1
|
Réthoré L, Guihot A, Grimaud L, Proux C, Barré B, Guillonneau F, Guette C, Boissard A, Henry C, Cayon J, Perrot R, Henrion D, Legros C, Legendre C. A Novel Function of Na V Channel β3 Subunit in Endothelial Cell Alignment Through Autophagy Modulation. FASEB J 2025; 39:e70663. [PMID: 40445729 PMCID: PMC12124425 DOI: 10.1096/fj.202401558rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 04/25/2025] [Accepted: 05/14/2025] [Indexed: 06/02/2025]
Abstract
Endothelial cells (EC) play a pivotal role in vascular homeostasis. By sensing shear stress generated by blood flow, EC endorse vasculoprotection through mechanotransduction signaling pathways. Various ion channels are involved in mechanosignaling, and here, we investigated the endothelial voltage-gated Na+ channels (NaV channels), since their mechanosensitivity has been previously demonstrated in cardiomyocytes. First, we showed that EC from aorta (TeloHAEC) behave as EC from umbilical vein (HUVEC) under laminar shear stress (LSS). For both EC models, cell alignment and elongation occurred with the activation of the KLF2/KLF4 atheroprotective signaling pathways. We found that LSS decreased the expression of SCN5A, encoding NaV1.5, while LSS increased that of SCN3B, encoding NaVβ3. We demonstrated that the KLF4 transcription factor is involved in SCN3B expression under both static and LSS conditions. Interestingly, SCN3B silencing impaired EC alignment induced by LSS. The characterization of NaVβ3 interactome by coimmunoprecipitation and proteomic analysis revealed that mTOR, implicated in autophagy, binds to NaVβ3. This result was evidenced by the colocalization between NaVβ3 and mTOR inside cells. Moreover, we showed that SCN3B silencing led to the decrease in LC3B expression and the number of LC3B positive autophagosomes. Furthermore, we showed that NaVβ3 is retained within the cell and colocalized with LAMP1 and LC3B. Finally, we found that resveratrol, a stimulating-autophagy and vasculoprotective molecule, induced KLF4 together with NaVβ3 expression. Altogether, our findings highlight a novel role of NaVβ3 in endothelial function and cell alignment as an actor in shear stress vasculoprotective intracellular pathway through autophagy modulation.
Collapse
Affiliation(s)
- Léa Réthoré
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Anne‐Laure Guihot
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Linda Grimaud
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Coralyne Proux
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Benjamin Barré
- Univ AngersAngersFrance
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
| | - François Guillonneau
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
- INSERM, CNRS, CRCI2NA, Nantes Université, Univ AngersAngersFrance
| | - Catherine Guette
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
- INSERM, CNRS, CRCI2NA, Nantes Université, Univ AngersAngersFrance
| | - Alice Boissard
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
| | - Cécile Henry
- Institut de Cancérologie de l'Ouest (ICO)AngersFrance
- Prot'ICO (ICO Proteomic Core Facility)AngersFrance
| | - Jérôme Cayon
- SFR ICAT, PACeM (Plateforme d'Analyse Cellulaire et Moléculaire), Univ AngersAngersFrance
| | - Rodolphe Perrot
- SFR ICAT, SCIAM (Service Commun d'Imageries et d'Analyses Microscopiques), Univ AngersAngersFrance
| | - Daniel Henrion
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Christian Legros
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| | - Claire Legendre
- INSERM, CNRS, MITOVASC, Equipe CarME, SFR ICAT, Univ AngersAngersFrance
| |
Collapse
|
2
|
Mora Massad K, Dai Z, Petrache I, Ventetuolo CE, Lahm T. Lung endothelial cell heterogeneity in health and pulmonary vascular disease. Am J Physiol Lung Cell Mol Physiol 2025; 328:L877-L884. [PMID: 39772753 PMCID: PMC12116231 DOI: 10.1152/ajplung.00296.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 12/16/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Lung endothelial cells (ECs) are essential for maintaining organ function and homeostasis. Despite sharing some common features with ECs from organ systems, lung ECs exhibit significant heterogeneity in morphology, function, and gene expression. This heterogeneity is increasingly recognized as a key contributor to the development of pulmonary diseases like pulmonary hypertension (PH). In this mini-review, we explore the evolving understanding of lung EC heterogeneity, particularly through the lens of single-cell RNA sequencing (scRNA-seq) technologies. These advances have provided unprecedented insights into the diverse EC subpopulations, their specific roles, and the disturbances in their homeostatic functions that contribute to PH pathogenesis. In particular, these studies identified novel and functionally distinct cell types such as aerocytes and general capillary ECs that are critical for maintaining lung function in health and disease. In addition, multiple novel pathways and mechanisms have been identified that contribute to aberrant pulmonary vascular remodeling in PH. Emerging techniques like single-nucleus RNA sequencing and spatial transcriptomics have further pushed the field forward by discovering novel disease mediators. As research continues to leverage these advanced techniques, the field is poised to uncover novel EC subtypes and disease mechanisms, paving the way for new therapeutic targets in PH and other lung diseases.
Collapse
Grants
- 4IPA1275127 American Heart Association (AHA)
- R01 HL077328 NHLBI NIH HHS
- P01 HL158507 NHLBI NIH HHS
- Reuben M. Chernaick Fellowship
- R01HL169509 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- I01 BX002042 BLRD VA
- R01 HL141268 NHLBI NIH HHS
- Borstein Family Foundation
- R01HL144727 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- Colorado Pulmonary Vascular Disease Award
- R01 HL170096 NHLBI NIH HHS
- R01HL170096 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL158596 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL144727 NHLBI NIH HHS
- HL077328 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01 HL169509 NHLBI NIH HHS
- R01 HL158596 NHLBI NIH HHS
- R01 HL162794 NHLBI NIH HHS
- R01-HL141268 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R01HL62794 HHS | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Karina Mora Massad
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
| | - Zhiyu Dai
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Washington University School of Medicine in St. Louis, Saint Louis, Missouri, United States
| | - Irina Petrache
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
| | - Corey E Ventetuolo
- Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States
- Department of Health Services, Policy and Practice, Brown University, Providence, Rhode Island, United States
| | - Tim Lahm
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado, United States
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, National Jewish Health, Denver, Colorado, United States
- Rocky Mountain Regional VA Medical Center, Aurora, Colorado, United States
| |
Collapse
|
3
|
Gao J, Pan H, Guo X, Huang Y, Luo JY. Endothelial Krüppel-like factor 2/4: Regulation and function in cardiovascular diseases. Cell Signal 2025; 130:111699. [PMID: 40023301 DOI: 10.1016/j.cellsig.2025.111699] [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: 12/02/2024] [Revised: 02/09/2025] [Accepted: 02/20/2025] [Indexed: 03/04/2025]
Abstract
This review presents an overview of the regulation, function, disease-relevance and pharmacological regulation of the critical endothelial transcription factors KLF2/4 in vasculature. The regulatory mechanisms of KLF2/4 expression and activity in vascular endothelium in response to hemodynamic forces and biochemical stimuli are depicted. The functional effects mediated by direct or indirect target genes of KLF2/4 in endothelial cells are systematically summarized. The contributory roles that dysregulated KLF2/4 play in relevant cardiovascular pathologies, such as atherosclerotic vascular lesions, pulmonary arterial hypertension and vascular complications of diabetes were reviewed. Moreover, this review also discusses the pharmacological regulation of KLF2/4 by drugs used in clinics and therapeutic possibility by directly targeting these two transcription factors for treating atherosclerotic cardiovascular diseases. Finally, prospective opinions on the gaps in disclosing novel vascular function mediated by KLF2/4 and future research needs are expressed.
Collapse
Affiliation(s)
- Jing Gao
- Department of Cardiology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Hongjie Pan
- Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital Affiliated to Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaogang Guo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong Special Administrative Region, China.
| | - Jiang-Yun Luo
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Strohm L, Mihalikova D, Czarnowski A, Schwaibold Z, Daiber A, Stamm P. Sex-Specific Antioxidant and Anti-Inflammatory Protective Effects of AMPK in Cardiovascular Diseases. Antioxidants (Basel) 2025; 14:615. [PMID: 40427496 PMCID: PMC12108612 DOI: 10.3390/antiox14050615] [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] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/02/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Cardiovascular diseases such as coronary heart disease, heart failure, or stroke are the most common cause of death worldwide and are regularly based on risk factors like diabetes mellitus, hypertension, or obesity. At the same time, both diseases and risk factors are significantly influenced by sex hormones. In order to better understand this influence and also specifically improve the therapy of female patients, medical research has recently focused increasingly on gender-specific differences. The goal is to develop personalized, gender-specific therapy concepts for these diseases to further enhance health outcomes. The enzyme adenosine monophosphate-activated protein kinase (AMPK) is a central regulator of energy metabolism, protecting the cardiovascular system from energy depletion, thereby promoting vascular health and preventing cellular damage. AMPK confers cardioprotective effects by preventing endothelial and vascular dysfunction, and by controlling or regulating oxidative stress and inflammatory processes. For AMPK, sex-specific effects were reported, influencing metabolic and cardiovascular responses. Exercise and metabolic stress generally cause higher AMPK activity in males. At the same time, females exhibit protective mechanisms against insulin resistance or oxidative stress, particularly in conditions like obesity. Additionally, males subject to AMPK deficiency seem to experience greater cardiac and mitochondrial dysfunction. In contrast, females show improvement in cardiovascular function after pharmacological AMPK activation. These differences, influenced by hormones, body composition, and gene expression, highlight the potential to develop personalized, sex-specific AMPK-targeted therapeutic strategies for cardiovascular diseases in the future. Here, we discuss the most actual scientific background, focusing on the protective, gender-specific effects of AMPK, and highlight potential clinical applications.
Collapse
Affiliation(s)
- Lea Strohm
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (L.S.); (D.M.); (A.C.); (Z.S.)
| | - Dominika Mihalikova
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (L.S.); (D.M.); (A.C.); (Z.S.)
| | - Alexander Czarnowski
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (L.S.); (D.M.); (A.C.); (Z.S.)
| | - Zita Schwaibold
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (L.S.); (D.M.); (A.C.); (Z.S.)
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (L.S.); (D.M.); (A.C.); (Z.S.)
- German Center for Cardiovascular Research (DZHK), Partnersite Rhine-Main, 55131 Mainz, Germany
| | - Paul Stamm
- Department of Cardiology, Cardiology I, University Medical Center of the Johannes Gutenberg-University, 55131 Mainz, Germany; (L.S.); (D.M.); (A.C.); (Z.S.)
| |
Collapse
|
5
|
Zhu J, Hotchkiss HL, Shores KL, Truskey GA, Maskarinec SA. Strategies for improved endothelial cell adhesion in microphysiological vascular model systems. PLoS One 2025; 20:e0323080. [PMID: 40388439 PMCID: PMC12088046 DOI: 10.1371/journal.pone.0323080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/02/2025] [Indexed: 05/21/2025] Open
Abstract
Human tissue-engineered blood vessels (TEBVs) have been applied as model systems to study a wide range of vascular diseases including Hutchinson-Gilford Progeria Syndrome and early atherosclerosis. Central to the utility of TEBVs as an in vitro blood vessel model is the maintenance of a functional endothelium under physiologically relevant shear stresses. Establishing and maintaining a confluent endothelial monolayer is challenging. In this protocol, we outline an optimized procedure for the endothelialization of TEBVs. We optimized the following key conditions affecting endothelial cell (EC) adherence in the vessel: EC seeding density, rotation time, and the application of perfusion. This protocol results in TEBVs with sustained EC luminal coverage that demonstrate alignment in the direction of applied flow and responsiveness to inflammatory stimuli. To facilitate rapid screening of EC coverage during the fabrication and perfusion steps, we re-designed TEBV chambers to include a viewing window that allows for efficient monitoring and assessment of the endothelialization process using fluorescence microscopy. By identifying key factors that affect EC attachment in TEBVs, this protocol may serve as a valuable resource for researchers seeking to achieve successful endothelialization of engineered blood vessel constructs.
Collapse
Affiliation(s)
- Jingyi Zhu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Halie L. Hotchkiss
- Division of Infectious Diseases, Duke University Health System, Durham, North Carolina, United States of America
| | - Kevin L. Shores
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - George A. Truskey
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Stacey A. Maskarinec
- Division of Infectious Diseases, Duke University Health System, Durham, North Carolina, United States of America
| |
Collapse
|
6
|
Wang M, Preckel B, Zuurbier CJ, Weber NC. Effects of SGLT2 inhibitors on ion channels in heart failure: focus on the endothelium. Basic Res Cardiol 2025:10.1007/s00395-025-01115-y. [PMID: 40366385 DOI: 10.1007/s00395-025-01115-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 05/06/2025] [Accepted: 05/09/2025] [Indexed: 05/15/2025]
Abstract
Heart failure (HF) is a life-threatening cardiovascular disease associated with high mortality, diminished quality of life, and a significant economic burden on both patients and society. The pathogenesis of HF is closely related to the endothelium, where endothelial ion channels play an important role in regulating intracellular Ca2+ signals. These ion channels are essential to maintain vascular function, including endothelium-dependent vascular tone, inflammation response, and oxidative stress. Sodium-glucose cotransporter 2 inhibitors (SGLT2i) have shown promising cardiovascular benefits in HF patients, reducing mortality risk and hospitalization in several large clinical trials. Clinical and preclinical studies indicate that the cardioprotective effects of SGLT2i in HF are mediated by endothelial nitric oxide (NO) pathways, as well as by reducing inflammation and reactive oxygen species in cardiac endothelial cells. Additionally, SGLT2i may confer endothelial protection by lowering intracellular Ca2+ level through the inhibition of sodium-hydrogen exchanger 1 (NHE1) and sodium-calcium exchanger (NCX) in endothelial cells. In this review, we discuss present knowledge regarding the expression and role of Ca2+-related ion channels in endothelial cells in HF, focusing on the effects of SGLT2i on endothelial NHE1, NCX as well as on vascular tone.
Collapse
Affiliation(s)
- Mengnan Wang
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Benedikt Preckel
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands
| | - Nina C Weber
- Department of Anesthesiology - Laboratory of Experimental Intensive Care and Anesthesiology-L.E.I.C.A, Amsterdam University Medical Centers, Amsterdam Cardiovascular Science, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
7
|
Nagi JS, Doiron AL. 20 nm nanoparticles trigger calcium influx to endothelial cells via a TRPV4 channel. Biomater Sci 2025; 13:2728-2743. [PMID: 40192740 DOI: 10.1039/d4bm01691b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/14/2025]
Abstract
While increased intracellular calcium (Ca2+) has been identified as a key effect of nanoparticles on endothelial cells, the mechanism has not been fully elucidated or examined under shear stress. Here, we show the effect of several types of 20 nm particles on Ca2+ in the presence of shear stress in human umbilical vein endothelial cells (HUVECs), human coronary artery endothelial cells (HCAECs), and human cardiac microvascular endothelial cells (HMVEC-Cs). Intracellular Ca2+ levels increased by nearly three-fold in these cell types upon exposure to 100 μg mL-1 20 nm Au particles, which was not seen in response to larger or smaller particles. An antagonist to the calcium channel - transient receptor potential vanilloid-type 4 (TRPV4) - drastically reduced the amount of calcium by 9.3-fold in HUVECs exposed to 0.6 Pa shear stress and 100 μg mL-1 20 nm gold particles, a trend upheld in both HCAECs and HMVEC-Cs. Cell alignment in the direction of fluid flow is a well-known phenomenon in endothelial cells, and interestingly, cells in the presence of 20 nm particles with fluid flow had a higher alignment index than cells in the fluid flow alone. When compared with previous works, these results indicated that 20 nm particles may be inducing endothelial permeability by activating the TRPV4 channel in vitro. The potential of nanoparticle delivery technologies hinges on an improved understanding of this effect toward improved delivery with limited toxicity.
