1
|
Xu S, Chuang CY, Hawkins CL, Hägglund P, Davies MJ. Quantitative analysis of the proteome and protein oxidative modifications in primary human coronary artery endothelial cells and associated extracellular matrix. Redox Biol 2025; 81:103524. [PMID: 39954365 PMCID: PMC11875191 DOI: 10.1016/j.redox.2025.103524] [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: 12/05/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025] Open
Abstract
Vascular endothelial cells (ECs) play a key role in physiology by controlling arterial contraction and relaxation, and molecular transport. EC dysfunction is associated with multiple pathologies. Here, we characterize the cellular and extracellular matrix (ECM) proteomes of primary human coronary artery ECs, from multiple donors, and oxidation/nitration products formed on these during cell culture, using liquid chromatography-mass spectrometry. In total ∼9900 proteins were identified in cells from 3 donors, with ∼7000 proteins per donor. Of these ∼5300 were consistently identified, indicating some heterogeneity across the donors, with age a possible cause. Multiple endogenous oxidation products were detected on both ECM and cellular proteins (and particularly endoplasmic reticulum species). In contrast, nitration was mostly detected on cell proteins and particularly cytoskeletal proteins, consistent with intracellular generation of nitrating agents, possibly from endothelial nitric oxide synthase (eNOS) or peroxidase enzymes. The modifications are ascribed to both physiological enzymatic activity (hydroxylation at proline/lysine; predominantly on ECM proteins and especially collagens) and the formation of reactive species (oxidation at tryptophan/tyrosine/histidine; nitration at tryptophan/tyrosine). The identified sites are present on a limited number of peptides (104 oxidized; 23 nitrated) from a modest number of proteins. A small number of proteins were detected with multiple modifications, consistent with these being selective and specific targets. Several nitrated peptides were consistently detected across all donors, and also in human smooth muscle cells suggesting that these are major targets in the vascular proteome. These data provide a 'background' proteome dataset for studies of endothelial dysfunction in disease.
Collapse
Affiliation(s)
- Shuqi Xu
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark; Department of Cardiovascular Medicine, The Affiliated Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Christine Y Chuang
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Clare L Hawkins
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark
| | - Per Hägglund
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| | - Michael J Davies
- Department of Biomedical Sciences, Panum Institute, University of Copenhagen, Denmark.
| |
Collapse
|
2
|
Zhong J, Gao RR, Zhang X, Yang JX, Liu Y, Ma J, Chen Q. Dissecting endothelial cell heterogeneity with new tools. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:10. [PMID: 40121354 PMCID: PMC11929667 DOI: 10.1186/s13619-025-00223-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/25/2025]
Abstract
The formation of a blood vessel network is crucial for organ development and regeneration. Over the past three decades, the central molecular mechanisms governing blood vessel growth have been extensively studied. Recent evidence indicates that vascular endothelial cells-the specialized cells lining the inner surface of blood vessels-exhibit significant heterogeneity to meet the specific needs of different organs. This review focuses on the current understanding of endothelial cell heterogeneity, which includes both intra-organ and inter-organ heterogeneity. Intra-organ heterogeneity encompasses arterio-venous and tip-stalk endothelial cell specialization, while inter-organ heterogeneity refers to organ-specific transcriptomic profiles and functions. Advances in single-cell RNA sequencing (scRNA-seq) have enabled the identification of new endothelial subpopulations and the comparison of gene expression patterns across different subsets of endothelial cells. Integrating scRNA-seq with other high-throughput sequencing technologies promises to deepen our understanding of endothelial cell heterogeneity at the epigenetic level and in a spatially resolved context. To further explore human endothelial cell heterogeneity, vascular organoids offer powerful tools for studying gene function in three-dimensional culture systems and for investigating endothelial-tissue interactions using human cells. Developing organ-specific vascular organoids presents unique opportunities to unravel inter-organ endothelial cell heterogeneity and its implications for human disease. Emerging technologies, such as scRNA-seq and vascular organoids, are poised to transform our understanding of endothelial cell heterogeneity and pave the way for innovative therapeutic strategies to address human vascular diseases.
Collapse
Affiliation(s)
- Jing Zhong
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Rong-Rong Gao
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan 250117, Shandong, China
| | - Xin Zhang
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
- University of Chinese Academy of Sciences, Beijing, China
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Jia-Xin Yang
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China
| | - Yang Liu
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
| | - Jinjin Ma
- The Innovation Centre of Ministry of Education for Development and Diseases, School of Medicine, South China University of Technology, Guangzhou, 510006, China.
- The Institute of Future Health, South China of Technology, Guangzhou International Campus, Guangzhou, 511442, China.
| | - Qi Chen
- Center for Cell Lineage Atlas, CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
- China-New Zealand Belt and Road Joint Laboratory on Biomedicine and Health, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangdong-Hong Kong Joint Laboratory for Stem Cell and Regenerative Medicine, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Center for Cell Lineage Atlas, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China.
- Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences); Key Lab for Rare & Uncommon Diseases of Shandong Province, Ji'nan 250117, Shandong, China.
| |
Collapse
|
3
|
He J, Chen Y, Li Y, Feng Y. Molecular mechanisms and therapeutic interventions in acute kidney injury: a literature review. BMC Nephrol 2025; 26:144. [PMID: 40121405 PMCID: PMC11929251 DOI: 10.1186/s12882-025-04077-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025] Open
Abstract
Acute kidney injury (AKI) is a clinical challenge characterized by elevated morbidity and a substantial impact on individual health and socioeconomic factors. A comprehensive examination of the molecular pathways behind AKI is essential for its prevention and management. In recent years, vigorous research in the domain of AKI has concentrated on pathophysiological characteristics, early identification, and therapeutic approaches across many aetiologies and highlighted the principal themes of oxidative stress, inflammatory response, apoptosis, necrosis, and immunological response. This review comprehensively reviewed the molecular mechanisms underlying AKI, including oxidative stress, inflammatory pathways, immune cell-mediated injury, activation of the renin-angiotensin-aldosterone (RAAS) system, mitochondrial damage and autophagy, apoptosis, necrosis, etc. Inflammatory pathways are involved in the injuries in all four structural components of the kidney. We also summarized therapeutic techniques and pharmacological agents associated with the aforementioned molecular pathways. This work aims to clarify the molecular mechanisms of AKI thoroughly, offer novel insights for further investigations of AKI, and facilitate the formulation of efficient therapeutic methods to avert the progression of AKI.
