18/2023]
Abstract
With the continuous progress of atherosclerosis research, the significant pathological change of it--vascular calcification (VC), gains increasing attention. In recent years, numerous studies have demonstrated that it is an independent predictor of death risk of cardiovascular disease, and it has a strong correlation with poor clinical prognosis. As the world's population continues to age, the occurrence of VC is expected to reach its highest point in the near future. Therefore, it is essential to investigate ways to prevent or even reverse this process for clinical purposes. Endothelial-to-mesenchymal transition (EndMT) describes the progressive differentiation of endothelial cells into mesenchymal stem cells (MSCs) under various stimuli and acquisition of pluripotent cell characteristics. More and more studies show that EndMT plays a vital role in various cardiovascular diseases, including atherosclerosis, vascular calcification and heart valvular disease. EndMT is also involved in the formation and progression of VC. This review vividly describes the history, characteristics of EndMT and how it affects the endothelial cell process, then focuses on the relationship between vascular endothelium, EndMT, amino acid metabolism, and vascular calcification. Finally, it overviews the signal pathway of EndMT and drugs targeting EndMT, hoping to provide new ideas and a theoretical basis for studying potential therapeutic targets of VC.
Collapse
Affiliation(s)
- Dingkun Lu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Han Jiang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Ting Zou
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Yuanwang Jia
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Yunyun Zhao
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China.
| |
Collapse
|
49
|
Xie R, Liu X, Wu H, Liu M, Zhang Y. Associations between systemic immune-inflammation index and abdominal aortic calcification: Results of a nationwide survey. Nutr Metab Cardiovasc Dis 2023; 33:1437-1443. [PMID: 37156667 DOI: 10.1016/j.numecd.2023.04.015] [Citation(s) in RCA: 44] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/10/2023]
Abstract
BACKGROUND AND AIM The Systemic Immune-Inflammation Index (SII) is a novel index of inflammation assessment that appears to be superior to the common single blood index in the assessment of cardiovascular disease. The purpose of this study was to investigate the association between SII and abdominal aortic calcification (AAC) in adults. METHODS AND RESULTS Multivariate logistic regression, sensitivity analysis, and smoothing curve fitting were used to investigate the relationship between SII and AAC based on data from the National Health and Nutrition Examination Survey (NHANES) 2013-2014. Subgroup analysis and interaction tests were used to investigate whether this association was stable across populations. There was a positive association between SII and ACC in 3036 participants >40 years of age. In the fully adjusted model, each 100-unit increase in SII was associated with a 4% increase in the risk of developing severe AAC [1.04 (1.02, 1.07)]. Participants in the highest quartile of SII had a 47% higher risk of developing severe AAC than those in the lowest quartile [1.47 (1.10, 1.99)]. This positive association was more pronounced in older adults >60 years of age. CONCLUSIONS SII is positively associated with AAC in US adults. Our findings imply that SII has the potential to improve AAC prevention in the general population.
Collapse
Affiliation(s)
- Ruijie Xie
- Department of Hand & Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, China; Division of Clinical Epidemiology and Aging Research, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 581, Heidelberg, 69120, Germany; University of Heidelberg, Heidelberg, 69117, Germany
| | - Xiaozhu Liu
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyang Wu
- Department of Graduate School of Tianjin Medical University, No. 22 Qixiangtai Road, Tianjin, 300070, China; Duke Molecular Physiology Institute, Duke University School of Medicine, Duke University, Durham, NC, USA.
| | - Mingjiang Liu
- Department of Hand & Microsurgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, China.
| | - Ya Zhang
- Department of Gland Surgery, The Affiliated Nanhua Hospital, Hengyang Medical School, University of South China, Hengyang, 421002, China.
| |
Collapse
|
50
|
Immanuel J, Yun S. Vascular Inflammatory Diseases and Endothelial Phenotypes. Cells 2023; 12:1640. [PMID: 37371110 PMCID: PMC10297687 DOI: 10.3390/cells12121640] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 06/06/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
The physiological functions of endothelial cells control vascular tone, permeability, inflammation, and angiogenesis, which significantly help to maintain a healthy vascular system. Several cardiovascular diseases are characterized by endothelial cell activation or dysfunction triggered by external stimuli such as disturbed flow, hypoxia, growth factors, and cytokines in response to high levels of low-density lipoprotein and cholesterol, hypertension, diabetes, aging, drugs, and smoking. Increasing evidence suggests that uncontrolled proinflammatory signaling and further alteration in endothelial cell phenotypes such as barrier disruption, increased permeability, endothelial to mesenchymal transition (EndMT), and metabolic reprogramming further induce vascular diseases, and multiple studies are focusing on finding the pathways and mechanisms involved in it. This review highlights the main proinflammatory stimuli and their effects on endothelial cell function. In order to provide a rational direction for future research, we also compiled the most recent data regarding the impact of endothelial cell dysfunction on vascular diseases and potential targets that impede the pathogenic process.
Collapse
Affiliation(s)
| | - Sanguk Yun
- Department of Biotechnology, Inje University, Gimhae-si 50834, Republic of Korea;
| |
Collapse
|