Collapse
Affiliation(s)
- Jaspreet Singh Nagi
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, VT 05405, USA.
| | - Amber L Doiron
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, VT 05405, USA.
| |
Collapse
|
8
|
Luo P, Huang M, Ye Y, Wang R, Yan W, Zhu L, Liu S, Tang Y, Liu K, Gao W. Effects of high-intensity interval training vs. moderate-intensity continuous training on arterial stiffness in adults: A systematic review and meta-analysis of randomized controlled trials. Arch Gerontol Geriatr 2025; 136:105890. [PMID: 40382987 DOI: 10.1016/j.archger.2025.105890] [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: 03/16/2025] [Revised: 04/15/2025] [Accepted: 05/04/2025] [Indexed: 05/20/2025]
Abstract
OBJECTIVE Arterial stiffness (AS) is regarded as an independent predictor of cardiovascular events and all-cause mortality, and it is significantly associated with global mortality rates. Physical activity (PA) plays a positive role in reducing AS and improving cardiovascular health. The aim of this study is to compare the differences between high-intensity interval training (HIIT) and moderate-intensity continuous training (MICT) in their effects on reducing AS. METHODS We conducted a comprehensive search of the PubMed, Cochrane, Embase, Web of Science, and EBSCO electronic databases, covering the period from their inception to January 10, 2025. We used a fixed-effect model to compare the changes in pulse wave velocity (PWV) before and after intervention between the HIIT group and the MICT group. Data were reported using the weighted mean difference (WMD) and 95 % confidence interval (95 % CI). RESULTS This study included 619 participants from 22 studies. Compared to MICT, HIIT demonstrated a more significant reduction in PWV (-0.10 m/s [95 % CI:0.16 to -0.03], P = 0.005). Additionally, we found that HIIT was superior in reducing CF-PWV (-0.10 m/s [95 % CI:0.17 to -0.02], P = 0.01). CONCLUSION HIIT is more effective than MICT in improving PWV and promoting arterial health in adults.
Collapse
Affiliation(s)
- Ping Luo
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Mingxuan Huang
- College of Music and Dance, South-Central Minzu University, Wuhan, China
| | - Yufang Ye
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Ruixue Wang
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Weiyi Yan
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Lin Zhu
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Shanqi Liu
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Yao Tang
- School of Physical Education, Wuhan Sports University, Wuhan, China
| | - Kang Liu
- School of Physical Education and Health, Nanning Normal University, Nanning, China
| | - Weifeng Gao
- School of Physical Education, Wuhan Sports University, Wuhan, China.
| |
Collapse
|
9
|
Meecham A, McCurdy S, Frias-Anaya E, Li W, Gallego-Gutierrez H, Nguyen P, Li YS, Chien S, Shyy JYJ, Ginsberg MH, Lopez-Ramirez MA. Silencing KRIT1 Partially Reverses the Effects of Disturbed Flow on the Endothelial Cell Transcriptome. Int J Mol Sci 2025; 26:4340. [PMID: 40362576 PMCID: PMC12072803 DOI: 10.3390/ijms26094340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Revised: 04/19/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Endothelial cells respond to forces generated by laminar blood flow with changes in vasodilation, anticoagulant, fibrinolytic, or anti-inflammatory functions which preserve vessel patency. These responses to flow shear stress are primarily mediated by the modulation of the following transcription factors: Krüppel-like factors 2 and 4 (KLF2 and KLF4). Notably, disturbed flow patterns, which are found in vascular areas predisposed to atherosclerosis, significantly reduce the endothelial expression of KLF2 and KLF4, resulting in changes in the transcriptome that exacerbate inflammation and thrombosis. The endothelial CCM (Cerebral Cavernous Malformation) complex, comprising KRIT1 (Krev1 interaction trapped gene 1), CCM2 (Malcavernin), and CCM3 (Programmed cell death protein 10), suppresses the expression of KLF2 and KLF4. Loss of function of the CCM complex has recently been suggested to protect from coronary atherosclerosis in humans. We thus hypothesized that the silencing of KRIT1, the central scaffold of the CCM complex, can normalize the atherogenic effects of disturbed flow on the human endothelial transcriptome. Bulk RNA sequencing (RNA-seq) was conducted on human umbilical vein endothelial cells (HUVECs) after the expression of KRIT1 was silenced using specific small interfering RNA (siRNA). The endothelial cells were exposed to three different conditions for 24 h, as follows: pulsatile shear stress (laminar flow), oscillatory shear stress (disturbed flow), and static conditions (no flow). We found that silencing the KRIT1 expression in HUVECs restored the expression of the transcription factors KLF2 and KLF4 under oscillatory shear stress. This treatment resulted in a transcriptomic profile similar to that of endothelial cells under pulsatile shear stress. These findings suggest that inhibition of the CCM complex in endothelium plays a vasoprotective role by reactivating a protective gene program to help endothelial cells resist disturbed blood flow. Targeting CCM genes can activate well-known vasoprotective gene programs that enhance endothelial resilience to inflammation, hypoxia, and angiogenesis under disturbed flow conditions, providing a novel pathway for preventing atherothrombosis.
Collapse
Affiliation(s)
- Amelia Meecham
- Department of Medicine, University of California, La Jolla, CA 92093, USA; (A.M.); (S.M.); (E.F.-A.); (W.L.); (H.G.-G.)
| | - Sara McCurdy
- Department of Medicine, University of California, La Jolla, CA 92093, USA; (A.M.); (S.M.); (E.F.-A.); (W.L.); (H.G.-G.)
| | - Eduardo Frias-Anaya
- Department of Medicine, University of California, La Jolla, CA 92093, USA; (A.M.); (S.M.); (E.F.-A.); (W.L.); (H.G.-G.)
| | - Wenqing Li
- Department of Medicine, University of California, La Jolla, CA 92093, USA; (A.M.); (S.M.); (E.F.-A.); (W.L.); (H.G.-G.)
| | - Helios Gallego-Gutierrez
- Department of Medicine, University of California, La Jolla, CA 92093, USA; (A.M.); (S.M.); (E.F.-A.); (W.L.); (H.G.-G.)
| | - Phu Nguyen
- Department of Bioengineering, University of California, La Jolla, CA 92093, USA; (P.N.); (Y.-S.L.); (S.C.); (J.Y.-J.S.)
| | - Yi-Shuan Li
- Department of Bioengineering, University of California, La Jolla, CA 92093, USA; (P.N.); (Y.-S.L.); (S.C.); (J.Y.-J.S.)
| | - Shu Chien
- Department of Bioengineering, University of California, La Jolla, CA 92093, USA; (P.N.); (Y.-S.L.); (S.C.); (J.Y.-J.S.)
| | - John Y.-J. Shyy
- Department of Bioengineering, University of California, La Jolla, CA 92093, USA; (P.N.); (Y.-S.L.); (S.C.); (J.Y.-J.S.)
| | - Mark H. Ginsberg
- Department of Medicine, University of California, La Jolla, CA 92093, USA; (A.M.); (S.M.); (E.F.-A.); (W.L.); (H.G.-G.)
| | - Miguel Alejandro Lopez-Ramirez
- Department of Medicine, University of California, La Jolla, CA 92093, USA; (A.M.); (S.M.); (E.F.-A.); (W.L.); (H.G.-G.)
- Department of Pharmacology, University of California, La Jolla, CA 92093, USA
| |
Collapse
|
10
|
Jang J, Park HJ. Epigenetic Regulation of Endothelial-Mesenchymal Transition in Vascular Atherosclerosis Development. Korean Circ J 2025; 55:456-458. [PMID: 40306732 PMCID: PMC12088988 DOI: 10.4070/kcj.2025.0006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 03/03/2025] [Accepted: 03/09/2025] [Indexed: 05/02/2025] Open
Affiliation(s)
- JaeHyuk Jang
- Division of Cardiology, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Uijeongbu, Korea
| | - Hun-Jun Park
- Division of Cardiology, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Uijeongbu, Korea.
| |
Collapse
|
11
|
Kelly MP, Nikolaev VO, Gobejishvili L, Lugnier C, Hesslinger C, Nickolaus P, Kass DA, Pereira de Vasconcelos W, Fischmeister R, Brocke S, Epstein PM, Piazza GA, Keeton AB, Zhou G, Abdel-Halim M, Abadi AH, Baillie GS, Giembycz MA, Bolger G, Snyder G, Tasken K, Saidu NEB, Schmidt M, Zaccolo M, Schermuly RT, Ke H, Cote RH, Mohammadi Jouabadi S, Roks AJM. Cyclic nucleotide phosphodiesterases as drug targets. Pharmacol Rev 2025; 77:100042. [PMID: 40081105 DOI: 10.1016/j.pharmr.2025.100042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/13/2025] [Indexed: 03/15/2025] Open
Abstract
Cyclic nucleotides are synthesized by adenylyl and/or guanylyl cyclase, and downstream of this synthesis, the cyclic nucleotide phosphodiesterase families (PDEs) specifically hydrolyze cyclic nucleotides. PDEs control cyclic adenosine-3',5'monophosphate (cAMP) and cyclic guanosine-3',5'-monophosphate (cGMP) intracellular levels by mediating their quick return to the basal steady state levels. This often takes place in subcellular nanodomains. Thus, PDEs govern short-term protein phosphorylation, long-term protein expression, and even epigenetic mechanisms by modulating cyclic nucleotide levels. Consequently, their involvement in both health and disease is extensively investigated. PDE inhibition has emerged as a promising clinical intervention method, with ongoing developments aiming to enhance its efficacy and applicability. In this comprehensive review, we extensively look into the intricate landscape of PDEs biochemistry, exploring their diverse roles in various tissues. Furthermore, we outline the underlying mechanisms of PDEs in different pathophysiological conditions. Additionally, we review the application of PDE inhibition in related diseases, shedding light on current advancements and future prospects for clinical intervention. SIGNIFICANCE STATEMENT: Regulating PDEs is a critical checkpoint for numerous (patho)physiological conditions. However, despite the development of several PDE inhibitors aimed at controlling overactivated PDEs, their applicability in clinical settings poses challenges. In this context, our focus is on pharmacodynamics and the structure activity of PDEs, aiming to illustrate how selectivity and efficacy can be optimized. Additionally, this review points to current preclinical and clinical evidence that depicts various optimization efforts and indications.
Collapse
Affiliation(s)
- Michy P Kelly
- Department of Neurobiology, Center for Research on Aging, University of Maryland School of Medicine, Baltimore, Maryland
| | - Viacheslav O Nikolaev
- Institute of Experimental Cardiovascular Research, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Leila Gobejishvili
- Department of Physiology, School of Medicine, University of Louisville, Kentucky, Louisville
| | - Claire Lugnier
- Translational CardioVascular Medicine, CRBS, UR 3074, Strasbourg, France
| | | | - Peter Nickolaus
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - David A Kass
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Rodolphe Fischmeister
- Université Paris-Saclay, Inserm, Signaling and Cardiovascular Pathophysiology, UMR-S 1180, Orsay, France
| | - Stefan Brocke
- Department of Immunology, UConn Health, Farmington, Connecticut
| | - Paul M Epstein
- Department of Cell Biology, UConn Health, Farmington, Connecticut
| | - Gary A Piazza
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Adam B Keeton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, Alabama
| | - Gang Zhou
- Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Mohammad Abdel-Halim
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - Ashraf H Abadi
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy and Biotechnology, German University in Cairo, Cairo, Egypt
| | - George S Baillie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark A Giembycz
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | | | - Gretchen Snyder
- Molecular Neuropharmacology, Intra-Cellular Therapies Inc (ITI), New York, New York
| | - Kjetil Tasken
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway; Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Nathaniel E B Saidu
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
| | - Martina Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands; Groningen Research Institute for Asthma and COPD, GRIAC, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Manuela Zaccolo
- Department of Physiology, Anatomy and Genetics and National Institute for Health and Care Research Oxford Biomedical Research Centre, University of Oxford, Oxford, United Kingdom
| | - Ralph T Schermuly
- Department of internal Medicine, Justus Liebig University of Giessen, Giessen, Germany
| | - Hengming Ke
- Department of Biochemistry and Biophysics, The University of North Carolina, Chapel Hill, North Carolina
| | - Rick H Cote
- Department of Molecular, Cellular, and Biomedical Sciences, University of New Hampshire, Durham, New Hampshire
| | - Soroush Mohammadi Jouabadi
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands
| | - Anton J M Roks
- Section of Vascular and Metabolic Disease, Department of Internal Medicine, Erasmus MC University Medical Center, Erasmus University Rotterdam, Rotterdam, The Netherlands.
| |
Collapse
|
12
|
Yan L, Zhou Z, Chen S, Feng X, Mao J, Luo F, Zhu J, Chen X, Hu Y, Wang Y, Wu B, Du L, Wang C, Gong L, Zhu Y. Reshaping the chromatin landscape in HUVECs from small-for-gestational-age newborns. JCI Insight 2025; 10:e186812. [PMID: 40260916 PMCID: PMC12038915 DOI: 10.1172/jci.insight.186812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 03/11/2025] [Indexed: 04/24/2025] Open
Abstract
Small for gestational age (SGA), with increased risk of adult-onset cardiovascular diseases and metabolic syndromes, is known to associate with endothelial dysfunction, but the pathogenic mechanisms remain unclear. In this study, the pathological state of human umbilical vein endothelial cells (HUVECs) from SGA individuals was characterized by presenting increased angiogenesis, migration, proliferation, and wound healing ability relative to their normal counterparts. Genome-wide mapping of transcriptomes and open chromatins unveiled global gene expression alterations and chromatin remodeling in SGA-HUVECs. Specifically, we revealed increased chromatin accessibility at active enhancers, along with dysregulation of genes associated with angiogenesis, and further identified CD44 as the key gene driving HUVECs' dysfunction by regulating pro-angiogenic genes' expression and activating phosphorylated ERK1/2 and phosphorylated endothelial NOS expression in SGA. In SGA-HUVECs, CD44 was abnormally upregulated by 3 active enhancers that displayed increased chromatin accessibility and interacted with CD44 promoter. Subsequent motif analysis uncovered activating protein-1 (AP-1) as a crucial transcription factor regulating CD44 expression by binding to CD44 promoter and associated enhancers. Enhancers CRISPR interference and AP-1 inhibition restored CD44 expression and alleviated the hyperangiogenesis of SGA-HUVECs. Together, our study provides a foundational understanding of the epigenetic alterations driving pathological angiogenesis and offers potential therapeutic insights into addressing endothelial dysfunction in SGA.
Collapse
Affiliation(s)
- Lingling Yan
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | - Xin Feng
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Junwen Mao
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fang Luo
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianfang Zhu
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiuying Chen
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yingying Hu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Yuan Wang
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Bingbing Wu
- Department of Obstetrics and Gynecology, Center for Reproductive Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, China
| | - Lizhong Du
- Department of Pediatrics, Children’s Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Chunlin Wang
- Department of Pediatrics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Liang Gong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, China
| | | |
Collapse
|
13
|
Grego A, Fernandes C, Fonseca I, Dias-Neto M, Costa R, Leite-Moreira A, Oliveira SM, Trindade F, Nogueira-Ferreira R. Endothelial dysfunction in cardiovascular diseases: mechanisms and in vitro models. Mol Cell Biochem 2025:10.1007/s11010-025-05289-w. [PMID: 40259179 DOI: 10.1007/s11010-025-05289-w] [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: 02/09/2025] [Accepted: 04/08/2025] [Indexed: 04/23/2025]
Abstract
Endothelial cells (ECs) are arranged side-by-side to create a semi-permeable monolayer, forming the inner lining of every blood vessel (micro and macrocirculation). Serving as the first barrier for circulating molecules and cells, ECs represent the main regulators of vascular homeostasis being able to respond to environmental changes, either physical or chemical signals, by producing several factors that regulate vascular tone and cellular adhesion. Healthy endothelium has anticoagulant properties that prevent the adhesion of leukocytes and platelets to the vessel walls, contributing to resistance to thrombus formation, and regulating inflammation, and vascular smooth muscle cell proliferation. Many risk factors of cardiovascular diseases (CVDs) promote the endothelial expression of chemokines, cytokines, and adhesion molecules. The resultant endothelial activation can lead to endothelial cell dysfunction (ECD). In vitro models of ECD allow the study of cellular and molecular mechanisms of disease and provide a research platform for screening potential therapeutic agents. Even though alternative models are available, such as animal models or ex vivo models, in vitro models offer higher experimental flexibility and reproducibility, making them a valuable tool for the understanding of pathophysiological mechanisms of several diseases, such as CVDs. Therefore, this review aims to synthesize the currently available in vitro models regarding ECD, emphasizing CVDs. This work will focus on 2D cell culture models (endothelial cell lines and primary ECs), 3D cell culture systems (scaffold-free and scaffold-based), and 3D cell culture models (such as organ-on-a-chip). We will dissect the role of external stimuli-chemical and mechanical-in triggering ECD.