Collapse
Affiliation(s)
- Jiajia He
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yanqin Chen
- Department of Nephrology, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Yi Li
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China
| | - Yunlin Feng
- Department of Nephrology and Institute of Nephrology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Sichuan Clinical Research Centre for Kidney Diseases, Chengdu, 610072, China.
| |
Collapse
|
4
|
Xu M, Zhang QT, Zhou L, Cai YW, Liu H, Zhao QL, Tian JH, Huang YK, Ren P, Huang X. Ferulic acid in Chaihu Shugan San modulates depression-like behavior, endothelial and gastrointestinal dysfunction in rats via the Ghrl-Edn1/Mecp2/P-mTOR/VEGFA pathway: A multi-omics study. JOURNAL OF ETHNOPHARMACOLOGY 2025; 346:119624. [PMID: 40127829 DOI: 10.1016/j.jep.2025.119624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 01/16/2025] [Accepted: 03/10/2025] [Indexed: 03/26/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE In a global context of escalating multimorbidity, characterized by the co-occurrence of major depressive disorder (MDD), endothelial dysfunction (ED), and gastrointestinal dysregulation (GD), the quest for effective treatments has become paramount. Central to these interconnected conditions is oxidative stress (OS), a pivotal factor that has been extensively studied yet remains inadequately addressed. This study introduces Chaihu-Shugan-San (CSS) and its absorbed component ferulic acid (FA), a potent antioxidant derived from medicinal plants, as a novel therapeutic approach with the unique ability to counter the multifaceted effects of acute forced swimming (AFS)-induced depression, ED, and GD. Unlike traditional single-disease-focused studies, our research explores the synergistic effects of CSS and FA across these interrelated disorders, offering a groundbreaking perspective. AIM OF THE STUDY This study aims to evaluate CSS and FA in treating depression-related multimorbidity triggered by AFS and to uncover the shared underlying mechanisms of FA. MATERIALS AND METHODS A depression-like model in rats was induced by AFS, and an OS model was established in endothelial cells (ECs) through hydrogen peroxide treatment. We investigated the effects of CSS and FA on MDD, ED, and GD in rats and OS levels in ECs. Our assessments included hematoxylin and eosin (HE) staining, biochemical assays, and behavioral studies. We conducted an integrated analysis of transcriptomics, proteomics, and phosphoproteomics data to elucidate the underlying mechanisms. The identification of relevant targets was confirmed through Western blotting (WB), real-time quantitative polymerase chain reaction (RT-qPCR), molecular docking studies, and an extensive literature review. RESULTS Our findings indicate that CSS and FA not only significantly mitigate AFS-induced abnormalities in the open field test (OFT), forced swim test (FST), and related behaviors such as gastric emptying and intestinal transit in rats but also ameliorate depression, ED, GD, inflammation and OS-related biomarker levels, alongside HE staining in gastric sinus and aorta slices. The study also highlights that FA can influence OS and endothelial function in ECs. Moreover, a combined multi-omics analysis unveiled several OS-related pathways, including the mTOR and p53 signaling pathways. Our research elucidates that the Ghrl-Edn1/Mecp2/P-mTOR/Vegfa-associated OS signaling pathway is pivotal in countering AFS-induced multimorbidity, expanding beyond the conventional disease-specific focus. CONCLUSIONS This pioneering study underscores capability of CSS and FA to tackle AFS-induced multimorbidity concurrently and intricately details FA's antioxidative mechanisms within ECs. The insights gleaned offer a novel perspective on FA's role in multimorbidity regulation and its potential to modulate OS, especially in the complex environment of ECs. Given the urgent global health challenges, our research positions FA as a promising therapeutic contender, advocating for a paradigm shift in multimorbidity management.
Collapse
Affiliation(s)
- Min Xu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qian-Tao Zhang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Zhou
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ya-Wen Cai
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Hao Liu
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Qiu-Long Zhao
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing-Hua Tian
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China
| | - Yun-Ke Huang
- Women's Hospital, Zhejiang University School of Medicine, Gynecology Department, Zhejiang, China
| | - Ping Ren
- Department of Geriatrics, Jiangsu Province Hospital of TCM, Nanjing, China
| | - Xi Huang
- Institute of TCM-Related Comorbid Depression, Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
5
|
Sheptulina AF, Liusina EO, Zlobovskaya OA, Kiselev AR, Drapkina OM. Possible Role of Platelets in the Development and Progression of Non-Alcoholic Fatty Liver Disease. FRONT BIOSCI-LANDMRK 2025; 30:26748. [PMID: 40152376 DOI: 10.31083/fbl26748] [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: 09/26/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 03/29/2025]
Abstract
To date, an increasing body of evidence supports the potential role of activated platelets in the pathogenesis of non-alcoholic fatty liver disease (NAFLD). This is likely due to their ability to secrete biologically active substances that regulate liver regeneration processes, ensure hemostasis, and participate in the immune response. Additionally, several studies have demonstrated the efficacy of antiplatelet agents in reducing inflammation, the severity of liver fibrosis, and the progression of fibrosis in non-alcoholic steatohepatitis (NASH). Since NAFLD is not an independent indication for antiplatelet therapy, the primary evidence regarding their efficacy in NAFLD has been derived from studies using animal models of NAFLD or in patients with concomitant cardiovascular diseases. This narrative review will discuss the main functions of platelets, their unique interactions with liver cells, and the outcomes of these interactions, as well as the results of studies evaluating the efficacy and safety of antiplatelet therapy in patients with NAFLD.
Collapse
Affiliation(s)
- Anna F Sheptulina
- Department of Fundamental and Applied Aspects of Obesity, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
- Department of Therapy and Preventive Medicine, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
| | - Ekaterina O Liusina
- Department of Fundamental and Applied Aspects of Obesity, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
| | - Olga A Zlobovskaya
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 123182 Moscow, Russia
| | - Anton R Kiselev
- Coordinating Center for Fundamental Research, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
| | - Oxana M Drapkina
- Department of Fundamental and Applied Aspects of Obesity, National Medical Research Center for Therapy and Preventive Medicine, 101990 Moscow, Russia
- Department of Therapy and Preventive Medicine, A.I. Evdokimov Moscow State University of Medicine and Dentistry, 127473 Moscow, Russia
| |
Collapse
|
6
|
Čiapienė I, Vėžys J, Lesauskaitė V, Matulevičiūtė I, Meškauskaitė U, Skipskis V, Strazdauskas A, Trumbeckaitė S, Bubulis A, Jūrėnas V, Ostaševičius V, Tamakauskas V, Tatarūnas V. Synergistic Effects of Low-Frequency Ultrasound and Therapeutic Agents on Endothelial and Renal Cells: Emphasis on Cell Functionality, Oxidative Stress, and Inflammatory Markers. Pharmaceuticals (Basel) 2025; 18:404. [PMID: 40143180 PMCID: PMC11945135 DOI: 10.3390/ph18030404] [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: 02/12/2025] [Revised: 03/06/2025] [Accepted: 03/10/2025] [Indexed: 03/28/2025] Open
Abstract
Background: Ischemic heart disease remains the leading cause of death worldwide, with coronary microvascular dysfunction (CMD) as a key complication after ST-elevation myocardial infarction (STEMI). Endothelial dysfunction contributes to CMD, impairing vascular tone and increasing inflammation. While angiotensin-converting enzyme (ACE) inhibitors and angiotensin II receptor blockers (ARBs) aid vascular health, their efficacy may improve with therapeutic ultrasound, which enhances drug delivery and endothelial response. This study explores the combined effects of ultrasound and pharmacological treatment on the ACE axis and inflammation in endothelial and renal cells. Methods: Human umbilical vein endothelial cells (HUVECs) and human renal proximal tubular epithelial cell line RPTEC/TERT1 were treated with captopril, losartan, and dexamethasone, alone or combined with low-frequency ultrasound (LFU). Cell viability and wound-healing assays assessed cellular function, while nitric oxide (NO) and reactive oxygen species (ROS) assays were used to evaluate redox signaling. Gene expression related to the ACE axis, inflammation, and vascular and renal cell function was analyzed via qPCR. Results: Captopril and losartan combined with LFU improved endothelial cell viability, wound healing, and NO production at various concentrations, whereas only losartan with LFU enhanced cell viability and wound healing in renal cells. Dexamethasone with LFU increased ROS levels and had variable effects on RPTEC/TERT1 cell survival. Gene expression analysis showed that LFU alone reduced pro-inflammatory markers VCAM-1, ICAM-1, and PTGS2 in captopril-treated HUVECs and similarly affected CYP4F2 in losartan-treated HUVECs. LFU also decreased PTGS2 expression at higher dexamethasone concentrations. In RPTEC/TERT1 cells, LFU alone did not impact SGLT2 or GGT1 expression, but captopril with LFU downregulated GGT1, and dexamethasone with LFU upregulated SGLT2 at higher concentrations. Conclusions: This study demonstrates that LFU enhances the effects of RAS inhibitors by promoting NO synthesis and reducing oxidative stress, while its combination with dexamethasone may have variable, potentially cytotoxic effects on renal cells. Gene expression patterns suggest LFU's anti-inflammatory potential and its role in modulating drug efficacy.