Collapse
Affiliation(s)
- Ana Grego
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Cristiana Fernandes
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Ivo Fonseca
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Marina Dias-Neto
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Angiology and Vascular Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Raquel Costa
- Universidade Católica Portuguesa, CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, 4169-005, Porto, Portugal
| | - Adelino Leite-Moreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
- Department of Cardiothoracic Surgery, Unidade Local de Saúde de São João, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Sandra Marisa Oliveira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Fábio Trindade
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal
| | - Rita Nogueira-Ferreira
- RISE-Health, Department of Surgery and Physiology, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319, Porto, Portugal.
| |
Collapse
|
14
|
Bridges CA, Fu L, Yeow J, Huang X, Jackson M, Kuchel RP, Sterling JD, Baker SM, Lord MS. The interplay between endothelial glycocalyx maturity and both the toxicity and intracellular uptake of charged nanoparticles. Acta Biomater 2025; 196:293-306. [PMID: 40058617 DOI: 10.1016/j.actbio.2025.03.012] [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: 10/29/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 04/05/2025]
Abstract
Nanoparticles are widely studied for delivering treatments to target tissues, but few have reached clinical use. Most nanoparticles encounter blood vessels on their way to target tissues. The inner surface of these vessels is lined with endothelial cells covered by a glycocalyx, an extracellular matrix rich in anionic glycans. The role of the glycocalyx in nanoparticle interactions is not well understood. Here, we demonstrate that endothelial cells need extended culture times to synthesize a mature glycocalyx. Our research shows that branched polyethyleneimine functionalized gold nanoparticles bind to endothelial cells expressing either a developing or mature glycocalyx, with the interaction involving hyaluronan and heparan sulfate. These nanoparticles are subsequently internalized. Similar results were seen with poly(L-arginine). A mature glycocalyx protects cells by reducing the toxicity of these cationic nanoparticles. In contrast, lipoic acid-functionalized gold nanoparticles are internalized by cells with a developing glycocalyx, but not a mature one. Poly(L-glutamic acid) only interacts with cells when major glycans in the glycocalyx are degraded. These findings highlight the complex relationship between nanoparticle charge and structure, and their effects on toxicity, binding, and uptake by endothelial cells. This offers important insights for improving nanoparticle interactions with blood vessels in health and disease. STATEMENT OF SIGNIFICANCE: Endothelial cells lining blood vessels form a barrier through which nanoparticles must cross to reach target tissues. These cells are covered with a layer called the glycocalyx, which is rich in anionic glycans. However, the role of the glycocalyx in how nanoparticles interact with cells remains underexplored. Our research revealed that cells with a mature glycocalyx internalize cationic nanoparticles and experience reduced cytotoxicity. Conversely, a mature glycocalyx prevents anionic nanoparticles from entering cells. These results suggest that the structure of both the nanoparticles and the glycocalyx should be considered in future studies to improve the use of nanoparticles for medical applications.
Collapse
Affiliation(s)
- Claire A Bridges
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Lu Fu
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Jonathan Yeow
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Xiaojing Huang
- Molecular Surface Interaction Laboratory, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - Miriam Jackson
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
| | - Rhiannon P Kuchel
- Electron Microscope Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW 2052, Australia
| | - James D Sterling
- College of Innovation, Entrepreneurship, and Economic Development, Missouri University of Science and Technology, Rolla, MO 65409, USA
| | | | - Megan S Lord
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia.
| |
Collapse
|
15
|
Shen S, Pan T, Liu P, Tian Y, Shi Y, Zhu W. The mechanisms and applications of endothelial progenitor cell therapy in the treatment of intracranial aneurysm. J Transl Med 2025; 23:377. [PMID: 40148864 PMCID: PMC11951544 DOI: 10.1186/s12967-025-06401-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
The pathophysiological mechanism of intracranial aneurysm (IA) involves the dynamic interaction of ECM abnormalities, hemodynamic stress, and inflammatory response. The rupture of intracranial aneurysm will cause serious consequences. Multiple studies have confirmed the important role and potential application of endothelial progenitor cells (EPCs) in vascular repair. This review focuses on the specific mechanism of EPCs in the treatment of intracranial aneurysms, which promote re-endothelialization and angiogenesis through bone marrow mobilization, targeted migration to the site of injury, differentiation into mature endothelial cells, and secretion of angiogenic factors. In addition, EPCs maintain ECM homeostasis by regulating MMP/IMP balance, inhibiting aneurysm wall thinning and structural damage. Based on the vascular repair mechanism of EPCs, new treatment strategies such as "biologically active" spring coils (loaded with EPCs or SDF-1α) and flow diverters(FDs) combined with EPCs therapy have been developed to synergistically promote carotid endothelialization of aneurysms and reduce the risk of recurrence. Future research needs to further validate the long-term efficacy and precise regulatory mechanisms of EPCs in clinical translation, providing new directions for IA treatment.
Collapse
Affiliation(s)
- Shiyu Shen
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Tonglin Pan
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Peixi Liu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yanlong Tian
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China
- National Center for Neurological Disorders, Shanghai, China
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China
- Neurosurgical Institute of Fudan University, Shanghai, China
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China
| | - Yuan Shi
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| | - Wei Zhu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai, China.
- National Center for Neurological Disorders, Shanghai, China.
- Shanghai Key Laboratory of Brain Function and Restoration and Neural Regeneration, Shanghai, China.
- Neurosurgical Institute of Fudan University, Shanghai, China.
- Shanghai Clinical Medical Center of Neurosurgery, Shanghai, China.
| |
Collapse
|
16
|
Yang L, Li X, Ni L, Lin Y. Treatment of endothelial cell dysfunction in atherosclerosis: a new perspective integrating traditional and modern approaches. Front Physiol 2025; 16:1555118. [PMID: 40206381 PMCID: PMC11979162 DOI: 10.3389/fphys.2025.1555118] [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: 01/03/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Atherosclerosis (AS), a prime causative factor in cardiovascular disease, originates from endothelial cell dysfunction (ECD). Comprising a vital part of the vascular endothelium, endothelial cells play a crucial role in maintaining vascular homeostasis, optimizing redox balance, and regulating inflammatory responses. More evidence shows that ECD not only serves as an early harbinger of AS but also exhibits a strong association with disease progression. In recent years, the treatment strategies for ECD have been continuously evolving, encompassing interventions ranging from lifestyle modifications to traditional pharmacotherapy aimed at reducing risk factors, which also have demonstrated the ability to improve endothelial cell function. Additionally, novel strategies such as promising biotherapy and gene therapy have drawn attention. These methods have demonstrated enormous potential and promising prospects in improving endothelial function and reversing AS. However, it is essential to remain cognizant that the current treatments still present significant challenges regarding therapeutic efficacy, long-term safety, and ethical issues. This article aims to provide a systematic review of these treatment methods, analyze the mechanisms and efficacy of various therapeutic strategies, with the goal of offering insights and guidance for clinical practice, and further advancing the prevention and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
| | | | | | - Yuanyuan Lin
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
17
|
Calvier L, Wasser CR, Solow EB, Wu S, Evers BM, Karp DS, Kounnas MZ, Herz J. Genetic or therapeutic disruption of the Reelin/Apoer2 signaling pathway improves inflammatory arthritis outcomes. Proc Natl Acad Sci U S A 2025; 122:e2418642122. [PMID: 40073057 PMCID: PMC11929474 DOI: 10.1073/pnas.2418642122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 02/07/2025] [Indexed: 03/14/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by synovial inflammation, pannus formation, and progressive joint destruction. The inflammatory milieu in RA drives endothelial cell activation and upregulation of adhesion molecules, thus facilitating leukocyte infiltration into the synovium. Reelin, a circulating glycoprotein previously implicated in endothelial activation and leukocyte recruitment in diseases such as atherosclerosis and multiple sclerosis, has emerged as a potential upstream regulator of these processes. However, its role in RA pathogenesis remains poorly understood. Here, we demonstrate that Reelin levels are markedly elevated in the plasma of both RA patients and mouse models of arthritis, with higher concentrations correlating with greater disease severity. Genetic deletion of the Reelin receptor Apoer2 conferred significant protection against serum transfer arthritis (STA), underscoring the relevance of this pathway in disease progression. Furthermore, therapeutic inhibition of Reelin using the CR-50 antibody yielded robust anti-inflammatory effects in multiple preclinical arthritis models, including STA, K/BxN, and collagen-induced arthritis. Notably, CR-50 treatment not only reduced leukocyte infiltration and synovial inflammation but also mitigated pannus formation. Importantly, these benefits were achieved without the gastrointestinal side effects commonly associated with nonsteroidal anti-inflammatory drugs like diclofenac. Our findings position Reelin as a proinflammatory endothelial biomarker and therapeutic target in RA. By modulating endothelial activation and leukocyte recruitment, anti-Reelin strategies offer an alternative approach to attenuate synovial inflammation and joint damage. These results provide a compelling rationale for further exploration of Reelin-targeted therapies as alternatives to conventional immunosuppressive treatments in RA and other chronic inflammatory diseases.
Collapse
MESH Headings
- Reelin Protein
- Animals
- Cell Adhesion Molecules, Neuronal/metabolism
- Cell Adhesion Molecules, Neuronal/genetics
- Cell Adhesion Molecules, Neuronal/antagonists & inhibitors
- Cell Adhesion Molecules, Neuronal/blood
- Cell Adhesion Molecules, Neuronal/immunology
- Extracellular Matrix Proteins/metabolism
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/antagonists & inhibitors
- Extracellular Matrix Proteins/blood
- Extracellular Matrix Proteins/immunology
- Serine Endopeptidases/metabolism
- Serine Endopeptidases/genetics
- Serine Endopeptidases/blood
- Serine Endopeptidases/immunology
- Nerve Tissue Proteins/metabolism
- Nerve Tissue Proteins/genetics
- Nerve Tissue Proteins/antagonists & inhibitors
- Nerve Tissue Proteins/blood
- Nerve Tissue Proteins/immunology
- Mice
- Signal Transduction
- Humans
- Arthritis, Rheumatoid/genetics
- Arthritis, Rheumatoid/metabolism
- Arthritis, Rheumatoid/pathology
- Arthritis, Rheumatoid/drug therapy
- LDL-Receptor Related Proteins/genetics
- LDL-Receptor Related Proteins/metabolism
- Arthritis, Experimental/genetics
- Arthritis, Experimental/pathology
- Mice, Knockout
- Inflammation
- Female
- Male
- Mice, Inbred C57BL
- Disease Models, Animal
Collapse
Affiliation(s)
- Laurent Calvier
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
| | - Catherine R. Wasser
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
| | - E. Blair Solow
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
| | - Sharon Wu
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
| | - Bret M. Evers
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
| | - David S. Karp
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
| | | | - Joachim Herz
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- O’Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX75390-9046
| |
Collapse
|
18
|
Meecham A, McCurdy S, Frias-Anaya E, Li W, Gallego-Gutierrez H, Ngyuen P, Li JYS, Chien S, Shyy JYJ, Ginsberg MH, Lopez-Ramirez MA. Silencing KRIT1 Partially Reverses the Effects of Disturbed Flow on the Endothelial Cell Transcriptome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.12.642862. [PMID: 40161739 PMCID: PMC11952409 DOI: 10.1101/2025.03.12.642862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Background Endothelial cells respond to forces generated by laminar blood flow with changes in vasodilation, anticoagulant, fibrinolytic, or anti-inflammatory functions which preserve vessel patency. These responses to flow sheer stress are primarily mediated by the modulation of transcription factors Krüppel-like factors 2 and 4 (KLF2 and KLF4). Notably, disturbed flow patterns, which are found in vascular areas predisposed to atherosclerosis, significantly reduce the endothelial expression of KLF2 and KLF4, resulting in changes in the transcriptome that exacerbate inflammation and thrombosis. The endothelial CCM complex, comprising KRIT1, CCM2, and CCM3, suppresses the expression of KLF2 and KLF4. Loss of function of the CCM complex has recently been suggested to protect from coronary atherosclerosis in humans. We thus hypothesized that silencing of KRIT1, the central scaffold of the CCM complex, can normalize the atherogenic effects of disturbed flow on the human endothelial transcriptome. Methods Bulk RNA sequencing (RNA-seq) was conducted on human umbilical vein endothelial cells (HUVECs) after the expression of KRIT1 was silenced using specific siRNAs. The endothelial cells were exposed to three different conditions for 24 hours: pulsatile shear stress (laminar flow), oscillatory shear stress (disturbed flow), and static conditions (no flow). Results We found that silencing KRIT1 expression in HUVECs restored the expression of the transcription factors KLF2 and KLF4 under oscillatory shear stress. This treatment resulted in a transcriptomic profile similar to that of endothelial cells under pulsatile shear stress. These findings suggest that inhibition of the CCM complex in endothelium plays a vasoprotective role by reactivating a protective gene program to help endothelial cells resist disturbed blood flow. Conclusions Targeting CCM genes can activate well-known vasoprotective gene programs that enhance endothelial resilience to inflammation, hypoxia, and angiogenesis under disturbed flow conditions, providing a novel pathway for preventing atherosclerosis.
Collapse
Affiliation(s)
- Amelia Meecham
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Sara McCurdy
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Eduardo Frias-Anaya
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Wenqing Li
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | | | - Phu Ngyuen
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Julie Yi-Shuan Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - Shu Chien
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093
| | - John Y.-J. Shyy
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Mark H. Ginsberg
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Miguel A. Lopez-Ramirez
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
- Department of Pharmacology, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
19
|
Guo L, Yuan Y, Zheng F, Zhan C, Li X. Computational Design and In Vitro and In Vivo Characterization of an ApoE-Based Synthetic High-Density Lipoprotein for Sepsis Therapy. Biomolecules 2025; 15:397. [PMID: 40149933 PMCID: PMC11940477 DOI: 10.3390/biom15030397] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/27/2025] [Accepted: 03/04/2025] [Indexed: 03/29/2025] Open
Abstract
Introduction: Septic patients have low levels of high-density lipoproteins (HDLs), which is a risk factor. Replenishing HDLs with synthetic HDLs (sHDLs) has shown promise as a therapy for sepsis. This study aimed to develop a computational approach to design and test new types of sHDLs for sepsis treatment. Methods: We used a three-step computational approach to design sHDL nanoparticles based on the structure of HDLs and their binding to endotoxins. We tested the efficacy of these sHDLs in two sepsis mouse models-cecal ligation and puncture (CLP)-induced and P. aeruginosa-induced sepsis models-and assessed their impact on inflammatory signaling in cells. Results: We designed four sHDL nanoparticles: two based on the ApoA-I sequence (YGZL1 and YGZL2) and two based on the ApoE sequence (YGZL3 and YGZL4). We demonstrated that an ApoE-based sHDL nanoparticle, YGZL3, provides effective protection against CLP- and P. aeruginosa-induced sepsis. The sHDLs effectively suppressed inflammatory signaling in HEK-blue or RAW264 cells. Conclusions: Unlike earlier approaches, we developed a new approach that employs computational simulations to design a new type of sHDL based on HDL's structure and function. We found that YGZL3, an ApoE sequence-based sHDL, provides effective protection against sepsis in two mouse models.
Collapse
Affiliation(s)
- Ling Guo
- Saha Cardiovascular Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
| | - Yaxia Yuan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Fang Zheng
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Changguo Zhan
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
- Molecular Modeling and Biopharmaceutical Center, College of Pharmacy, University of Kentucky, Lexington, KY 40536, USA
| | - Xiangan Li
- Saha Cardiovascular Research Center, Department of Physiology, College of Medicine, University of Kentucky, Lexington, KY 40536, USA
- Lexington VA Health Care System, Lexington, KY 40502, USA
| |
Collapse
|
20
|
Vasilieva ME, Kashchenko VA, Shmidt EV, Mizeva IA, Dolotovskaya PM, Zaytsev VV, Margaryants NB, Kamshilin AA. Improvement of Microvascular Function in Patients with Morbid Obesity After Bariatric Surgery Revealed by Imaging Photoplethysmography. Obes Surg 2025; 35:1001-1008. [PMID: 39934520 DOI: 10.1007/s11695-025-07741-8] [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: 11/23/2024] [Revised: 01/29/2025] [Accepted: 02/04/2025] [Indexed: 02/13/2025]
Abstract
BACKGROUND Morbid obesity leads to serious complications such as diabetes mellitus, arterial hypertension, and atherosclerosis. Bariatric surgery aims to reduce body weight and correct metabolic disorders and associated macro- and microvascular dysfunction. Metabolic disturbances can be assessed by biochemical markers, whereas microvascular function can be assessed by the response to provocative stimuli. The aim of this work is to quantitatively assess the cutaneous microcirculatory change caused by bariatric surgery using imaging photoplethysmography. METHODS The study included 20 patients with obesity whose body mass index (BMI) was greater than 40 kg/m2 who underwent bariatric surgery and 20 control subjects. Microvascular function was assessed twice, before and 6 months after surgery, by measuring the perfusion response to local forearm heating up to 40 °C using imaging photoplethysmography. RESULTS The perfusion response to local heating was found to be significantly lower in patients with obesity before surgery compared to the control group, but 6 months after surgery, it approaches indicators of the control group, along with a decrease in BMI from 48 ± 5 to 36 ± 5 kg/m2, P < 0.001. Besides, bariatric surgery improves biochemical markers of metabolic syndrome (glycated hemoglobin decreases from 6.3 ± 1.0 to 5.2 ± 0.4% and cholesterol from 5.2 ± 1.4 to 4.2 ± 0.8 mmol/l). CONCLUSION Based on these results, we conclude that microvascular abnormalities caused by obesity could be repaired after bariatric surgery and subsequent conservative treatment.
Collapse
Affiliation(s)
- Maria E Vasilieva
- North-Western District Scientific and Clinical Center Named After L.G. Sokolov, FMBA, Saint Petersburg, Russian Federation
| | - Victor A Kashchenko
- North-Western District Scientific and Clinical Center Named After L.G. Sokolov, FMBA, Saint Petersburg, Russian Federation
- Saint Petersburg State University, Saint Petersburg, Russian Federation
- Beloostrov Clinic, Vsevolozhsk District, Leningrad Region, Russian Federation
| | - Elena V Shmidt
- North-Western District Scientific and Clinical Center Named After L.G. Sokolov, FMBA, Saint Petersburg, Russian Federation
- Saint Petersburg State University, Saint Petersburg, Russian Federation
| | - Irina A Mizeva
- Institute of Continuous Media Mechanics, Perm, Russian Federation
| | | | - Valery V Zaytsev
- North-Western District Scientific and Clinical Center Named After L.G. Sokolov, FMBA, Saint Petersburg, Russian Federation
| | | | - Alexei A Kamshilin
- North-Western District Scientific and Clinical Center Named After L.G. Sokolov, FMBA, Saint Petersburg, Russian Federation.
- Institute of Automation and Control Processes, Primorskiy, Russian Federation.
| |
Collapse
|
21
|
Yao Y, Huang V, Seah V, Kim JE. Impact of Quantity and Type of Dietary Protein on Cardiovascular Disease Risk Factors Using Standard and Network Meta-analyses of Randomized Controlled Trials. Nutr Rev 2025; 83:e814-e828. [PMID: 39013196 PMCID: PMC11819482 DOI: 10.1093/nutrit/nuae086] [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: 07/18/2024] Open
Abstract
CONTEXT Higher protein diets (HPDs) have shown favorable outcomes on weight maintenance and body-composition management; however, their protective effects against cardiovascular diseases (CVDs) remain uncertain and contentious. Furthermore, it is important to consider the influence of other macronutrients in the diet and type of dietary protein when studying HPDs, because this aspect has been overlooked in previous studies. OBJECTIVE We assessed the impacts of quantity and type of dietary protein on CVD risk factors. DATA SOURCES A database search was conducted in PubMed, Embase, CINAHL, Web of Science, and Cochrane Library and a total of 100 articles met the eligibility criteria. DATA EXTRACTION Extracted data from 100 articles were analyzed using standard meta-analysis, and 41 articles were also analyzed using network meta-analysis. DATA ANALYSIS In the standard meta-analysis, an HPD had significant favorable effects on systolic blood pressure (SBP) (mean difference [MD] = -1.51 mmHg; 95% CI: -2.77, -0.25), diastolic blood pressure (DBP) (MD = -1.08 mmHg; 95% CI: -1.81, -0.35), and flow-mediated dilation (MD = 0.78%; 95% CI: 0.09, 1.47) compared with lower protein diets. The further network meta-analysis supported that the high-protein, high-carbohydrate, low-fat diet was the most recommended diet to ensure a maximum decrease in SBP, DBP, total cholesterol (TC), and low-density-lipoprotein cholesterol (LDL-C). In comparison to animal-protein-rich diets, plant-protein-rich diets (PPRs) exhibited a significant favorable effects on improving TC (MD = -0.12 mmol/L; 95% CI: -0.19, -0.05), triglyceride (MD = -0.05 mmol/L; 95% CI: -0.09, -0.01), LDL-C (MD = -0.11 mmol/L; 95% CI: -0.18, -0.04), and high-density-lipoprotein cholesterol (MD = 0.03 mmol/L; 95% CI: 0.02, 0.04) levels. CONCLUSION Consumption of HPDs and PPRs supports improvements in vascular health and lipid-lipoprotein profiles, respectively. Furthermore, macronutrient composition should be carefully designed in the dietary approach to maximize the effectiveness of HPDs in improving CVD risk factors. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42022369931.
Collapse
Affiliation(s)
- Yueying Yao
- Department of Food Science and Technology, National University of Singapore, 117543 Singapore
| | - Vanessa Huang
- Department of Food Science and Technology, National University of Singapore, 117543 Singapore
| | - Vernice Seah
- Department of Food Science and Technology, National University of Singapore, 117543 Singapore
| | - Jung Eun Kim
- Department of Food Science and Technology, National University of Singapore, 117543 Singapore
| |
Collapse
|
22
|
Blankenship AE, Kemna R, Kueck PJ, John C, Vitztum M, Yoksh L, Mahnken JD, Vidoni ED, Morris JK, Geiger PC. Improving glycemic control via heat therapy in older adults at risk for Alzheimer's disease (FIGHT-AD): a pilot study. J Appl Physiol (1985) 2025; 138:720-730. [PMID: 39829076 DOI: 10.1152/japplphysiol.00396.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/26/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
Impaired glycemic control increases the risk of type 2 diabetes (T2D) and Alzheimer's disease (AD). Heat therapy (HT), via hot water immersion (HWI), has shown promise in improving shared mechanisms implicated in both T2D and AD, like blood glucose regulation, insulin sensitivity, and inflammation. The potential for HT to improve brain health in individuals at risk for AD has not been examined. This pilot study aimed to assess the feasibility and adherence of using HT in cognitively healthy older individuals at risk for AD due to existing metabolic risk factors. Participants underwent 4 wk of HT (three sessions/week) via HWI, alongside cognitive screening, self-reported sleep characterization, glucose tolerance tests, and MRI scans pre- and postintervention. A total of 18 participants (9 males, 9 females; mean age: 71.1 ± 3.9 yr), demonstrating metabolic risk, completed the intervention. Participant adherence for the study was 96% (8 missed sessions out of 216 total sessions), with one study-related mild adverse event (mild dizziness/nausea). Overall, the research participants responded to a postintervention survey saying they enjoyed participating in the study and it was not a burden on their schedules. Secondary outcomes of the HT intervention demonstrated significant changes in mean arterial pressure, diastolic blood pressure, and cerebral blood flow (P < 0.05), with a trend toward improved body mass index (P = 0.06). Future studies, including longer durations and a thermoneutral control group, are needed to fully understand heat therapy's impact on glucose homeostasis and the potential to improve brain health.NEW & NOTEWORTHY Our pilot study demonstrated promising results for heat therapy (HT) via hot water immersion in older adults at risk for Alzheimer's disease due to metabolic factors. Despite a relatively short intervention, significant improvements in mean arterial pressure, diastolic blood pressure, and cerebral blood flow postintervention were observed. High participant adherence, overall satisfaction, and minimal adverse events suggest HT's feasibility. These findings highlight HT's potential as an effective alternative intervention for cardiometabolic dysfunction in at-risk populations.
Collapse
Affiliation(s)
- Anneka E Blankenship
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Riley Kemna
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Paul J Kueck
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Casey John
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Michelle Vitztum
- KU Diabetes Institute, Department of Internal Medicine, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Lauren Yoksh
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Jonathan D Mahnken
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Biostatistics and Data Science, University of Kansas Medical Center, Kansas City, Kansas, United States
- Frontiers Clinical and Translational Science Institute, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Eric D Vidoni
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Jill K Morris
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Neurology, University of Kansas Medical Center, Kansas City, Kansas, United States
| | - Paige C Geiger
- University of Kansas Alzheimer's Disease Research Center, University of Kansas Medical Center, Fairway, Kansas, United States
- Department of Cell Biology and Physiology, University of Kansas Medical Center, Kansas City, Kansas, United States
| |
Collapse
|
23
|
Costa ADSD, Vadym K, Park K. Engineered endothelium model enables recapitulation of vascular function and early atherosclerosis development. Biomaterials 2025; 314:122889. [PMID: 39423515 DOI: 10.1016/j.biomaterials.2024.122889] [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: 04/30/2024] [Revised: 10/07/2024] [Accepted: 10/09/2024] [Indexed: 10/21/2024]
Abstract
Human health relies heavily on the vascular endothelium. Here, we propose a novel engineered endothelium model (EEM), which recapitulated both normal vascular function and pathology. An artificial basement membrane (aBM), where porous polyvinyl alcohol hydrogel was securely integrated with human fibroblast-derived, decellularized extracellular matrix on both sides was fabricated first and followed by endothelial cells (ECs) and pericytes (PCs) adhesion, respectively. Our EEM formed robust adherens junction (VE-cad) and built an impermeable barrier with time, along with the nitric oxide (NO) secretion. In our EEM, ECs and PCs interacted each other via aBM and led to hemoglobin alpha 1 (Hb-α1) development, which was involved in NO control and was strongly interconnected with VE-cad as well. A resilient property of EEM under inflammatory milieu was also confirmed by VE-cad and barrier recovery with time. In particular interest, foam cells formation, a hallmark of atherosclerotic initiation was successfully recapitulated in our EEM, where a series of sequential events were confirmed: human monocytes adhesion, transendothelial migration, and oxidized low-density lipoprotein uptake by macrophages. Collectively, our EEM is excellent in recapitulating not only normal endothelium but early pathologic one, thereby enabling EEM to be a physiologically relevant model for vascular study and disease modeling.
Collapse
Affiliation(s)
| | - Kopych Vadym
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
| | - Kwideok Park
- Center for Biomaterials, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea.
| |
Collapse
|
24
|
Sullivan AE, Courvan MCS, Aday AW, Wasserman DH, Niswender KD, Shardelow EM, Wells EK, Wells QS, Freiberg MS, Beckman JA. The Role of Serum Free Fatty Acids in Endothelium-Dependent Microvascular Function. Endocrinol Diabetes Metab 2025; 8:e70031. [PMID: 39888728 PMCID: PMC11784902 DOI: 10.1002/edm2.70031] [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: 10/09/2024] [Revised: 12/28/2024] [Accepted: 01/11/2025] [Indexed: 02/02/2025] Open
Abstract
BACKGROUND Elevated serum free fatty acid (FFA) concentration is associated with insulin resistance and is a hallmark of metabolic syndrome. A pathological feature of insulin resistance is impaired endothelial function. OBJECTIVE To investigate the effect of FFA reduction with either acipimox, a nicotinic acid derivative that impairs lipolysis, or salsalate, a salicylate that reduces basal and inflammation-induced lipolysis, on insulin-mediated endothelium-dependent vasodilation. METHODS This was a post hoc, combined analysis of two randomised, double-blind, placebo-controlled crossover trials. Sixteen subjects were recruited (6 with metabolic syndrome and 10 controls) and randomised to acipimox 250 mg orally every 6 h for 7 days or placebo. Nineteen subjects were recruited (13 with metabolic syndrome and 6 controls) and randomised to receive salsalate 4.5 g/day for 4 weeks or placebo. The primary outcome was the association between FFA concentration and insulin-mediated vasodilation, measured by venous-occlusion strain-gauge plethysmography at baseline and following FFA modulation with the study drugs. RESULTS At baseline, FFA concentration (R = -0.35, p = 0.043) and insulin sensitivity (HOMA-IR: R = -0.42, p = 0.016, Adipo-IR: R = -0.39, p = 0.025) predicted insulin-mediated vasodilation. FFA levels were significantly reduced after drug pretreatment (0.604 vs. 0.491 mmol/L, p = 0.036) while insulin levels, insulin sensitivity and inflammatory markers were unchanged. Despite a reduction in circulating FFA with drug therapy, neither insulin-stimulated vasodilation nor insulin sensitivity improved. CONCLUSIONS Short-term reduction of FFA concentration does not improve insulin-stimulated vasodilation in patients with metabolic syndrome. TRIAL REGISTRATION ClinicalTrials.gov identifier: NCT00759291 and NCT00760019 (formerly NCT00762827).
Collapse
Affiliation(s)
- Alexander E. Sullivan
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | | | - Aaron W. Aday
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - David H. Wasserman
- Department of Molecular Physiology and BiophysicsVanderbilt University School of MedicineNashvilleTennesseeUSA
| | - Kevin D. Niswender
- Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Department of Veteran AffairsTennessee Valley Healthcare SystemNashvilleTennesseeUSA
| | - Emily M. Shardelow
- Vanderbilt University Medical CenterProgram for Metabolic Bone DisordersNashvilleTennesseeUSA
| | - Emily K. Wells
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Quinn S. Wells
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Vanderbilt Translational and Clinical Cardiovascular Research Center, Division of Cardiovascular MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Matthew S. Freiberg
- Division of Cardiovascular Medicine, Department of MedicineVanderbilt University Medical CenterNashvilleTennesseeUSA
- Geriatric Research Education and Clinical Centers (GRECC)Veterans Affairs Tennessee Valley Healthcare SystemNashvilleTennesseeUSA
| | - Joshua A. Beckman
- Division of Vascular Medicine, Department of MedicineUniversity of Texas SouthwesternDallasTexasUSA
| |
Collapse
|
25
|
Tao M, Shan L, Zhang W, Wei L, Guo M, Fang Z, Zhao J, Gao J. METTL3-mediated m6A modification of EGR1 mRNA promotes T2DM vasculopathy. Cell Signal 2025; 127:111564. [PMID: 39674391 DOI: 10.1016/j.cellsig.2024.111564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/11/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024]
Abstract
Vascular endothelial dysfunction is one of the leading causes of developing vascular lesions in Type 2 diabetes mellitus (T2DM). In the development of vascular lesions, when endothelial cells are stimulated by hyperglycemia, inflammation and other external conditions, endothelial cell dysfunction will occur, which promotes endothelial cells to lose its typical phenotype and gain mesenchymal characteristics, with the occurrence of endothelial-to-mesenchymal transition (EndMT). At the same time promote endothelial cell proliferation and migration, induce vascular injury. m6A methylation modification enzyme METTL3 is involved in the development of vascular lesions in T2DM. However, the mechanisms by which METTL3 is involved in T2DM vascular lesions are unclear. In this study, we induced T2DM vascular lesions in human umbilical vein endothelial cells (HUVECs) mimicking high glucose and TNF-α (H + T) levels. The effects of METTL3 on HUVECs EndMT, proliferation and migration have been revealed. Protein expression of endothelial calmodulin (VE-Cadherin) and smooth muscle actin (α-SMA) was visualised by western blot and immunofluorescence techniques to evaluate the occurrence of EndMT. In addition, MeRIP-seq revealed a METTL3-mediated m6A modification profile. MeRIP-qPCR combined with m6A site prediction verified the methylation levels of downstream targets and identified EGR1 as a target of METTL3. Activation of EGR1 successfully rescued EndMT in METTL3-deficient HUVECs. In summary, targeting METTL3 may become an important molecular target for intervention in diabetic vascular lesions.