Collapse
Affiliation(s)
- Ieva Čiapienė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Joris Vėžys
- Department of Mechanical Engineering, Faculty of Mechanical Engineering and Design, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania;
| | - Vaiva Lesauskaitė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Indrė Matulevičiūtė
- Department of Ophthalmology, Lithuanian University of Health Sciences, Eiveniu 2, LT-50161 Kaunas, Lithuania;
| | - Ugnė Meškauskaitė
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Vilius Skipskis
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Arvydas Strazdauskas
- Department of Biochemistry, Faculty of Medicine, Lithuanian University of Health Sciences, Eiveniu 4, LT-50161 Kaunas, Lithuania;
| | - Sonata Trumbeckaitė
- Department of Pharmacognosy, Faculty of Pharmacy, Lithuanian University of Health Sciences, Sukileliu 13, LT-50162 Kaunas, Lithuania;
| | - Algimantas Bubulis
- Institute of Mechatronics, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania; (A.B.); (V.J.); (V.O.)
| | - Vytautas Jūrėnas
- Institute of Mechatronics, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania; (A.B.); (V.J.); (V.O.)
| | - Vytautas Ostaševičius
- Institute of Mechatronics, Kaunas University of Technology, Studentu 56, LT-51424 Kaunas, Lithuania; (A.B.); (V.J.); (V.O.)
| | - Vytenis Tamakauskas
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| | - Vacis Tatarūnas
- Institute of Cardiology, Lithuanian University of Health Sciences, Sukileliu 15, LT-50103 Kaunas, Lithuania; (I.Č.); (V.L.); (U.M.); (V.S.); (V.T.)
| |
Collapse
|
7
|
Mostafa RE, Ali DE, El-Shiekh RA, El-Alfy AN, Hafeez MSAE, Reda AM, Fayek NM. Therapeutic applications of natural products in the management of venous diseases: a comprehensive review. Inflammopharmacology 2025:10.1007/s10787-025-01688-z. [PMID: 40074995 DOI: 10.1007/s10787-025-01688-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025]
Abstract
The occurrence of venous diseases among adults is approximately 77% in females and 57% in males. These conditions are prevalent, progressive disorders that significantly affect individuals socially, physically, and psychologically, often resulting in various venous abnormalities that hinder effective blood circulation in the lower limbs. This review provides a comprehensive overview of venous diseases, focusing on their pathophysiology, symptoms, causes, risk factors, diagnosis, and complications. The symptoms associated with venous diseases are diverse and can include pain, heaviness, swelling, ulcers, and skin changes. Risk factors such as age, obesity, hormonal influences, and genetic predispositions are discussed in relation to their contribution to disease progression. The therapeutic modalities for managing venous diseases are explored, with a particular emphasis on natural products in alleviating symptoms and improving vascular health. Natural compounds, i.e., flavonoids, play a vital role in the circulatory system, supporting blood vessels and promoting healthy blood flow, in addition to their vasoprotective, antioxidant, anti-inflammatory, and anti-platelet properties. Overall, the ongoing research efforts on the efficacy of natural products will significantly enhance the management of several venous diseases in the coming years.
Collapse
Affiliation(s)
- Rasha E Mostafa
- Pharmacology Department, Medical Research and Clinical Studies Institute, National Research Centre, 33 El-Bohouth St., Dokki, P.O. 12622, Cairo, Egypt
| | - Dalia E Ali
- Pharmacognosy and Natural Products Department, Faculty of Pharmacy, Pharos University in Alexandria, Alexandria, 21648, Egypt
| | - Riham A El-Shiekh
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Ahmed N El-Alfy
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, 11829, Egypt
| | - Mohamed S Abd El Hafeez
- Department of Pharmacognosy, Faculty of Pharmacy, Egyptian Russian University, Cairo-Suez Road, Badr City, 11829, Egypt
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
| | - Ahmed M Reda
- Department of Pharmacy, Kut University College, Al Kut, Wasit, 52001, Iraq
- Department of Biochemistry, Faculty of Pharmacy, Egyptian Russian University, Badr City, Cairo, Egypt
| | - Nesrin M Fayek
- Department of Pharmacognosy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
8
|
Saputro RE, Chou CC, Lin YY, Tarumi T, Liao YH. Exercise-mediated modulation of autonomic nervous system and inflammatory response in sleep-deprived individuals: A narrative reviews of implications for cardiovascular health. Auton Neurosci 2025; 259:103256. [PMID: 40073691 DOI: 10.1016/j.autneu.2025.103256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/25/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025]
Abstract
Sleep deprivation is a growing concern in cardiovascular risk, causing physiological disruptions like autonomic dysregulation and inflammation. Recent research indicates that sleep deprivation increases sympathetic nervous activity while decreasing parasympathetic activity, leading to increased blood pressure, impaired endothelial function, and heightened inflammation. Exercise has emerged as a non-pharmacological approach to increase cardiovascular health. However, the impact of exercise on sleep deprivation-induced changes in autonomic activity and inflammation remains unclear. To explore this, we reviewed studies investigating the effects of acute exercise on autonomic regulation and inflammatory markers following sleep deprivation. We conducted a narrative review of the literature. PubMed/MEDLINE, Google Scholar, and Web of Science (WOS) searched the articles between May 2022 and April 2023. The papers had to: [1] focus on recent studies between 2000 and 2023; [2] consist of sleep deprivation participants; [3] be published in English. Acute moderate- to high-intensity exercise after sleep deprivation may reduce parasympathetic activity, trigger pro-inflammatory cytokines, and delay recovery to normal levels. In contrast, regular exercise routines may mitigate the adverse effects of sleep deprivation on autonomic regulation and reduce systemic inflammation. Sleep deprivation can lead to autonomic imbalance, increased blood pressure, and increased inflammatory responses, which are further amplified by acute exercise, increasing the cardiovascular burden. When sleep deprivation occurs, exercise intensity and timing should be carefully chosen to avoid adverse cardiovascular health risks.