Collapse
Affiliation(s)
- Meng Tao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China; College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - Li Shan
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China
| | - Wei Zhang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei 230011, Anhui, China
| | - LiangBing Wei
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China
| | - MingFei Guo
- Department of Scientific Research, The First Affiliated Hospital of Anhui Medical University, Hefei 230012, Anhui, China; Anhui Public Health Clinical Center, Hefei 230012, Anhui, China
| | - ZhaoHui Fang
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China; Diabetes Prevention Institute of Traditional Chinese Medicine, Anhui Academy of Traditional Chinese Medicine, Hefei 230031, Anhui, China
| | - JinDong Zhao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China; Diabetes Prevention Institute of Traditional Chinese Medicine, Anhui Academy of Traditional Chinese Medicine, Hefei 230031, Anhui, China
| | - JiaRong Gao
- Department of Pharmacy, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, Anhui, China.
| |
Collapse
|
26
|
Van Eijgen J, Van Winckel L, Hanssen H, Kotliar K, Vanassche T, Van Craenenbroeck EM, Cornelissen V, Van Craenenbroeck AH, Jones E, Stalmans I. Retinal vessel analysis to assess microvascular function in the healthy eye: A systematic review on the response to acute physiological and pathological stressors. Surv Ophthalmol 2025; 70:200-214. [PMID: 39592075 DOI: 10.1016/j.survophthal.2024.11.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024]
Abstract
The retina allows noninvasive in vivo assessment of the microcirculation. Autoregulation of the retinal microvasculature meets the changing requirements of local metabolic demand and maintains adequate blood flow. Analysis of the retinal vascular reactivity contributes to the understanding of regulatory physiology and its relationship to the systemic microcirculation. We conducted a literature review on the effect of different acute stimuli onto the retinal vasculature was conducted in accordance with the PRISMA guidelines. A literature search between 1-1-2005 and 17-10-2022 was performed in Medline, Embase, Web of Science and the Cochrane Library. We report the retinal vascular behavior of healthy individuals in response to both physiological and pathological stressors in 106 included articles. We provide ables of methodological characteristics for each stressor. Hypoxia, hypercapnia, high altitude, flicker light stimulation, rise of core temperature, blood pressure lowering, and the condition immediately after endurance exercise associate with larger retinal vessels. Hyperoxia, hypocapnia, blood pressure rise (Bayliss effect), and the condition during isometric exercise associate with smaller retinal vessels. The retinal vasculature is highly reactive to physiological and pathological stressors. This autoregulatory capacity is hypothesized to be a source of biomarkers for vascular health. Dynamic and static retinal vessel analysis are noninvasive methods to assess this (micro)vascular function. Exploring its diagnostic potential and application into clinical practice requires the development of standardized assessment methods, for which some recommendations are made.
Collapse
Affiliation(s)
- Jan Van Eijgen
- Department of Ophthalmology, University Hospitals UZ Leuven, Leuven, Belgium; Research Group Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium.
| | - Lien Van Winckel
- Department of Ophthalmology, University Hospitals UZ Leuven, Leuven, Belgium.
| | - Henner Hanssen
- Department of Sports, Exercise and Health, Medical Faculty, University of Basel, Basel, Switzerland.
| | - Konstantin Kotliar
- Department of Medical Engineering and Technomathematics, FH Aachen University of Applied Sciences, Campus Jülich, Heinrich-Mussmann-Str. 1, Jülich 52428, Germany.
| | - Thomas Vanassche
- Centre for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium.
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR Department, University of Antwerp, Campus Drie Eiken D.T.2.28, Universiteitsplein 1, Antwerp 2610, Belgium; Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem 2650, Belgium.
| | - Véronique Cornelissen
- Research Group of Rehabilitation of Internal Disorders, Department of Rehabilitation Sciences, Faculty of Movement and Rehabilitation Sciences, KU Leuven, Leuven, Belgium.
| | - Amaryllis H Van Craenenbroeck
- Division of Nephrology, University Hospitals UZ leuven, Leuven, Belgium; Nephrology and Renal Transplantation Research Group, Department of Microbiology, Immunology, and Transplantation, KU Leuven, Leuven, Belgium.
| | - Elisabeth Jones
- Centre for Molecular and Vascular Biology, Herestraat 49, Bus 911, KU, Leuven 3000, Belgium; Department of Cardiology, CARIM School for Cardiovascular Diseases, Maastricht University, Universiteitssingel 50, Maastricht 6229 ER, Netherlands.
| | - Ingeborg Stalmans
- Department of Ophthalmology, University Hospitals UZ Leuven, Leuven, Belgium; Research Group Ophthalmology, Department of Neurosciences, KU Leuven, Leuven, Belgium.
| |
Collapse
|
27
|
Li K, Feng J, Li M, Han L, Wu Y. Systematic Review of Interleukin-35 in Endothelial Dysfunction: A New Target for Therapeutic Intervention. Mediators Inflamm 2025; 2025:2003124. [PMID: 39974277 PMCID: PMC11839265 DOI: 10.1155/mi/2003124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 01/21/2025] [Indexed: 02/21/2025] Open
Abstract
Endothelial dysfunction is a significant factor in the pathogenesis of various diseases. In pathological states, endothelial cells (ECs) undergo activation, resulting in dysfunction characterized by the stimulation of inflammatory responses, oxidative stress, cell proliferation, blood coagulation, and vascular adhesions. Interleukin-35 (IL-35), a novel member of the IL-12 family, is primarily secreted by regulatory T cells (Tregs) and regulatory B cells (Bregs). The role of IL-35 in immunomodulation, antioxidative stress, resistance to apoptosis, control of EC activation, adhesion, and angiogenesis in ECs remains incompletely understood, as the specific mechanisms of IL-35 action and its regulation have yet to be fully elucidated. Therefore, this systematic review aims to comprehensively investigate the impact of IL-35 on ECs and their physiological roles in a range of conditions, including cardiovascular diseases, tumors, sepsis, and rheumatoid arthritis (RA), with the objective of elucidating the potential of IL-35 as a therapeutic target for these ailments.
Collapse
Affiliation(s)
- Kai Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Jie Feng
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Meng Li
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Leilei Han
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| | - Yanqing Wu
- Department of Cardiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, No. 1 Minde Road, Nanchang 330006, Jiangxi, China
| |
Collapse
|
28
|
Ramírez-Melo LM, Estrada-Luna D, Rubio-Ruiz ME, Castañeda-Ovando A, Fernández-Martínez E, Jiménez-Osorio AS, Pérez-Méndez Ó, Carreón-Torres E. Relevance of Lipoprotein Composition in Endothelial Dysfunction and the Development of Hypertension. Int J Mol Sci 2025; 26:1125. [PMID: 39940892 PMCID: PMC11817739 DOI: 10.3390/ijms26031125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/18/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Endothelial dysfunction and chronic inflammation are determining factors in the development and progression of chronic degenerative diseases, such as hypertension and atherosclerosis. Among the shared pathophysiological characteristics of these two diseases is a metabolic disorder of lipids and lipoproteins. Therefore, the contents and quality of the lipids and proteins of lipoproteins become the targets of therapeutic objective. One of the stages of lipoprotein formation occurs through the incorporation of dietary lipids by enterocytes into the chylomicrons. Consequently, the composition, structure, and especially the properties of lipoproteins could be modified through the intake of bioactive compounds. The objective of this review is to describe the roles of the different lipid and protein components of lipoproteins and their receptors in endothelial dysfunction and the development of hypertension. In addition, we review the use of some non-pharmacological treatments that could improve endothelial function and/or prevent endothelial damage. The reviewed information contributes to the understanding of lipoproteins as vehicles of regulatory factors involved in the modulation of inflammatory and hemostatic processes, the attenuation of oxidative stress, and the neutralization of toxins, rather than only cholesterol and phospholipid transporters. For this review, a bibliographic search was carried out in different online metabases.
Collapse
Affiliation(s)
- Lisette Monsibaez Ramírez-Melo
- Nutrition Academic Area Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico;
| | - Diego Estrada-Luna
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - María Esther Rubio-Ruiz
- Department of Physiology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Tlalpan, Mexico City 14080, Mexico;
| | - Araceli Castañeda-Ovando
- Chemistry Academic Area, Instituto de Ciencias Básicas e Ingeniería, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Eduardo Fernández-Martínez
- Medicine Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Pachuca 42039, Hidalgo, Mexico;
| | - Angélica Saraí Jiménez-Osorio
- Nursing Academic Area, Instituto de Ciencias de la Salud, Universidad Autónoma del Estado de Hidalgo, Circuito Ex Hacienda La Concepción S/N, Carretera Pachuca-Actopan, San Agustín Tlaxiaca 42160, Hidalgo, Mexico; (D.E.-L.); (A.S.J.-O.)
| | - Óscar Pérez-Méndez
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
- Tecnológico de Monterrey, Campus Ciudad de México, Mexico City 14380, Mexico
| | - Elizabeth Carreón-Torres
- Department of Molecular Biology, Instituto Nacional de Cardiología Ignacio Chávez, Juan Badiano 1, Mexico City 14080, Mexico;
| |
Collapse
|
29
|
Rooney M, Lambe J, O'Connor A, Dunne S, Mills A, Feeney EL, Gibney ER. Bovine dairy products and flow mediated dilation (FMD): a systematic review of the published evidence. Eur J Nutr 2025; 64:66. [PMID: 39853454 PMCID: PMC11761514 DOI: 10.1007/s00394-024-03574-w] [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: 03/25/2024] [Accepted: 12/17/2024] [Indexed: 01/26/2025]
Abstract
PURPOSE Evidence suggests bovine dairy products may have neutral or beneficial effects on cardiometabolic health, despite being a source of saturated fat. The dairy matrix, the structure and combination of protein, fat, and other nutrients, and how they interact with each other, is purported to be responsible for these beneficial health effects. Whether this relationship extends to endothelial function, as assessed by flow mediated dilation (FMD), remains to be elucidated. METHODS Three electronic databases (PubMed, Embase and Cochrane Central) were searched from inception until 5th September 2024. This review included randomised controlled trials (RCT) investigating any bovine dairy intervention which considered endothelial function using FMD in humans with a non-dairy or alternative dairy control. RESULTS Of 4,220 records identified, 18 reports from 11 RCT including 508 (53.3% male) participants, examined endothelial function by FMD and were eligible for evidence synthesis. Eight papers reported an improvement, nine reported no effect and one reported a decrease in FMD. The greatest effects were found in those with impaired health at baseline, with whey protein and high dairy intakes observed to be most beneficial. CONCLUSION Bovine dairy intake has neutral or beneficial effects on cardiometabolic health. This review demonstrates that this relationship extends to endothelial function as assessed by FMD. Whey protein and high dairy intakes may be most effective, although further high quality RCT in this area are warranted.
Collapse
Affiliation(s)
- Martina Rooney
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- Institute of Food and Health, School of Agriculture and Food Sciences, University College Dublin, Dublin 4, Republic of Ireland
| | - Joyce Lambe
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- Institute of Food and Health, School of Agriculture and Food Sciences, University College Dublin, Dublin 4, Republic of Ireland
| | - Aileen O'Connor
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- Institute of Food and Health, School of Agriculture and Food Sciences, University College Dublin, Dublin 4, Republic of Ireland
| | - Simone Dunne
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- Institute of Food and Health, School of Agriculture and Food Sciences, University College Dublin, Dublin 4, Republic of Ireland
| | - Andrea Mills
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- Institute of Food and Health, School of Agriculture and Food Sciences, University College Dublin, Dublin 4, Republic of Ireland
| | - Emma L Feeney
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland
- Institute of Food and Health, School of Agriculture and Food Sciences, University College Dublin, Dublin 4, Republic of Ireland
| | - Eileen R Gibney
- Food for Health Ireland, University College Dublin, Dublin 4, Republic of Ireland.
- Institute of Food and Health, School of Agriculture and Food Sciences, University College Dublin, Dublin 4, Republic of Ireland.
| |
Collapse
|
30
|
Shi M, Sun L, Wei J, Shen Y, Wang J, Zhang P, Yang X, Ding Y, Yin W, Lu X, Yang X, Wang G, Li R. Quercetin alleviates endothelial dysfunction in preeclampsia by inhibiting ferroptosis and inflammation through EGFR binding. Commun Biol 2025; 8:90. [PMID: 39833490 PMCID: PMC11747637 DOI: 10.1038/s42003-025-07547-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 01/14/2025] [Indexed: 01/22/2025] Open
Abstract
Preeclampsia (PE) is a pregnancy-specific complication and there remains no effective treatment. Given the limitations on medication use during pregnancy, exploring natural, safe, and effective drugs for PE is worthwhile. We investigate the causal relationship between ferroptosis, inflammation, and PE, and determine the protective effects of quercetin (QCT), a representative compound that is classified as a flavanol, against endothelial dysfunction. Then, the target of QCT is predicted and verified. The prophylactic addition of a low dose of QCT rescues endothelial dysfunction, aiding in endothelial repair. Furthermore, QCT alleviates PE-like maternal manifestations and endothelial dysfunction in the placenta of the selective reduced uteroplacental perfusion (sRUPP) rat model through binding to the epidermal growth factor receptor (EGFR). The potential applications of QCT are expanded, offering the possibility of further development as a safe and effective preventive molecule for PE.
Collapse
Affiliation(s)
- Meiting Shi
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
| | - Lu Sun
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Jiachun Wei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Yao Shen
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China
| | - Jian Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Ping Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaofeng Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Yuzhen Ding
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Wanchang Yin
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xinyao Lu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xuesong Yang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China.
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China.
- Clinical Research Center, Clifford Hospital, Guangzhou, China.
| | - Guang Wang
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, School of Medicine, Jinan University, Guangzhou, China.
- Key Laboratory for Regenerative Medicine of the Ministry of Education, Jinan University, Guangzhou, China.
- Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Guangdong Second Provincial General Hospital, School of Medicine, Jinan University, Guangzhou, China.
| | - Ruiman Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
31
|
Hong S, Lee DB, Yoon DW, Yoo SL, Kim J. The Effect of Sleep Disruption on Cardiometabolic Health. Life (Basel) 2025; 15:60. [PMID: 39860000 PMCID: PMC11766988 DOI: 10.3390/life15010060] [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: 12/17/2024] [Revised: 01/03/2025] [Accepted: 01/04/2025] [Indexed: 01/27/2025] Open
Abstract
Sleep disruption has emerged as a significant public health concern with profound implications for metabolic health. This review synthesizes current evidence demonstrating the intricate relationships between sleep disturbances and cardiometabolic dysfunction. Epidemiological studies have consistently demonstrated that insufficient sleep duration (<7 h) and poor sleep quality are associated with increased risks of obesity, type 2 diabetes, and cardiovascular disease. The underlying mechanisms are multifaceted, involving the disruption of circadian clock genes, alterations in glucose and lipid metabolism, the activation of inflammatory pathways, and the modulation of the gut microbiome. Sleep loss affects key metabolic regulators, including AMPK signaling and disrupts the secretion of metabolic hormones such as leptin and ghrelin. The latest evidence points to the role of sleep-induced changes in the composition and function of gut microbiota, which may contribute to metabolic dysfunction through modifications in the intestinal barrier and inflammatory responses. The NLRP3 inflammasome and NF-κB signaling pathways have been identified as crucial mediators linking sleep disruption to metabolic inflammation. An understanding of these mechanisms has significant implications for public health and clinical practice, suggesting that improving sleep quality could be an effective strategy for preventing and treating cardiometabolic disorders in modern society.
Collapse
Affiliation(s)
- SeokHyun Hong
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.); (D.-B.L.); (S.-L.Y.)
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea
| | - Da-Been Lee
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.); (D.-B.L.); (S.-L.Y.)
| | - Dae-Wui Yoon
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.); (D.-B.L.); (S.-L.Y.)
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea
| | - Seung-Lim Yoo
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.); (D.-B.L.); (S.-L.Y.)
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea
| | - Jinkwan Kim
- Sleep Medicine Institute, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea; (S.H.); (D.-B.L.); (S.-L.Y.)
- Department of Biomedical Laboratory Science, College of Health Science, Jungwon University, Goesan-gun 28204, Chungcheongbuk-do, Republic of Korea
| |
Collapse
|
32
|
Sun WT, Du JY, Wang J, Wang YL, Dong ED. Potential preservative mechanisms of cardiac rehabilitation pathways on endothelial function in coronary heart disease. SCIENCE CHINA. LIFE SCIENCES 2025; 68:158-175. [PMID: 39395086 DOI: 10.1007/s11427-024-2656-6] [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: 03/18/2024] [Accepted: 06/17/2024] [Indexed: 10/14/2024]
Abstract
Cardiac rehabilitation, a comprehensive exercise-based lifestyle and medical management, is effective in decreasing morbidity and improving life quality in patients with coronary heart disease. Endothelial function, an irreplaceable indicator in coronary heart disease progression, is measured by various methods in traditional cardiac rehabilitation pathways, including medicinal treatment, aerobic training, and smoking cessation. Nevertheless, studies on the effect of some emerging cardiac rehabilitation programs on endothelial function are limited. This article briefly reviewed the endothelium-beneficial effects of different cardiac rehabilitation pathways, including exercise training, lifestyle modification and psychological intervention in patients with coronary heart disease, and related experimental models, and summarized both uncovered and potential cellular and molecular mechanisms of the beneficial roles of various cardiac rehabilitation pathways on endothelial function. In exercise training and some lifestyle interventions, the enhanced bioavailability of nitric oxide, increased circulating endothelial progenitor cells (EPCs), and decreased oxidative stress are major contributors to preventing endothelial dysfunction in coronary heart disease. Moreover, the preservation of endothelial-dependent hyperpolarizing factors and inflammatory suppression play roles. On the one hand, to develop more endothelium-protective rehabilitation methods in coronary heart disease, adequately designed and sized randomized multicenter clinical trials should be advanced using standardized cardiac rehabilitation programs and existing assessment methods. On the other hand, additional studies using suitable experimental models are warranted to elucidate the relationship between some new interventions and endothelial protection in both macro- and microvasculature.
Collapse
Affiliation(s)
- Wen-Tao Sun
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China.
| | - Jian-Yong Du
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Jia Wang
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Yi-Long Wang
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China
| | - Er-Dan Dong
- Research Center for Cardiopulmonary Rehabilitation, University of Health and Rehabilitation Sciences Qingdao Hospital (Qingdao Municipal Hospital), School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, 266071, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing, 100191, China.
- The Institute of Cardiovascular Sciences, Peking University, Beijing, 100191, China.
- State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, 100191, China.
| |
Collapse
|
33
|
Chahal S, Raj RG, Kumar R. Risk of Type 1 Diabetes Mellitus in SARS-CoV-2 Patients. Curr Diabetes Rev 2025; 21:e240524230298. [PMID: 38798206 DOI: 10.2174/0115733998290807240522045553] [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: 01/23/2024] [Revised: 04/19/2024] [Accepted: 05/09/2024] [Indexed: 05/29/2024]
Abstract
Recent studies have found that a link between people with type 1 diabetes mellitus (T1DM) are at higher risk of morbidity as well as mortality from COVID-19 infection, indicating a need for vaccination. T1DM appears to impair innate and adaptive immunity. The overabundance of pro-inflammatory cytokines produced in COVID-19 illness that is severe and potentially fatal is known as a "cytokine storm." Numerous cohorts have revealed chronic inflammation as a key risk factor for unfavorable COVID-19 outcomes. TNF-α, interleukin (IL)-1a, IL-1, IL-2, IL-6, and other cytokines were found in higher concentrations in patients with T1DM. Even more importantly, oxidative stress contributes significantly to the severity and course of COVID- 19's significant role in the progression and severity of COVID-19 diseases. Severe glucose excursions, a defining characteristic of type 1 diabetes, are widely recognized for their potent role as mediating agents of oxidative stress via several routes, such as heightened production of advanced glycation end products (AGEs) and activation of protein kinase C (PKC). Furthermore, persistent endothelial dysfunction and hypercoagulation found in T1DM may impair microcirculation and endothelium, which could result in the development of various organ failure and acute breathing syndrome.
Collapse
Affiliation(s)
- Shweta Chahal
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Rojin G Raj
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Ranjeet Kumar
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
34
|
Jiang Y, Hu J, Cui C, Peng Z, Yang S, Lei J, Li B, Yang X, Qin J, Yin M, Liu X, Ye K, Xu Z, Zhang X, Lu X. Netrin1-Enriched Exosomes From Genetically Modified ADSCs as a Novel Treatment for Diabetic Limb Ischemia. Adv Healthc Mater 2025; 14:e2403521. [PMID: 39440618 DOI: 10.1002/adhm.202403521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Indexed: 10/25/2024]
Abstract
Diabetic limb ischemia (DLI) is a frequent complication of diabetes and the leading cause of non-traumatic amputation. Traditional treatments like stent placement and bypass surgery may not suit all patients. Exosome transplantation has emerged as a promising therapy. Netrin1, a protective cardiovascular factor, has an unclear role in DLI. This study investigates the role of Netrin1 in DLI patients and evaluates the therapeutic potential of exosomes derived from Netrin1-overexpressing adipose-derived stem cells (N-ADSCs). The expression of Netrin1 is significantly decreased in both endothelial cells and serum of DLI patients, highlighting its potential as a biomarker or therapeutic target. In vitro, Netrin1-enriched exosomes (N-Exos) promoted human umbilical vein endothelial cell (HUVEC) proliferation, migration, tube formation, and increased resistance to apoptosis under high glucose conditions. These protective effects are mediated through PI3K/AKT/eNOS and MEK/ERK pathways, and N-Exos further facilitated macrophage polarization from M1 to M2. In vivo, N-Exos demonstrates superior therapeutic effects over ADSC exosomes (Exos), including enhanced angiogenesis, improved collateral artery remodeling, reduced inflammation, and muscle protection. Collectively, these findings identify Netrin1 as a critical factor in DLI and underscore its significance in disease progression and therapeutic strategies. N-Exos offers a promising non-cellular therapeutic approach for the treatment of DLI.
Collapse
Affiliation(s)
- Yihong Jiang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Jiateng Hu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Chaoyi Cui
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Zhaoxi Peng
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Sen Yang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Jiahao Lei
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Xinrui Yang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Jinbao Qin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Minyi Yin
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Xiaobing Liu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Kaichuang Ye
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Zhijue Xu
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xing Zhang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, China
| |
Collapse
|
35
|
Schini-Kerth VB, Diouf I, Muzammel H, Said A, Auger C. Natural Products to Promote Vascular Health. Handb Exp Pharmacol 2025; 287:33-60. [PMID: 39317849 DOI: 10.1007/164_2024_721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Maintaining good vascular health is a major component in healthy ageing as it reduces the risk of cardiovascular diseases. Endothelial dysfunction, in particular, is a key mechanism in the development of major cardiovascular diseases including hypertension, atherosclerosis and diabetes. Recently, endothelial senescence has emerged as a pivotal early event in age-related endothelial dysfunction. Endothelial function is characterized by an imbalance between the endothelial formation of vasoprotective mechanisms, including the formation of nitric oxide (NO) and endothelium-dependent hyperpolarization responses, and an increased level of oxidative stress involving several pro-oxidant enzymes such as NADPH oxidases and, often also, the appearance of cyclooxygenase-derived vasoconstrictors. Pre-clinical studies have indicated that natural products, in particular several polyphenol-rich foods, can trigger activating pathways in endothelial cells promoting an increased formation of NO and endothelium-dependent hyperpolarization. In addition, some can even exert beneficial effects on endothelial senescence. Moreover, some of these products have been associated with the prevention and/or improvement of established endothelial dysfunction in several experimental models of cardiovascular diseases and in humans with cardiovascular diseases. Therefore, intake of certain natural products, such as dietary and plant-derived polyphenol-rich products, appears to be an attractive approach for a healthy vascular system in ageing.
Collapse
Affiliation(s)
- Valérie B Schini-Kerth
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France.
| | - Ibrahima Diouf
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Hira Muzammel
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Amissi Said
- Translational Cardiovascular Medicine, UR 3074, CRBS, University of Strasbourg, Strasbourg, France
| | - Cyril Auger
- Regenerative Nanomedicine, INSERM UMR 1260, CRBS, University of Strasbourg, Strasbourg, France
| |
Collapse
|
36
|
Liu Y, Li Z, Xu Y, Mao H, Huang N. Uric Acid and Atherosclerosis in Patients with Chronic Kidney Disease: Recent Progress, Mechanisms, and Prospect. KIDNEY DISEASES (BASEL, SWITZERLAND) 2025; 11:112-127. [PMID: 40124130 PMCID: PMC11928073 DOI: 10.1159/000543781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/16/2025] [Indexed: 03/25/2025]
Abstract
Background Chronic kidney disease (CKD) is a prevalent global health concern, significantly linked to increased cardiovascular morbidity and mortality. Among various risk factors, uric acid (UA) has emerged as a potentially modifiable contributor to cardiovascular complications in CKD patients. Summary Elevated serum uric acid levels frequently occur in individuals with CKD and are associated with the development of atherosclerosis (AS). Uric acid has been demonstrated to exacerbate inflammatory processes, promote oxidative stress, and cause endothelial dysfunction, which are critical factors that drive the formation of atherosclerotic plaques. Furthermore, high uric acid levels can worsen renal function, establishing a detrimental cycle that amplifies cardiovascular risk. Key Messages This review investigates the complex interconnection between UA and AS in patients with CKD, highlighting the underlying mechanisms and therapeutic considerations. A more profound comprehension of this relationship is essential for enhancing cardiovascular health and outcomes in this vulnerable population.
Collapse
Affiliation(s)
- Yuchu Liu
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Zeyu Li
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Yuanwen Xu
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Haiping Mao
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| | - Naya Huang
- Department of Nephrology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
- Key Laboratory of Nephrology, Ministry of Health of China, Guangdong Provincial Key Laboratory of Nephrology, Guangzhou, China
| |
Collapse
|
37
|
Li Z, Gu M, Zaparte A, Fu X, Mahen K, Mrdjen M, Li XS, Yang Z, Ma J, Thoudam T, Chandler K, Hesler M, Heathers L, Gorse K, Van TT, Wong D, Gibson AM, Wang Z, Taylor CM, Quijada P, Makarewich CA, Hazen SL, Liangpunsakul S, Brown JM, Lefer DJ, Welsh DA, Sharp TE. Alcohol-induced gut microbial reorganization and associated overproduction of phenylacetylglutamine promotes cardiovascular disease. Nat Commun 2024; 15:10788. [PMID: 39738016 PMCID: PMC11685538 DOI: 10.1038/s41467-024-55084-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Accepted: 11/27/2024] [Indexed: 01/01/2025] Open
Abstract
The mechanism(s) underlying gut microbial metabolite (GMM) contribution towards alcohol-mediated cardiovascular disease (CVD) is unknown. Herein we observe elevation in circulating phenylacetylglutamine (PAGln), a known CVD-associated GMM, in individuals living with alcohol use disorder. In a male murine binge-on-chronic alcohol model, we confirm gut microbial reorganization, elevation in PAGln levels, and the presence of cardiovascular pathophysiology. Fecal microbiota transplantation from pair-/alcohol-fed mice into naïve male mice demonstrates the transmissibility of PAGln production and the CVD phenotype. Independent of alcohol exposure, pharmacological-mediated increases in PAGln elicits direct cardiac and vascular dysfunction. PAGln induced hypercontractility and altered calcium cycling in isolated cardiomyocytes providing evidence of improper relaxation which corresponds to elevated filling pressures observed in vivo. Furthermore, PAGln directly induces vascular endothelial cell activation through induction of oxidative stress leading to endothelial cell dysfunction. We thus reveal that the alcohol-induced microbial reorganization and resultant GMM elevation, specifically PAGln, directly contributes to CVD.
Collapse
Affiliation(s)
- Zhen Li
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Min Gu
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Science Center, New Orleans, LA, USA
- International Flavors and Fragrances Health and Bioscience, Shanghai, China
| | - Aline Zaparte
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Science Center, New Orleans, LA, USA
- Comprehensive Alcohol Research Center, School of Medicine, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Xiaoming Fu
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Learner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Kala Mahen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Learner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center (NOAC), Cleveland Clinic, Cleveland, OH, USA
| | - Marko Mrdjen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Learner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center (NOAC), Cleveland Clinic, Cleveland, OH, USA
| | - Xinmin S Li
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Learner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Zhihong Yang
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jing Ma
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Themis Thoudam
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kristina Chandler
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Maggie Hesler
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Laura Heathers
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Kiersten Gorse
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Thanh Trung Van
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - David Wong
- Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Aaron M Gibson
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zeneng Wang
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Christopher M Taylor
- Comprehensive Alcohol Research Center, School of Medicine, Louisiana State University Health Science Center, New Orleans, LA, USA
- Department of Microbiology, Immunology and Parasitology, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Pearl Quijada
- Integrative Biology and Physiology, University of California, Los Angeles, CA, USA
| | - Catherine A Makarewich
- The Heart Institute, Division of Molecular Cardiovascular Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Stanley L Hazen
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Learner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Heart and Vascular and Thoracic Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Suthat Liangpunsakul
- Division of Gastroenterology and Hepatology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush Veterans Administration Medical Center, Indianapolis, IN, USA
| | - J Mark Brown
- Department of Cardiovascular & Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Center for Microbiome and Human Health, Learner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
- Northern Ohio Alcohol Center (NOAC), Cleveland Clinic, Cleveland, OH, USA
| | - David J Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - David A Welsh
- Section of Pulmonary/Critical Care and Allergy/Immunology, Department of Medicine, Louisiana State University Health Science Center, New Orleans, LA, USA
- Comprehensive Alcohol Research Center, School of Medicine, Louisiana State University Health Science Center, New Orleans, LA, USA
| | - Thomas E Sharp
- Department of Molecular Pharmacology and Physiology, Morsani College of Medicine, University of South Florida, Tampa, FL, USA.
- Heart Institute, Morsani College of Medicine, USF Health, University South Florida, Tampa, FL, USA.
| |
Collapse
|
38
|
Hassan M, Vinagolu-Baur J, Li V, Frasier K, Herrick G, Scotto T, Rankin E. E-cigarettes and arterial health: A review of the link between vaping and atherosclerosis progression. World J Cardiol 2024; 16:707-719. [PMID: 39734821 PMCID: PMC11669975 DOI: 10.4330/wjc.v16.i12.707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 11/26/2024] Open
Abstract
Recent studies have suggested an evolving understanding of the association between vaping, specifically electronic cigarette (e-cigarette) use, and the progression of atherosclerosis, a significant contributor to cardiovascular disease. Despite the prevailing perception of vaping as a safer alternative to traditional tobacco smoking, accumulating evidence suggests that the aerosols emitted by e-cigarettes contain harmful constituents that may promote endothelial dysfunction, oxidative stress, inflammation, and dyslipidemia-key mechanisms implicated in atherosclerosis pathogenesis. While past research, including experimental studies and clinical investigations, has shed light on the potential cardiovascular risks associated with vaping, gaps in knowledge persist. Future research endeavors should focus on interpreting the long-term effects of vaping on atherosclerosis development and progression, exploring the impact of different e-cigarette formulations and user demographics, and identifying effective strategies for mitigating the cardiovascular consequences of vaping. By identifying and addressing these research gaps, we can enhance our understanding of the cardiovascular implications of vaping and inform evidence-based interventions and policies to safeguard public health.