Collapse
Affiliation(s)
- Riki Edo Saputro
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan; Department of Physical Education, Universitas Wahid Hasyim, Semarang 50224, Indonesia
| | - Chun-Chung Chou
- Physical Education Office, National Taipei University of Technology, Taipei 10608, Taiwan
| | - Yi-Yuan Lin
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan.
| | - Takashi Tarumi
- Human Informatics and Interaction Research Institute, National Institute of Advanced Industrial Science and Technology, Higashi, Tsukuba, Ibaraki 305-8566, Japan
| | - Yi-Hung Liao
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei 11219, Taiwan.
| |
Collapse
|
9
|
Wu J, Wang J, Pei Z, Zhu Y, Zhang X, Zhou Z, Ye C, Song M, Hu Y, Xue P, Zhao G. Endothelial senescence induced by PAI-1 promotes endometrial fibrosis. Cell Death Discov 2025; 11:89. [PMID: 40050610 PMCID: PMC11885584 DOI: 10.1038/s41420-025-02377-0] [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: 01/14/2025] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/09/2025] Open
Abstract
Intrauterine adhesions (IUAs), also known as Asherman's syndrome (AS), represent a significant cause of uterine infertility for which effective treatment remains elusive. The endometrium's ability to regenerate cyclically depends heavily on the growth and regression of its blood vessels. However, trauma to the endometrial basal layer can disrupt the subepithelial capillary plexus, impeding regeneration. This damage results in the replacement of native cells with fibroblasts and myofibroblasts, ultimately leading to fibrosis. Endothelial cells (ECs) play a pivotal role in the vascular system, extending beyond their traditional barrier function. Through single-cell sequencing and experimental validation, we discovered that ECs undergo senescence in IUA patients, impairing angiogenesis and fostering stromal cell fibrosis. Further analysis revealed significant interactions between ECs and PAI-1+ stromal cells. PAI-1, derived from stromal cells, promotes EC senescence via the urokinase-type plasminogen activator receptor (uPAR). Notably, prior to fibrosis onset, TGF-β upregulates PAI-1 expression in stromal cells in a SMAD dependent manner. In an IUA mouse model, inhibiting PAI-1 mitigated EC senescence and endometrial fibrosis. Our findings underscore the crucial role of EC senescence in IUA pathogenesis, contributing to vascular reduction and fibrosis. Targeting PAI-1 represents a promising therapeutic strategy to suppress EC senescence and alleviate endometrial fibrosis, offering new insights into the treatment of IUAs.
Collapse
Affiliation(s)
- Jing Wu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Wang
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhongrui Pei
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Yaru Zhu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xier Zhang
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zihan Zhou
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Chunying Ye
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Minmin Song
- Obstetrics and Gynaecology Hospital, Fudan University, Shanghai, China
| | - Yali Hu
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Pingping Xue
- Department of Reproductive Medicine Center, Changzhou Maternal and Child Health Care Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, China.
| | - Guangfeng Zhao
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Obstetrics and Gynecology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| |
Collapse
|
10
|
Gan Q, Chi H, Meng X, Tang X, Xing J, Sheng X, Zhan W. Von Willebrand Factor in Weibel-Palade Bodies of Endothelial Cells Involved in the Immune Response to Hirame Novirhabdovirus Infection in a Teleost Paralichthys olivaceus. JOURNAL OF FISH DISEASES 2025; 48:e14069. [PMID: 39660701 DOI: 10.1111/jfd.14069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 12/12/2024]
Abstract
von Willebrand factor (vWF) is a large multimeric sialoglycoprotein that plays key roles in normal haemostasis, inflammation regulation, angiogenesis and cancer metastasis in mammals. The gene, protein sequences and functions of vWF in flounder Paralichthys olivaceus (PovWF) were analysed in this study. PovWF possesses an 8550-bp open reading frame (ORF) that encodes a 2849 amino acid protein. PovWF mRNA is highly expressed in the heart and gill, followed by the intestine, skin, spleen, kidney, muscle and liver. PovWF positive cells are mainly endothelial cells (ECs), predominantly located along the inner lining of blood vessels, enclosing the bloodstream. After being infected with hirame novirhabdovirus (HIRRV), flounder exhibits a dark body colour, congested fins and visceral membranes. Histopathologic analysis revealed that the ECs of diseased fish had compromised integrity, accompanied by a significant increase in number of cells within blood vessels. Immunofluorescence and ultrastructural studies showed that virions infect ECs can induce morphological and functional alterations, which lead to the release of vWF and facilitate the migration of neutrophils into tissues to exert antiviral functions. This research pinpoints the role of vWF in the immune response to HIRRV infection in teleost. It offers an in-depth and all-encompassing understanding of the pathophysiological interaction between HIRRV and endothelial cells during invasive infections in fish.
Collapse
Affiliation(s)
- Qiujie Gan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Heng Chi
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Xianghu Meng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
| | - Xiaoqian Tang
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Jing Xing
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Xiuzhen Sheng
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
| | - Wenbin Zhan
- Laboratory of Pathology and Immunology of Aquatic Animals, KLMME, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Qingdao Marine Science and Technology Center, Qingdao, China
| |
Collapse
|
11
|
Guan T, Zhang W, Li M, Wang Q, Guo L, Guo B, Luo X, Li Z, Lu M, Dong Z, Xu M, Liu M, Liu Y, Feng J. D-Ala2-GIP (1-30) promotes angiogenesis by facilitating endothelial cell migration via the Epac/Rap1/Cdc42 signaling pathway. Cell Signal 2025; 127:111615. [PMID: 39855534 DOI: 10.1016/j.cellsig.2025.111615] [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: 09/20/2024] [Revised: 01/07/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
Angiogenesis, a meticulously regulated process essential for both normal development and pathological conditions, necessitates a comprehensive understanding of the endothelial mechanisms governing its progression. Leveraging the zebrafish model and NgAgo knockdown system to identify target genes influencing angiogenesis, our study highlights the significant role of gastric inhibitory polypeptide (GIP) and its receptor (GIPR) in this process. While GIP has been extensively studied for its insulinotropic and glucagonotropic effects, its role in angiogenesis remains unexplored. This study demonstrated that GIPR knockdown induced developmental delays, morphological abnormalities, and pronounced angiogenic impairments in zebrafish embryos. Conversely, exogenous D-Ala2-GIP administration enhanced blood vessel formation in the yolk sac membrane of chick embryos. Consistent with these findings, D-Ala2-GIP treatment promoted microvessel formation in the tube formation assays and rat aortic ring models. Further investigation revealed that D-Ala2-GIP facilitated human umbilical vein endothelial cell (HUVEC) migration, a key step in angiogenesis, through the cyclic adenosine monophosphate (cAMP)-mediated activation of the Epac/Rap1/Cdc42 signaling pathway. This study provides novel insights into the angiogenic functions of GIP and its potential implications for cardiovascular biology.
Collapse
Affiliation(s)
- Tuchen Guan
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Wenxue Zhang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Mingxuan Li
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Qing Wang
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Longyu Guo
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Beibei Guo
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Xiaoqian Luo
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Zhen Li
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Muxing Lu
- Medical School of Nantong University, Nantong, Jiangsu Province 226001, China
| | - Zhangji Dong
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Man Xu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Mei Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China
| | - Yan Liu
- Key Laboratory of Neuroregeneration of Jiangsu Province and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province 226001, China.
| | - Jian Feng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital and Medical School of Nantong University, Nantong, Jiangsu Province 226001, China.