Collapse
Affiliation(s)
- Muhammad Hassan
- Department of Medicine, Nuvance Health, Vassar Brothers Medical Center, Poughkeepsie, NY 12601, United States
| | - Julia Vinagolu-Baur
- Department of Medical Education, State University of New York, Upstate Medical University, Syracuse, NY 13210, United States
| | - Vivian Li
- Department of Medicine, Nuvance Health, Vassar Brothers Medical Center, Poughkeepsie, NY 12601, United States.
| | - Kelly Frasier
- Department of Medicine, Nuvance Health, Vassar Brothers Medical Center, Poughkeepsie, NY 12601, United States
| | - Grace Herrick
- Department of Medical Education, Alabama College of Osteopathic Medicine, Dothan, AL 36303, United States
| | - Tiffany Scotto
- Department of Medicine, University of Florida Health, Jacksonville, FL 32209, United States
| | - Erica Rankin
- Department of Medical Education, Nova Southeastern University Kiran C. Patel College of Osteopathic Medicine, Fort Lauderdale, FL 33328, United States
| |
Collapse
|
39
|
Atallah R, Gindlhuber J, Platzer W, Rajesh R, Heinemann A. Succinate Regulates Endothelial Mitochondrial Function and Barrier Integrity. Antioxidants (Basel) 2024; 13:1579. [PMID: 39765906 PMCID: PMC11673088 DOI: 10.3390/antiox13121579] [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: 11/15/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Endothelial dysfunction is a hallmark of several pathological conditions, including cancer, cardiovascular disease and inflammatory disorders. In these conditions, perturbed TCA cycle and subsequent succinate accumulation have been reported. The role of succinate as a regulator of immunological responses and inflammation is increasingly being recognized. Nevertheless, how endothelial cell function and phenotype are altered by elevated intracellular succinate has not been addressed yet. Thus, we employed numerous in vitro functional assays using primary HUVECs and diethyl succinate (DES), a cell membrane-permeable succinate analogue. An MTS assay 1 h post stimulation with DES suggested reduced metabolic activity in HUVECs. Concurrently, elevated production of ROS, including mitochondrial superoxide, and a reduction in mitochondrial membrane potential were observed. These findings were corroborated by Seahorse mito-stress testing, which revealed that DES acutely lowered the OCR, maximal respiration and ATP production. Given the link between mitochondrial stress and apoptosis, we examined important survival signalling pathways. DES transiently reduced ERK1/2 phosphorylation, a response that was followed by a skewed pro-apoptotic shift in the BAX to BCL2L1 gene expression ratio, which coincided with upregulating VEGF gene expression. This indicated an induction of mixed pro-apoptotic and pro-survival signals in the cell. However, the BAX/BCL-XL protein ratio was unchanged, suggesting that the cells did not commit themselves to apoptosis. An MTS assay, caspase 3/7 activity assay and annexin V/propidium iodide staining confirmed this finding. By contrast, stimulation with DES induced acute endothelial barrier permeability, forming intercellular gaps, altering cell size and associated actin filaments without affecting cell count. Notably, during overnight DES exposure gradual recovery of the endothelial barrier and cell sprouting was observed, alongside mitochondrial membrane potential restoration, albeit with sustained ROS production. COX-2 inhibition and EP4 receptor blockade hindered barrier restoration, implicating a role of COX-2/PGE2/EP4 signalling in this process. Interestingly, ascorbic acid pre-treatment prevented DES-induced acute barrier disruption independently from ROS modulation. In conclusion, succinate acts as a significant regulator of endothelial mitochondrial function and barrier integrity, a response that is counterbalanced by upregulated VEGF and prostaglandin production by the endothelial cells.
Collapse
Affiliation(s)
- Reham Atallah
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Juergen Gindlhuber
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Physiology & Pathophysiology, Medical University of Graz, 8010 Graz, Austria
| | - Wolfgang Platzer
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Rishi Rajesh
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| | - Akos Heinemann
- Otto Loewi Research Center for Vascular Biology, Immunology and Inflammation, Division of Pharmacology, Medical University of Graz, 8010 Graz, Austria
| |
Collapse
|
40
|
Auger C, Muzammel H, Diouf I, Schini-Kerth VB. Potential of Anthocyanin-rich Products to Prevent and Improve Endothelial Function and Senescence: Focus on Anthocyanins. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:27590-27618. [PMID: 39629614 DOI: 10.1021/acs.jafc.4c04727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Endothelial dysfunction is a pivotal early event in the development of major cardiovascular diseases including hypertension, atherosclerosis, diabetes, and aging. The alteration of the endothelial function is often triggered by an imbalance between the endothelial formation of vasoprotective factors, including nitric oxide (NO) and endothelium-dependent hyperpolarization (EDH), and vasocontracting factors, such as arachidonic acid-derived mediators generated by cyclooxygenases, and an increased level of oxidative stress. Recently, endothelial senescence was reported to be an early trigger of endothelial dysfunction. Preclinical studies indicate that polyphenol-rich food, including anthocyanin-rich products, can activate pathways promoting an increased formation of vasoprotective factors and can prevent the induction of endothelial dysfunction in endothelial cells and isolated blood vessels. Similarly, intake of anthocyanin-rich products has been associated with the prevention and/or the improvement of an endothelial dysfunction in several experimental models of cardiovascular diseases, including physiological aging. Moreover, clinical data indicate that polyphenol-rich and anthocyanin-rich products can improve endothelial function and vascular health in humans with cardiovascular diseases. The present review will discuss both experimental and clinical evidence indicating that several polyphenol-rich foods and natural products, and especially anthocyanin-rich products, can promote endothelial and vascular health, as well as the underlying mechanisms.
Collapse
Affiliation(s)
- Cyril Auger
- University of Strasbourg, INSERM, Regenerative Nanomedicine UMR 1260, 67000 Strasbourg, France
| | - Hira Muzammel
- University of Strasbourg, Translational Cardiovascular Medicine UR 3074, 67000 Strasbourg, France
| | - Ibrahima Diouf
- University of Strasbourg, Translational Cardiovascular Medicine UR 3074, 67000 Strasbourg, France
| | - Valérie B Schini-Kerth
- University of Strasbourg, Translational Cardiovascular Medicine UR 3074, 67000 Strasbourg, France
| |
Collapse
|
41
|
Vordos Z, Deli I, Anifanti M, Kluzek S, Koutlianos N, Kouidi E, Deligiannis A. The Effect of Dark Chocolate Consumption on Arterial Function in Endurance Male Runners: Prospective Cohort Study. Sports (Basel) 2024; 12:344. [PMID: 39728884 DOI: 10.3390/sports12120344] [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: 11/20/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 12/28/2024] Open
Abstract
Foods rich in polyphenols have beneficial effects on health. This study aimed to examine the impact of dark chocolate on endurance runners' arterial function. Forty-six male amateur runners, aged 25-55, participated. The initial assessments included clinical testing, arterial stiffness measurements, and a cardiopulmonary exercise test. The participants then consumed 50 g of dark chocolate (70% cocoa) daily for two weeks, maintaining their usual training routine. After this period, the baseline assessment was repeated. The results showed significant improvements. Pulse wave velocity decreased by 11.82% (p < 0.001), and augmentation index by 19.47% (p < 0.001). Systolic brachial blood pressure reduced by 2.12% (p < 0.05), diastolic by 2.79% (p < 0.05), and mean pressure by 2.41% (p < 0.05). Central arterial pressure also decreased, with systolic by 1.24% (p < 0.05), diastolic by 2.80% (p < 0.05), and mean pressure by 2.43% (p < 0.05). Resting heart rate increased by 4.57% (p < 0.05) and left ventricular ejection time decreased by 4.89% (p < 0.05), particularly in athletes over 40. Exercise time increased by 2.16% (p < 0.05), heart rate (max) by 1.15% (p < 0.05), VO2max by 2.31% (p < 0.05), and anaerobic threshold shifted by 6.91% (p < 0.001) in exercise time and 6.93% (p < 0.001) in VO2max. In conclusion, dark chocolate improves arterial function in endurance runners, enhancing vascular health.
Collapse
Affiliation(s)
- Zacharias Vordos
- Laboratory of Sports Medicine, Department of Physical Education and Sports Science, Aristotle University of Thessaloniki, 57001 Thermi, Greece
| | - Ifigeneia Deli
- Laboratory of Sports Medicine, Department of Physical Education and Sports Science, Aristotle University of Thessaloniki, 57001 Thermi, Greece
| | - Maria Anifanti
- Laboratory of Sports Medicine, Department of Physical Education and Sports Science, Aristotle University of Thessaloniki, 57001 Thermi, Greece
| | - Stefan Kluzek
- Sports & Exercise Medicine and Trauma & Orthopaedic Surgery, University of Nottingham, Nottingham NG7 2UH, UK
| | - Nikolaos Koutlianos
- Laboratory of Sports Medicine, Department of Physical Education and Sports Science, Aristotle University of Thessaloniki, 57001 Thermi, Greece
| | - Evangelia Kouidi
- Laboratory of Sports Medicine, Department of Physical Education and Sports Science, Aristotle University of Thessaloniki, 57001 Thermi, Greece
| | - Asterios Deligiannis
- Laboratory of Sports Medicine, Department of Physical Education and Sports Science, Aristotle University of Thessaloniki, 57001 Thermi, Greece
| |
Collapse
|
42
|
Paolini L, Guida F, Calvaruso A, Andreozzi L, Pierantoni L, Lanari M, Fabi M. Endothelial Dysfunction: Molecular Mechanisms and Therapeutic Strategies in Kawasaki Disease. Int J Mol Sci 2024; 25:13322. [PMID: 39769085 PMCID: PMC11676170 DOI: 10.3390/ijms252413322] [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: 10/30/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 01/11/2025] Open
Abstract
The endothelium plays a key role in regulating vascular homeostasis by responding to a large spectrum of chemical and physical stimuli. Vasculitis is a group of inflammatory conditions affecting the vascular bed, and it is known that they are strongly linked to endothelial dysfunction (ED). Kawasaki disease (KD) is one childhood systemic vasculitis, and it represents the leading cause of acquired cardiac disease in children due to coronary damage and subsequent cardiovascular (CV) morbidity and mortality. We aimed to focus on the actual knowledge of ED in the pathogenesis of KD and its practical implications on therapeutical strategies to limit cardiovascular complications. Understanding ED in KD provides insight into the underlying mechanisms and identifies potential therapeutic targets to mitigate vascular damage, ultimately improving cardiovascular outcomes in both the acute and chronic stages of the disease. However, research gaps remain, particularly in translating findings from animal models into clinical applications for cardiovascular lesions and related morbidity in KD patients.
Collapse
Affiliation(s)
- Lucia Paolini
- Specialty School of Paediatrics, Alma Mater Studiorum, University of Bologna, 40139 Bologna, Italy; (L.P.); (A.C.)
| | - Fiorentina Guida
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| | - Antonino Calvaruso
- Specialty School of Paediatrics, Alma Mater Studiorum, University of Bologna, 40139 Bologna, Italy; (L.P.); (A.C.)
| | - Laura Andreozzi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| | - Luca Pierantoni
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| | - Marcello Lanari
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, 40139 Bologna, Italy
| | - Marianna Fabi
- Pediatric Emergency Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40139 Bologna, Italy; (F.G.); (L.A.); (L.P.); (M.L.)
| |
Collapse
|
43
|
de Melo IG, Tavares V, Savva-Bordalo J, Rei M, Liz-Pimenta J, Pereira D, Medeiros R. Endothelial Dysfunction Markers in Ovarian Cancer: VTE Risk and Tumour Prognostic Outcomes. Life (Basel) 2024; 14:1630. [PMID: 39768338 PMCID: PMC11678387 DOI: 10.3390/life14121630] [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: 11/03/2024] [Revised: 11/28/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Ovarian cancer (OC) presents daunting lethality rates worldwide, with frequent late-stage diagnosis and chemoresistance, highlighting the need for improved prognostic approaches. Venous thromboembolism (VTE), a major cancer mortality factor, is partially driven by endothelial dysfunction (ED). ED's pro-inflammatory state fosters tumour progression, suggesting a VTE-independent link between ED and cancer. Given this triad's interplay, ED markers may influence OC behaviour and patients' prognosis. Thus, the impact of ED-related genes and single-nucleotide polymorphisms (SNPs) on OC-related VTE and patient thrombogenesis-independent prognosis was investigated. NOS3 upregulation was linked to lower VTE incidence (χ2, p = 0.013), while SELP upregulation was associated with shorter overall survival (log-rank test, p = 0.048). Dismissing patients with VTE before OC diagnosis, SELP rs6136 T allele carriers presented lower progression-free survival (log-rank test, p = 0.038). Nevertheless, due to the SNP minor allele underrepresentation, further investigation is required. Taken together, ED markers seem to exhibit roles that depend on the clinical context, such as tumour-related thrombogenesis or cancer prognosis. Validation with larger cohorts and more in-depth functional studies are needed for data clarification and potential therapeutic strategies exploitation to tackle cancer progression and thrombosis in OC patients.
Collapse
Affiliation(s)
- Inês Guerra de Melo
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
| | - Valéria Tavares
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Joana Savva-Bordalo
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Mariana Rei
- Department of Gynaecology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal;
| | - Joana Liz-Pimenta
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- Department of Medical Oncology, Centro Hospitalar de Trás-os-Montes e Alto Douro (CHTMAD), 5000-508 Vila Real, Portugal
| | - Deolinda Pereira
- Department of Medical Oncology, Portuguese Institute of Oncology of Porto (IPO Porto), 4200-072 Porto, Portugal; (J.S.-B.); (D.P.)
| | - Rui Medeiros
- Molecular Oncology and Viral Pathology Group, Research Centre of IPO Porto (CI-IPOP), Pathology and Laboratory Medicine Department, Clinical Pathology SV/RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto. CCC), 4200-072 Porto, Portugal; (I.G.d.M.); (V.T.)
- Faculty of Medicine, University of Porto (FMUP), 4200-072 Porto, Portugal;
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
- Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- Research Department, Portuguese League Against Cancer (NRNorte), 4200-172 Porto, Portugal
| |
Collapse
|
44
|
Allan JM, Fox BM, Kasztan M, Kelly GC, Molina PA, King MA, Colson J, Wells L, Bowman L, Blackburn M, Kutlar A, Harris RA, Pollock DM, Pollock JS. Enhanced vasoconstriction in sickle cell disease is dependent on ETA receptor activation. Clin Sci (Lond) 2024; 138:1505-1520. [PMID: 39526571 DOI: 10.1042/cs20240625] [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: 03/31/2024] [Revised: 11/03/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024]
Abstract
Sickle cell disease (SCD) carries a significant risk for poor vascular health and vascular dysfunction. High levels of vascular reactive oxygen species (ROS) as well as elevated plasma endothelin-1 (ET-1), a potent vasoconstrictor with actions via the ETA receptor, are both common phenotypes in SCD. Alpha-1 adrenergic receptor activation is a major mediator of stress-induced vasoconstriction. However, the mechanism of the SCD enhanced vasoconstrictive response is unknown. We hypothesized that SCD induces enhanced alpha-1 adrenergic mediated vasoconstriction through the ET-1/ETA receptor pathway in arterial tissues. Utilizing humanized SCD (HbSS) and genetic control (HbAA) mice, alpha-1a, but not alpha-1b or alpha-1d, receptor expression was significantly greater in aortic tissue from HbSS mice compared to HbAA mice. Significantly enhanced vasoconstriction in aortic and carotid arterial segments were observed from HbSS mice compared with HbAA mice. Treatment with ambrisentan, a selective ETA receptor antagonist, and a ROS scavenger normalized the aortic vasoconstrictive response in HbSS mice. In a randomized translational study, patients with SCD were treated with placebo or ambrisentan for 3 months, with the treatment group showing an increase in the percent brachial arterial diameter. Taken together, these data suggest that the ETA receptor pathway interaction with the adrenergic receptor pathway contributes to enhanced aortic vasoconstriction in SCD. Findings indicate the potential of ETA antagonism as a therapeutic avenue for improving vascular health in SCD.
Collapse
Affiliation(s)
- John Miller Allan
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - Brandon M Fox
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - Malgorzata Kasztan
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
- Division of Hematology-Oncology, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - Gillian C Kelly
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - Patrick A Molina
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - McKenzi A King
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - Jackson Colson
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - Leigh Wells
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
- Division of Hematology and Oncology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Latanya Bowman
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
- Division of Hematology and Oncology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Marsha Blackburn
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
- Division of Hematology and Oncology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Abdullah Kutlar
- Division of Hematology and Oncology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - Ryan A Harris
- Georgia Prevention Institute, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, U.S.A
| | - David M Pollock
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| | - Jennifer S Pollock
- Section of Cardiorenal Physiology & Medicine, Division of Nephrology, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35233, U.S.A
| |
Collapse
|
45
|
Jansen J, Marshall PW, Benatar JR, Cross R, Lindbom TK, Kingsley M. Low-Intensity Resistance Exercise in Cardiac Rehabilitation: A Narrative Review of Mechanistic Evidence and Clinical Implications. J Clin Med 2024; 13:7338. [PMID: 39685797 DOI: 10.3390/jcm13237338] [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: 11/13/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/18/2024] Open
Abstract
Cardiac rehabilitation, a multi-component intervention designed to mitigate the impact of cardiovascular disease, often underutilises low-intensity resistance exercise despite its potential benefits. This narrative review critically examines the mechanistic and clinical evidence supporting the incorporation of low-intensity resistance exercise into cardiac rehabilitation programmes. Research indicates that low-intensity resistance exercise induces hypertrophic adaptations by maximising muscle fibre activation through the size principle, effectively recruiting larger motor units as it approaches maximal effort. This activation promotes adaptation in both type I and II muscle fibres, resulting in comparable increases in myofibrillar protein synthesis and phosphorylation of key signalling proteins when compared to high-intensity resistance exercise. Low-intensity resistance exercise provides equivalent improvements in muscular strength and hypertrophy compared to high-intensity protocols while addressing barriers to participation, such as concerns about safety and logistical challenges. By facilitating engagement through a more accessible exercise modality, low-intensity resistance exercise might improve adherence rates and patient outcomes in cardiac rehabilitation. Additionally, the ability of low-intensity resistance exercise to address sarcopenia and frailty syndrome, significant determinants of cardiovascular disease progression, can enhance the recovery and overall quality of life for patients. This review establishes evidence-based recommendations for the inclusion of low-intensity resistance exercise in cardiac rehabilitation, offering a promising pathway to enhance the effectiveness of these programmes.
Collapse
Affiliation(s)
- Jemima Jansen
- Department of Exercise Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Paul W Marshall
- Department of Exercise Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Jocelyne R Benatar
- Department of Exercise Sciences, University of Auckland, Auckland 1010, New Zealand
- Greenlane Cardiovascular Service, Auckland City Hospital, Auckland 1023, New Zealand
| | - Rebecca Cross
- Department of Health Sciences, Macquarie University, Sydney 2113, Australia
| | - Tia K Lindbom
- Department of Exercise Sciences, University of Auckland, Auckland 1010, New Zealand
| | - Michael Kingsley
- Department of Exercise Sciences, University of Auckland, Auckland 1010, New Zealand
- Holsworth Research Initiative, La Trobe Rural Health School, La Trobe University, Melbourne 3000, Australia
| |
Collapse
|
46
|
Bahreiny SS, Ahangarpour A, Harooni E, Amraei M, Aghaei M, Fard RM. Closer look at circulating nitric oxide levels and their association with polycystic ovary syndrome: A meta-analytical exploration. Int J Reprod Biomed 2024; 22:943-962. [PMID: 39968366 PMCID: PMC11830925 DOI: 10.18502/ijrm.v22i12.18061] [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: 05/21/2024] [Revised: 10/08/2024] [Accepted: 11/16/2024] [Indexed: 02/20/2025] Open
Abstract
Background Polycystic ovary syndrome (PCOS) casts a wide shadow over the reproductive health of millions of women worldwide, emerging as one of the most complex and multifaceted endocrine disorders. In addition, nitric oxide (NO) stands out as a pivotal signaling molecule, orchestrating a symphony of physiological processes. Objective This meta-analysis aims to elucidate the association between NO levels and PCOS, investigate the potential of NO as a biomarker for PCOS diagnosis, and evaluate its clinical significance. Materials and Methods A systematic review was conducted in several electronic databases, including PubMed, Web of Science, Cochrane Library, Scopus, EMBASE, and Google Scholar, to identify relevant studies published up to January 2024. Standardized mean difference and 95% CI were calculated using a random effects model to assess the overall effect size. Meta-regressions and subgroup analysis were performed to investigate sources of heterogeneity. Results A meta-analysis of 14 studies with 1171 participants showed that NO levels were significantly lower in the PCOS group than in the control group. The pooled analysis yielded a standardized mean difference of -0.482; 95% CI: -0.908 to -0.056; p = 0.027. Subgroup analyses further demonstrated variations in NO levels between different PCOS phenotypes and in relation to metabolic parameters. Conclusion This meta-analysis provides evidence for an association between PCOS and dysregulated NO levels and suggests a potential role of NO as a biomarker in the diagnosis and pathogenesis of PCOS.
Collapse
Affiliation(s)
- Seyed Sobhan Bahreiny
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Medical Basic Sciences Research Institute, Physiology Research Center, Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Akram Ahangarpour
- Medical Basic Sciences Research Institute, Physiology Research Center, Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Elnaz Harooni
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Medical Basic Sciences Research Institute, Physiology Research Center, Department of Physiology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mahdi Amraei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mojtaba Aghaei
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Reza Mohammadpour Fard
- Student Research Committee, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
- Department of Immunology, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
47
|
Wabel EA, Krieger-Burke T, Watts SW. Vascular chemerin from PVAT contributes to norepinephrine and serotonin-induced vasoconstriction and vascular stiffness in a sex-dependent manner. Am J Physiol Heart Circ Physiol 2024; 327:H1577-H1589. [PMID: 39453435 DOI: 10.1152/ajpheart.00475.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
The adipokine chemerin supports normal blood pressure and contributes to adiposity-associated hypertension, evidenced by falls in mean arterial pressure in Dahl SS rats given an antisense oligonucleotide against chemerin. In humans, circulating chemerin is positively associated with hypertension and aortic stiffness. Mechanisms of chemerin's influence on vascular health and disease remain unknown. We identified chemerin production in the vasculature-the blood vessel and its perivascular adipose tissue (PVAT). Here, using RNAScope, qPCR, isometric contractility, high-frequency ultrasound imaging, and Western blot in the Dahl SS rat, we test the hypothesis that endogenous chemerin amplifies agonist-induced vasoconstriction through the chemerin1 receptor and that chemerin drives aortic stiffness in the thoracic aorta. CMKLR1 (chemerin1) expression was higher in the media, and Rarres2 (chemerin) expression was higher in the PVAT. Chemerin1 receptor antagonism via selective inhibitor CCX832 reduced maximal contraction to norepinephrine (NE) and serotonin (5-HT), but not angiotensin II, in isolated thoracic aorta (PVAT intact) from male Dahl SS rat. In females, CCX832 did not alter contraction to NE or 5-HT. Male, but not female, genetic chemerin knockout Dahl SS rats had lower aortic arch pulse wave velocity than wild types, indicating chemerin's role in aortic stiffness. Aortic PVAT from females expressed less chemerin protein than males, suggesting PVAT as the primary source of active chemerin. We show that chemerin made by the PVAT amplifies NE and 5-HT-induced contraction and potentially induces aortic stiffening in a sex-dependent manner, highlighting the potential for chemerin to be a key factor in blood pressure control and aortic stiffening.NEW & NOTEWORTHY Chemerin1 receptor inhibition reduced norepinephrine (NE) and 5-HT-induced vasoconstriction in males. Genetic chemerin knockout (KO) resulted in lower pulse wave velocity in males. Differences in chemerin abundance in aorta perivascular adipose tissue (APVAT) may explain sex-dependent role of chemerin.
Collapse
Affiliation(s)
- Emma A Wabel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Teresa Krieger-Burke
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
48
|
Zhou W, Jiang X, Gao J. Extracellular vesicles for delivering therapeutic agents in ischemia/reperfusion injury. Asian J Pharm Sci 2024; 19:100965. [PMID: 39640057 PMCID: PMC11617990 DOI: 10.1016/j.ajps.2024.100965] [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: 05/07/2024] [Revised: 06/08/2024] [Accepted: 06/29/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemia/reperfusion (I/R) injury is marked by the restriction and subsequent restoration of blood supply to an organ. This process can exacerbate the initial tissue damage, leading to further disorders, disability, and even death. Extracellular vesicles (EVs) are crucial in cell communication by releasing cargo that regulates the physiological state of recipient cells. The development of EVs presents a novel avenue for delivering therapeutic agents in I/R therapy. The therapeutic potential of EVs derived from stem cells, endothelial cells, and plasma in I/R injury has been actively investigated. Therefore, this review aims to provide an overview of the pathological process of I/R injury and the biophysical properties of EVs. We noted that EVs serve as nontoxic, flexible, and multifunctional carriers for delivering therapeutic agents capable of intervening in I/R injury progression. The therapeutic efficacy of EVs can be enhanced through various engineering strategies. Improving the tropism of EVs via surface modification and modulating their contents via preconditioning are widely investigated in preclinical studies. Finally, we summarize the challenges in the production and delivery of EV-based therapy in I/R injury and discuss how it can advance. This review will encourage further exploration in developing efficient EV-based delivery systems for I/R treatment.
Collapse
Affiliation(s)
- Weihang Zhou
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Xinchi Jiang
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Jianqing Gao
- State Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
- Department of Pharmacy, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China
- Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
49
|
Lupoli R, Calcaterra I, Ambrosino P, Giacco R, Vitale M, Della Pepa G, Rivellese AA, Iannuzzo G, Bozzetto L, Di Minno M. Effects of Mediterranean Diet on Endothelial Reactivity in Individuals with High Cardiometabolic Risk: A Randomized Controlled Parallel-Group Preliminary Trial. Biomedicines 2024; 12:2595. [PMID: 39595161 PMCID: PMC11592348 DOI: 10.3390/biomedicines12112595] [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: 10/08/2024] [Revised: 10/30/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND Endothelial dysfunction is recognized as an early modification involved in the pathogenesis of vascular diseases. Evidence suggests that the Mediterranean Diet (MD) is associated with endothelial function improvement and, in turn, plays an important role in atherosclerosis development and progression. OBJECTIVES To evaluate both acute and sustained effects of the MD on endothelial function in patients with high cardiometabolic risk. METHODS A total of 25 subjects were randomly assigned to either the MD group or the Control Diet (CD) group according to a single-blind, parallel-group study design. Endothelial function was evaluated through non-invasive flow-mediated dilation (FMD) measurements at baseline (T0) and after 8 weeks (Tw8) of the MD or CD intervention, under both 12 h fast condition (fasting) and 2 h post-meal resembling the assigned diet (2 h). Assessments were conducted by a blinded sonographer. RESULTS FMD at T0-fasting was similar between MD and CD groups (6.11% ± 0.67 vs. 7.90% ± 1.65; p = 0.266). A significant difference in FMD between MD and CD groups was observed at T0-2h (12.14% ± 1.93 vs. 4.01% ± 1.03; p = 0.004), T8w-fasting (9.76% ± 1.18 vs. 5.03% ± 0.89; p = 0.008), and T8w-2h (8.99% ± 1.22 vs. 3.86% ± 0.52; p = 0.003). Oral glucose insulin sensitivity (OGIS) at T0 correlated with FMD percent changes from T0-fasting to T0-2h (r = 0.414, p = 0.044). After adjusting for age, gender, and OGIS, MD was an independent predictor of percent changes in FMD from T0-fasting to T0-2h (β: -0.582, p = 0.003), from T0-fasting to T8w-fasting (β: -0.498, p = 0.013), and from T0-fasting to T8w-2h (β: -0.479, p = 0.018). CONCLUSIONS Adherence to the MD may improve endothelial function in both the short- and medium-term among patients at high cardiometabolic risk.
Collapse
Affiliation(s)
- Roberta Lupoli
- Department of Molecular Medicine and Medical Biotechnology, Federico II University, 80131 Naples, Italy;
| | - Ilenia Calcaterra
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (I.C.); (M.V.); (G.D.P.); (A.A.R.); (G.I.); (L.B.); (M.D.M.)
| | - Pasquale Ambrosino
- Istituti Clinici Scientifici Maugeri IRCCS, Scientific Directorate of Telese Terme Institute, 82037 Telese Terme, Italy
| | - Rosalba Giacco
- Institute of Food Sciences, National Research Council, 83100 Avellino, Italy;
| | - Marilena Vitale
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (I.C.); (M.V.); (G.D.P.); (A.A.R.); (G.I.); (L.B.); (M.D.M.)
| | - Giuseppe Della Pepa
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (I.C.); (M.V.); (G.D.P.); (A.A.R.); (G.I.); (L.B.); (M.D.M.)
| | - Angela Albarosa Rivellese
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (I.C.); (M.V.); (G.D.P.); (A.A.R.); (G.I.); (L.B.); (M.D.M.)
| | - Gabriella Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (I.C.); (M.V.); (G.D.P.); (A.A.R.); (G.I.); (L.B.); (M.D.M.)
| | - Lutgarda Bozzetto
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (I.C.); (M.V.); (G.D.P.); (A.A.R.); (G.I.); (L.B.); (M.D.M.)
| | - Matteo Di Minno
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (I.C.); (M.V.); (G.D.P.); (A.A.R.); (G.I.); (L.B.); (M.D.M.)
| |
Collapse
|
50
|
Bock PM, Monteiro RB, Maraschin CK, Alegretti AP, Farias MG, Spagnol F, Lopez PLDC, Santos LP, Helal L, Moraes RS, Umpierre D, Schaan BD. Circulating endothelial progenitor cells and inflammatory markers in type 1 diabetes after an acute session of aerobic exercise. ARCHIVES OF ENDOCRINOLOGY AND METABOLISM 2024; 68:e230499. [PMID: 39876965 PMCID: PMC11771752 DOI: 10.20945/2359-4292-2023-0499] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 09/04/2024] [Indexed: 01/31/2025]
Abstract
Objective To determine circulating endothelial progenitor cells (EPC) counts and levels of inflammatory markers in individuals with and without type 1 diabetes mellitus (T1DM) in response to an intense aerobic exercise session. Subjects and methods In total, 15 adult men with T1DM and 15 healthy individuals underwent a 30-minute aerobic exercise session on a cycle ergometer at 60% of the peak heart rate. The EPC count (CD45dim/CD34+/KDR+), tumor necrosis factor-alpha (TNF-α) and interleukin 6 (IL-6) levels were measured before and 60 minutes after the session. Results We found no difference within or between groups regarding EPC counts before and after the aerobic exercise: healthy individuals, 0.02% change (95% confidence interval [CI] -0.04%-0.08%); individuals with T1DM, 0.00% (95%CI -0.01%-0.01%). We also found no difference in TNF-α levels before and after exercise in healthy individuals (210.2, interquartile range [IQR] 142.1-401.2 pg/mL and 191.3, IQR 136.4-350.5 pg/mL, respectively) and in patients with T1DM (463.8, IQR 201.4-4306.0 pg/mL and 482.7, IQR 143.8-4304.3 pg/mL, respectively). Similarly, no difference in IL-6 levels was observed before and after exercise in healthy individuals (148.2, IQR 147.5-148.6 pg/mL and 148.2, IQR 147.7-148.6 pg/mL, respectively) and individuals with T1DM (147.2, IQR 145.9-147.7 pg/mL and 147.2, IQR 146.8-147.8 pg/mL, respectively). Conclusions Patients with T1DM and healthy controls had comparable EPC responses to aerobic exercise, most likely due to the absence of a chronic inflammatory state.
Collapse
Affiliation(s)
- Patrícia Martins Bock
- Instituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em SaúdeHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilInstituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em Saúde (IATS) – CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio GrandeRio GrandeRSBrasilUniversidade Federal do Rio Grande, Rio Grande, RS, Brasil
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Raíssa Borges Monteiro
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Clara Krummenauer Maraschin
- Universidade Federal do Rio Grande do SulFaculdade de MedicinaDepartamento de Clínica MédicaPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
| | - Ana Paula Alegretti
- Serviço de Diagnóstico LaboratorialHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilServiço de Diagnóstico Laboratorial (SDLab), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Mariela Granero Farias
- Serviço de Diagnóstico LaboratorialHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilServiço de Diagnóstico Laboratorial (SDLab), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Fabiane Spagnol
- Serviço de Diagnóstico LaboratorialHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilServiço de Diagnóstico Laboratorial (SDLab), Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Patricia Luciana da Costa Lopez
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
| | - Lucas Porto Santos
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Lucas Helal
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Ruy Silveira Moraes
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Daniel Umpierre
- Instituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em SaúdeHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilInstituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em Saúde (IATS) – CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| | - Beatriz D. Schaan
- Instituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em SaúdeHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilInstituto Nacional de Ciência e Tecnologia para Avaliação de Tecnologias em Saúde (IATS) – CNPq/Brasil, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Centro de Pesquisa Clínica/Centro de Pesquisa ExperimentalHospital de Clínicas de Porto AlegrePorto AlegreRSBrasilCentro de Pesquisa Clínica/Centro de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulFaculdade de MedicinaDepartamento de Clínica MédicaPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Faculdade de Medicina, Departamento de Clínica Médica, Programa de Pós-graduação em Ciências Médicas: Endocrinologia, Porto Alegre, RS, Brasil
- Universidade Federal do Rio Grande do SulPorto AlegreRSBrasilUniversidade Federal do Rio Grande do Sul, Programa de Pós-graduação em Ciências da Saúde: Cardiologia, Porto Alegre, RS, Brasil
| |
Collapse
|