| |
Collapse
|
12
|
Zhang S, Wu J, Wang L, Zhang C, Zhang Y, Feng Y. Exploring the hepatic-ophthalmic axis through immune modulation and cellular dynamics in diabetic retinopathy and non-alcoholic fatty liver disease. Hum Genomics 2025; 19:19. [PMID: 40011971 DOI: 10.1186/s40246-025-00730-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 02/13/2025] [Indexed: 02/28/2025] Open
Abstract
BACKGROUND Dysfunctions within the liver system are intricately linked to the progression of diabetic retinopathy (DR) and non-alcoholic fatty liver disease (NAFLD). This study leverages systematic analysis to elucidate the complex cross-talk and communication pathways among diverse cell populations implicated in the pathogenesis of DR and NAFLD. METHODS Single-cell RNA sequencing data for proliferative diabetic retinopathy (PDR) and NAFLD were retrieved from the Gene Expression Omnibus (GEO) database. Differential gene expression analysis was conducted and followed by pseudo-time analysis to delineate dynamic changes in core cells and differentially expressed genes (DEGs). CellChat was employed to predict intercellular communication and signaling pathways. Additionally, gene set enrichment and variation analyses (GSEA and GSVA) were performed to uncover key functional enrichments. RESULTS Our comparative analysis of the two datasets focused on T cells, macrophages and endothelial cells, revealing SYNE2 as a notable DEG. Notably, common genes including PYHIN1, SLC38A1, ETS1 (T cells), PPFIBP1, LIFR, HSPG2 (endothelial cells), and MSR1 (macrophages), emerged among the top 50 DEGs across these cell types. The CD45 signaling pathway was pivotal for T cells and macrophages, exerting profound effects on other cells in both PDR and NAFLD. Moreover, GSEA and GSVA underscored their involvement in cellular communication, immune modulation, energy metabolism, mitotic processes. CONCLUSION The comprehensive investigation of T cells, macrophages, endothelial cells, and the CD45 signaling pathway advances our understanding of the intricate biological processes underpinning DR and NAFLD. This research underscores the imperative of exploring immune-related cell interactions, shedding light on novel therapeutic avenues in these disease contexts.
Collapse
Affiliation(s)
- Shuyan Zhang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Jiajun Wu
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Leilei Wang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai Eye Disease Prevention and Treatment Center, Shanghai, China
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Yinjian Zhang
- Department of Ophthalmology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China.
| |
Collapse
|
13
|
Zhang S, Guo J, He Y, Su Z, Feng Y, Zhang L, Jun Z, Weng X, Yuan Y. Roles of lncRNA in the crosstalk between osteogenesis and angiogenesis in the bone microenvironment. J Zhejiang Univ Sci B 2025; 26:107-123. [PMID: 40015932 PMCID: PMC11867785 DOI: 10.1631/jzus.b2300607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/16/2024] [Indexed: 03/01/2025]
Abstract
Bone is a highly calcified and vascularized tissue. The vascular system plays a vital role in supporting bone growth and repair, such as the provision of nutrients, growth factors, and metabolic waste transfer. Moreover, the additional functions of the bone vasculature, such as the secretion of various factors and the regulation of bone-related signaling pathways, are essential for maintaining bone health. In the bone microenvironment, bone tissue cells play a critical role in regulating angiogenesis, including osteoblasts, bone marrow mesenchymal stem cells (BMSCs), and osteoclasts. Osteogenesis and bone angiogenesis are closely linked. The decrease in osteogenesis and bone angiogenesis caused by aging leads to osteoporosis. Long noncoding RNAs (lncRNAs) are involved in various physiological processes, including osteogenesis and angiogenesis. Recent studies have shown that lncRNAs could mediate the crosstalk between angiogenesis and osteogenesis. However, the mechanism by which lncRNAs regulate angiogenesis‒osteogenesis crosstalk remains unclear. In this review, we describe in detail the ways in which lncRNAs regulate the crosstalk between osteogenesis and angiogenesis to promote bone health, aiming to provide new directions for the study of the mechanism by which lncRNAs regulate bone metabolism.
Collapse
Affiliation(s)
- Shihua Zhang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Jianmin Guo
- School of Life Sciences, South University of Science and Technology, Shenzhen 518055, China
| | - Yuting He
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Zhi'ang Su
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Yao Feng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Lan Zhang
- College of Sports and Health, Shandong Sport University, Jinan 250102, China
| | - Zou Jun
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China
| | - Xiquan Weng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China. ,
| | - Yu Yuan
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China.
| |
Collapse
|
14
|
Liu X, Liu H, Wang N, Lai S, Qiu C, Gao S, Huang T, Zhang W. The interactive toxic effect of homocysteine and copper on cardiac microvascular endothelial cells during ischemia-reperfusion injury. Chem Biol Interact 2025; 408:111387. [PMID: 39824432 DOI: 10.1016/j.cbi.2025.111387] [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] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/20/2025]
Abstract
Hyperhomocysteinemia (HHcy) is associated with the development and progression of chronic cardiovascular diseases through the deleterious effects of high levels of homocysteine (Hcy) on the cardiovascular system. However, the exact mechanism of action of Hcy on the acute injury of the cardiovascular system following ischemia/reperfusion (I/R) remains unclear. The present study demonstrated that copper mobilization occurs during cardiac I/R, and the interactive toxic effect of Hcy and mobile Cu2+ during cardiac I/R induces necroptosis of cardiac microvascular endothelial cells (CMECs) and thus enhances cardiac dysfunction. In the present study, we utilized three cardiac I/R model: isolated rat heart, in vivo model as well as cell culture, and demonstrated that copper mobilization occurs during cardiac I/R, and the interactive toxic effect of Hcy and mobile Cu2+ during cardiac I/R induces necroptosis of cardiac microvascular endothelial cells (CMECs) and thus enhances cardiac dysfunction. Furthermore, we proved that the Cu2+ chelator TTM significantly mitigated the deleterious effects of Hcy and Cu2+ on CMECs and cardiac function both in vitro and in vivo. Mechanismly, the combinative effect of Hcy and Cu2+ are associated with the production of reactive oxygen species (ROS) and nitric oxide (NO) by NADPH oxidase (NOX) and endothelial nitric oxide synthase (eNOS), respectively. Subsequently, the overproduction of toxic peroxynitrite (ONOO-) induces CMECs necroptosis. The application of ROS scavengers in CMECs resulted in a notable reduction in necroptosis mediated by Hcy and Cu2+ under hypoxia/reperfusion (H/R) condition. These findings indicate that the mechanism by which Hcy and Cu2+ enhances cardiac dysfunction under I/R condition may be attributed to the stimulation of both NOX and eNOS activity, resulting in the generation of excessive ONOO- and subsequent necroptosis of CMECs.
Collapse
Affiliation(s)
- Xiaoming Liu
- Department of Thoracic Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine, Nanchang, 330000, Jiangxi, PR China; Jiangxi Institute of Respiratory Disease, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, PR China
| | - Haipeng Liu
- Department of Thoracic Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine, Nanchang, 330000, Jiangxi, PR China; Jiangxi Institute of Respiratory Disease, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, PR China
| | - Ning Wang
- Department of Thoracic Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine, Nanchang, 330000, Jiangxi, PR China; Jiangxi Institute of Respiratory Disease, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, PR China
| | - Songqing Lai
- Department of Cardiovascular Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China
| | - Chengpeng Qiu
- Department of Thoracic Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine, Nanchang, 330000, Jiangxi, PR China; Jiangxi Institute of Respiratory Disease, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, PR China
| | - Shansong Gao
- Department of Thoracic Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine, Nanchang, 330000, Jiangxi, PR China; Jiangxi Institute of Respiratory Disease, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, PR China
| | - Tianxiang Huang
- Department of Thoracic Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine, Nanchang, 330000, Jiangxi, PR China
| | - Wan Zhang
- Department of Thoracic Surgery, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi, PR China; Jiangxi Hospital of China-Japan Friendship Hospital, National Regional Center for Respiratory Medicine, Nanchang, 330000, Jiangxi, PR China; Jiangxi Institute of Respiratory Disease, The 1st Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330000, Jiangxi, PR China.
| |
Collapse
|
15
|
Xu XX, Tian Y, Pu Y, Che B, Luo H, Liu Y, Liu YJ, Jing G. Bacterial Swimming and Accumulation on Endothelial Cell Surfaces. J Phys Chem B 2025. [PMID: 39983743 DOI: 10.1021/acs.jpcb.4c08666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2025]
Abstract
Flagellar-driven locomotion plays a critical role in the bacterial attachment and colonization of surfaces, contributing to the risks of contamination and infection. Extensive efforts to uncover the underlying principles governing bacterial motility near surfaces have relied on idealized assumptions about surrounding artificial surfaces. However, in the context of living systems, the role of cells from tissues and organs becomes increasingly critical, particularly in bacterial swimming and adhesion, yet it remains poorly understood. Here, we propose using biological surfaces composed of vascular endothelial cells to experimentally investigate bacterial motion and interaction behaviors. Our results reveal that bacterial trapping observed on inorganic surfaces is counteractively manifested with reduced radii of circular motion on cellular surfaces. Additionally, two distinct modes of bacterial adhesion were identified: tight and loose adhesion. Interestingly, the presence of living cells enhances bacterial surface enrichment, and imposed flow intensifies this accumulation via a bias-swimming effect. These results surprisingly indicate that physical effects remain the dominant factor regulating bacterial motility and accumulation at the single-cell-layer level in vitro, bridging the gap between simplified hydrodynamic mechanisms and complex biological surfaces with relevance to biofilm formation and bacterial contamination.
Collapse
Affiliation(s)
- Xin-Xin Xu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Yangguang Tian
- School of Physics, Northwest University, Xi'an 710127, China
| | - Yuhe Pu
- School of Physics, Northwest University, Xi'an 710127, China
| | - Bingchen Che
- Department of Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education), Tsinghua University, Beijing 100084, China
| | - Hao Luo
- School of Physics, Northwest University, Xi'an 710127, China
| | - Yanan Liu
- School of Physics, Northwest University, Xi'an 710127, China
| | - Yan-Jun Liu
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Guangyin Jing
- School of Physics, Northwest University, Xi'an 710127, China
| |
Collapse
|
16
|
Rimmer M, J Peacock W, Shivkumar K, Mori S. Rare bilateral anatomical variation of the lateral thoracic artery: duplicated arteries with unique origins and pathways. Surg Radiol Anat 2025; 47:80. [PMID: 39966115 PMCID: PMC11836202 DOI: 10.1007/s00276-025-03587-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2025] [Accepted: 01/28/2025] [Indexed: 02/20/2025]
Abstract
The lateral thoracic artery (LTA) is one of six main branches that originate from the axillary artery. The LTA has a textbook origin from the 2nd part of the axillary artery posterior to the pectoralis minor muscle. Contrary to the textbook origin, there are numerous reports of LTA variants that originate from the thoracoacromial artery, subscapular artery, and thoracodorsal artery, or even its duplication. This case report involves description of an additional unique duplicate variant of the LTA, bilaterally. The right duplicate LTA has its origin from the 3rd part of the axillary artery and then courses with the axillary sheath before coursing towards the breast. The right duplicate LTA variant also gives off two small subcutaneous branches in the medial upper arm. The left duplicate LTA has its origin from the brachial artery and courses directly to the breast through the axilla. Additional detail on the variants of the LTA could prove useful in surgical procedures that involve the lateral thorax, chest, and axilla and contribute to broader anatomical knowledge.
Collapse
Affiliation(s)
- Mark Rimmer
- Center for Interventional Programs, UCLA Health System, and the UCLA Cardiac Arrhythmia Center & EP Programs, David Geffen School of Medicine, Los Angeles, CA, USA
| | | | - Kalyanam Shivkumar
- Center for Interventional Programs, UCLA Health System, and the UCLA Cardiac Arrhythmia Center & EP Programs, David Geffen School of Medicine, Los Angeles, CA, USA
| | - Shumpei Mori
- Center for Interventional Programs, UCLA Health System, and the UCLA Cardiac Arrhythmia Center & EP Programs, David Geffen School of Medicine, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Chee YJ, Dalan R, Cheung C. The Interplay Between Immunity, Inflammation and Endothelial Dysfunction. Int J Mol Sci 2025; 26:1708. [PMID: 40004172 PMCID: PMC11855323 DOI: 10.3390/ijms26041708] [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: 12/09/2024] [Revised: 02/04/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
The endothelium is pivotal in multiple physiological processes, such as maintaining vascular homeostasis, metabolism, platelet function, and oxidative stress. Emerging evidence in the past decade highlighted the immunomodulatory function of endothelium, serving as a link between innate, adaptive immunity and inflammation. This review examines the regulation of the immune-inflammatory axis by the endothelium, discusses physiological immune functions, and explores pathophysiological processes leading to endothelial dysfunction in various metabolic disturbances, including hyperglycemia, obesity, hypertension, and dyslipidaemia. The final section focuses on the novel, repurposed, and emerging therapeutic targets that address the immune-inflammatory axis in endothelial dysfunction.
Collapse
Affiliation(s)
- Ying Jie Chee
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Rinkoo Dalan
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore 308433, Singapore;
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
| | - Christine Cheung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore;
- Institute of Molecular and Cell Biology, Agency for Science, Technology and Research, Singapore 138632, Singapore
| |
Collapse
|
18
|
Choi K, Cho Y, Chae Y, Cheon SY. Cell-cell communications in the brain of hepatic encephalopathy: The neurovascular unit. Life Sci 2025; 363:123413. [PMID: 39863020 DOI: 10.1016/j.lfs.2025.123413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 01/07/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Many patients with liver diseases are exposed to the risk of hepatic encephalopathy (HE). The incidence of HE in liver patients is high, showing various symptoms ranging from mild symptoms to coma. Liver transplantation is one of the ways to overcome HE. However, not all patients can receive liver transplantation. Moreover, patients who have received liver transplantation have limitations in that they are vulnerable to hepatocellular carcinoma, allograft rejection, and infection. To find other therapeutic strategies, it is important to understand pathological factors and mechanisms that lead to HE after liver disease. Oxidative stress, inflammatory response, hyperammonaemia and metabolic disorders seen after liver diseases have been reported as risk factors of HE. These are known to affect the brain and cause HE. These peripheral pathological factors can impair the blood-brain barrier, cause it to collapse and damage the neurovascular unit component of multiple cells, including vascular endothelial cells, astrocytes, microglia, and neurons, leading to HE. Many previous studies on HE have suggested the impairment of neurovascular unit and cell-cell communication in the pathogenesis of HE. This review focuses on pathological factors that appear in HE, cell type-specific pathological mechanisms, miscommunication/incorrect relationships, and therapeutic candidates between brain cells in HE. This review suggests that regulating communications and interactions between cells may be important in overcoming HE.
Collapse
Affiliation(s)
- Kyuwan Choi
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| | - Yena Cho
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| | - Yerin Chae
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea
| | - So Yeong Cheon
- Department of Biotechnology, College of Biomedical & Health Science, Konkuk University, Chungju, Republic of Korea; Research Institute for Biomedical & Health Science (RIBHS), Konkuk University, Chungju, Republic of Korea.
| |
Collapse
|
19
|
Li XF, Shen GZ, Gong PF, Yang Y, Tuerxun P. Mechanisms of action of the proline hydroxylase-adenosine pathway in regulating apoptosis and reducing myocardial ischemia-reperfusion injury. Clin Hemorheol Microcirc 2025:13860291241310148. [PMID: 39973430 DOI: 10.1177/13860291241310148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Objective: The aim of this study is to explore the protective mechanism of proline hydroxylase (PHD) in reducing myocardial ischemia-reperfusion injury (MIRI) through the hypoxia-inducible factor (HIF)-1α-adenosine-MAPK/ERK signaling pathway, with the goal of identifying potential drug targets and therapeutic strategies for the clinical management of MIRI. Methods: A rat model of MIRI was established using 45 male Sprague-Dawley (SD) rats, which were randomly divided into the following three groups: sham operation (n = 15), MIRI model (n = 15), and MIRI + FG-4592 preconditioning (n = 15) groups. Cardiac function was assessed by echocardiographic measurements of the left ventricular end-diastolic diameter (LVIDd), left ventricular contractile diameter (LVIDs), left ventricular shortening fraction (FS), and left ventricular ejection fraction (EF). Cardiomyocyte apoptosis was evaluated using hematoxylin-eosin (HE) and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining. Myocardial infarct size was determined with 23,5-triphenyltetrazolium chloride (TTC) staining, while levels of inflammatory factors such as interleukin-6 (IL-6), interleukin-1β (IL-1β), and tumor necrosis factor-α (TNF-α) were quantified using enzyme-linked immunosorbent assays (ELISA). Western blot (WB) analysis was performed to assess the expression of apoptotic proteins ERK1/2, phosphorylated-ERK1/2 (p-ERK1/2), AKT, phosphorylated-AKT (p-AKT), caspase-3, BCL-2, and BAX in the infarct boundary area. Adenosine levels within myocardial tissue were also measured. Results: FG-4592 preconditioning significantly improved cardiac function, lowered cardiomyocyte apoptosis and myocardial infarction size, reduced myocardial tissue damage, and inhibited inflammation. Additionally, FG-4592 increased the expression of anti-apoptotic proteins and enhanced adenosine levels in myocardial tissue in the treatment group compared with the MIRI model group. Conclusions: Inhibition of HIF-1α degradation plays a significant role in enhancing extracellular adenosine levels and reducing MIRI, possibly regulating apoptosis through the MAPK/ERK signaling pathway. These findings highlight the potential of targeting the PHD-HIF-adenosine axis in developing treatment strategies for MIRI, meriting future exploration.
Collapse
Affiliation(s)
- Xiu-Fen Li
- Department of Cardiology, Xinjiang Medical University Affiliated Traditional Chinese Medicine Hospital, Urumqi, China
| | - Gu-Zhuo Shen
- Department of Cardiology, The Fourth Clinical Medical College of Xinjiang Medical University, Urumqi, China
| | - Peng-Fei Gong
- Department of Cardiology, Xinjiang Medical University Affiliated Traditional Chinese Medicine Hospital, Urumqi, China
| | - Yan Yang
- Department of Cardiology, Xinjiang Medical University Affiliated Traditional Chinese Medicine Hospital, Urumqi, China
| | - Paerhati Tuerxun
- Department of Cardiology, Xinjiang Medical University Affiliated Traditional Chinese Medicine Hospital, Urumqi, China
| |
Collapse
|
20
|
Sebatana R, Kudzai KD, Magura A, Mdlophane A, Zeevaart JR, Sathekge M, Kahts M, Mdanda S, Witika BA. An Insight to Nanoliposomes as Smart Radiopharmaceutical Delivery Tools for Imaging Atherosclerotic Plaques: Positron Emission Tomography Applications. Pharmaceutics 2025; 17:240. [PMID: 40006607 PMCID: PMC11858949 DOI: 10.3390/pharmaceutics17020240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 01/27/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Atherosclerosis is a chronic progressive disease which is known to cause acute cardiovascular events as well as cerebrovascular events with high mortality. Unlike many other diseases, atherosclerosis is often diagnosed only after an acute or fatal event. At present, the clinical problems of atherosclerosis mainly involve the difficulty in confirming the plaques or identifying the stability of the plaques in the early phase. In recent years, the development of nanotechnology has come with various advantages including non-invasive imaging enhancement, which can be studied for the imaging of atherosclerosis. For targeted imaging and atherosclerosis treatment, nanoliposomes provide enhanced stability, drug administration, extended circulation, and less toxicity. This review discusses the current advances in the development of tailored liposomal nano-radiopharmaceutical-based techniques and their applications to atherosclerotic plaque diagnosis. This review further highlights liposomal nano-radiopharmaceutical localisation and biodistribution-key processes in the pathophysiology of atherosclerosis. Finally, this review discusses the direction and future of liposomal nano-radiopharmaceuticals as a potential clinical tool for the assessment and diagnosis of atherosclerotic plaque.
Collapse
Affiliation(s)
- Reabetswe Sebatana
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
| | - Kahwenga D. Kudzai
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Allan Magura
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Amanda Mdlophane
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Jan Rijn Zeevaart
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
- Radiochemistry, The South African Nuclear Energy Corporation (Necsa) SOC Ltd., Pelindaba 0240, South Africa
| | - Mike Sathekge
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Maryke Kahts
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| | - Sipho Mdanda
- Department of Nuclear Medicine, Steve Biko Academic Hospital, University of Pretoria, Pretoria 0028, South Africa; (A.M.); (M.S.)
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0028, South Africa;
| | - Bwalya Angel Witika
- Department of Pharmaceutical Sciences, School of Pharmacy, Sefako Makgatho Health Sciences University, Pretoria 0208, South Africa; (R.S.); (K.D.K.); (A.M.); (M.K.)
| |
Collapse
|
21
|
Guo W, Yi X. Advancements and future prospects in the study of panvascular disease. Clin Hemorheol Microcirc 2025:13860291241302593. [PMID: 39973436 DOI: 10.1177/13860291241302593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Panvascular disease is characterized by the involvement of blood vessels across multiple regions of the body, and is associated with high morbidity, disability, and mortality rates. Its pathogenesis is multifaceted, necessitating risk assessment and treatment approaches that differ from those applied to single-organ diseases. Given that panvascular disease affects multiple vital organs, an integrated, multi-system management strategy offers significant advantages over conventional, organ-specific approaches. This article provides a comprehensive review of the epidemiological features, traditional and emerging risk factors, pathophysiological mechanisms, screening and risk assessment methods, as well as new strategies for the prevention and management of panvascular disease. The objective is to offer a theoretical foundation and technical support for enhancing prevention and control measures for this condition.
Collapse
Affiliation(s)
- Wei Guo
- Department of Geriatrics, Jining No.1 People's Hospital, Jining, Shandong, China
| | - Xin Yi
- Department of Medical Laboratory, Jining No.1 People's Hospital, Jining, Shandong, China
| |
Collapse
|
22
|
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] [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
|
23
|
Cai B, Kilian D, Ghorbani S, Roth JG, Seymour AJ, Brunel LG, Mejia DR, Rios RJ, Szabo IM, Iranzo SC, Perez A, Rao RR, Shin S, Heilshorn SC. One-step bioprinting of endothelialized, self-supporting arterial and venous networks. Biofabrication 2025; 17:025012. [PMID: 39819775 DOI: 10.1088/1758-5090/adab26] [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: 09/24/2024] [Accepted: 01/16/2025] [Indexed: 01/19/2025]
Abstract
Advances in biofabrication have enabled the generation of freeform perfusable networks mimicking vasculature. However, key challenges remain in the effective endothelialization of these complex, vascular-like networks, including cell uniformity, seeding efficiency, and the ability to pattern multiple cell types. To overcome these challenges, we present an integrated fabrication and endothelialization strategy to directly generate branched, endothelial cell-lined networks using a diffusion-based, embedded 3D bioprinting process. In this strategy, a gelatin microparticle sacrificial ink delivering both cells and crosslinkers is extruded into a crosslinkable gel precursor support bath. A self-supporting, perfusable structure is formed by diffusion-induced crosslinking, after which the sacrificial ink is melted to allow cell release and adhesion to the printed lumen. This approach produces a uniform cell lining throughout networks with complex branching geometries, which are challenging to uniformly and efficiently endothelialize using conventional perfusion-based approaches. Furthermore, the biofabrication process enables high cell viability (>90%) and the formation of a confluent endothelial layer providing vascular-mimetic barrier function and shear stress response. Leveraging this strategy, we demonstrate for the first time the patterning of multiple endothelial cell types, including arterial and venous cells, within a single arterial-venous-like network. Altogether, this strategy enables the fabrication of multi-cellular engineered vasculature with enhanced geometric complexity and phenotypic heterogeneity.
Collapse
Affiliation(s)
- Betty Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - David Kilian
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Sadegh Ghorbani
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Julien G Roth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University, Stanford, CA 94305, United States of America
| | - Alexis J Seymour
- Department of Bioengineering, Stanford University, Stanford, CA 94305, United States of America
| | - Lucia G Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Daniel Ramos Mejia
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Ricardo J Rios
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Isabella M Szabo
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Sean Chryz Iranzo
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Andy Perez
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| | - Rameshwar R Rao
- Ben Towne Center for Childhood Cancer and Blood Disorders Research, Seattle Children's Research Institute, Seattle, WA 98101, United States of America
- Division of Pediatric Hematology, Oncology, Bone Marrow Transplant, and Cellular Therapies, Department of Pediatrics, University of Washington School of Medicine, Seattle, WA 98105, United States of America
| | - Sungchul Shin
- Department of Agriculture, Forestry, and Bioresources, Seoul National University, Seoul 08826, Republic of Korea
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, United States of America
| |
Collapse
|
24
|
Viana-Mattioli S, Fonseca-Alaniz MH, Pinheiro-de-Sousa I, Junior RR, Mastella MH, de Carvalho Cavalli R, Sandrim VC. Plasma from hypertensive pregnancy patients induce endothelial dysfunction even under atheroprotective shear stress. Sci Rep 2025; 15:4675. [PMID: 39920219 PMCID: PMC11805971 DOI: 10.1038/s41598-025-88902-8] [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: 06/29/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
Preeclampsia (PE) is a challenge in maternal healthcare due to its complex nature, characterized by high blood pressure, protein in the urine, and damage to various organs. There is evidence linking PE to endothelial dysfunction (ED), triggered by substances released from an oxygen-deprived placenta. Previous in vitro studies have not considered the impact of in vivo elements, such as the different patterns of blood flow, and laminar (LSS) vs. oscillatory (OSS) shear stress, on the development of ED. We investigated the impact of plasma from healthy pregnant women (HP), subjects with gestational hypertension (GH), and PE patients on global gene expression of human coronary endothelial cells (HCAECs) under LSS and OSS. Our findings revealed a unique transcriptional profile of endothelial cells induced by plasma incubation in LSS. Notably, OSS resulted in similar transcriptomes irrespective of plasma treatment. Under LSS, GH plasma resulted in a proliferative profile, whereas PE plasma was linked to pro-inflammatory and antioxidant profiles compared to HP plasma. Our findings demonstrate that shear stress levels influence the endothelial cell transcriptome in response to plasma from hypertensive pregnancy patients. Both PE and GH can induce endothelial dysfunction under atheroprotective LSS, with a more significant effect observed with PE-derived plasma.
Collapse
Affiliation(s)
- Sarah Viana-Mattioli
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Miriam Helena Fonseca-Alaniz
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Iguaracy Pinheiro-de-Sousa
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
- European Molecular Biology Laboratory, European Bioinformatics Institute, Hinxton, UK
| | - Ricardo Rosa Junior
- Laboratorio de Genetica e Cardiologia Molecular, Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, SP, Brazil
| | - Moises Henrique Mastella
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil
| | - Ricardo de Carvalho Cavalli
- Department of Gynecology and Obstetrics, Hospital das Clínicas, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, SP, Brazil
| | - Valeria Cristina Sandrim
- Department of Biophysics and Pharmacology, Institute of Biosciences of Botucatu, Universidade Estadual Paulista (UNESP), Distrito Rubião Júnior, Botucatu, São Paulo, SP, Brazil.
| |
Collapse
|
25
|
Yao X, Cai X, Zhang S, Yang Y, Yang X, Ma W, Jiang Z. Mendelian randomization study of serum uric acid levels and urate-lowering drugs on pulmonary arterial hypertension outcomes. Sci Rep 2025; 15:4460. [PMID: 39915571 PMCID: PMC11802783 DOI: 10.1038/s41598-025-88887-4] [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: 10/11/2024] [Accepted: 01/31/2025] [Indexed: 02/09/2025] Open
Abstract
This study aims to explore the causal relationships between serum uric acid level and pulmonary arterial hypertension (PAH) using the Mendelian randomization (MR) approach, and to assess the therapeutic impacts of urate-lowering drugs on PAH. Utilizing published genome-wide association study (GWAS) data, we applied MR and colocalization analysis to assess the link between serum uric acid levesl and PAH across four GWAS datasets from two distinct European populations. The validity and reliability of these findings were confirmed through multiple statistical methods, along with an MR analysis of urate-lowering drug targets to investigate their potential effects on PAH treatment. MR analysis revealed a significant positive correlation between serum uric acid levels and PAH (odds ratio (OR) 1.106, 95% confidence intervals (CI) 1.021-1.200, P = 0.014), corroborated by a replication MR analysis (OR 1.859, 95% CI 1.130-3.057, P = 0.015). No significant heterogeneity or horizontal pleiotropy was found in the sensitivity analyses. However, urate-lowering drugs did not demonstrate a significant direct therapeutic effect on PAH. This study establishes a genetic basis for a causal link between serum uric acid levels and PAH. However, urate-lowering drugs do not appear to have a direct causal effect on improving PAH. These findings provide a novel reference point for developing future therapeutic strategies for PAH.
Collapse
Affiliation(s)
- Xiaoling Yao
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Xin Cai
- Department of Rheumatology and Immunology, The First People's Hospital of Guiyang, Guiyang, 550001, China
| | - Shaoqin Zhang
- Department of Rheumatology and Immunology, The First People's Hospital of Guiyang, Guiyang, 550001, China
| | - Yuzheng Yang
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China
| | - Xiangyan Yang
- Department of Rheumatology and Immunology, The First People's Hospital of Guiyang, Guiyang, 550001, China
| | - Wukai Ma
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
- Institute of the Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China.
| | - Zong Jiang
- Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, 550001, China.
- Institute of the Second Clinical Medical College, Guizhou University of Traditional Chinese Medicine, Guiyang, Guizhou, China.
| |
Collapse
|
26
|
Ławkowska K, Bonowicz K, Jerka D, Bai Y, Gagat M. Integrins in Cardiovascular Health and Disease: Molecular Mechanisms and Therapeutic Opportunities. Biomolecules 2025; 15:233. [PMID: 40001536 PMCID: PMC11853560 DOI: 10.3390/biom15020233] [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: 12/18/2024] [Revised: 01/30/2025] [Accepted: 02/01/2025] [Indexed: 02/27/2025] Open
Abstract
Cardiovascular diseases, including atherosclerosis, hypertension, and heart failure, remain the leading cause of global mortality, with endothelial dysfunction and vascular remodeling as critical contributors. Integrins, as transmembrane adhesion proteins, are central regulators of cell adhesion, migration, and signaling, playing a pivotal role in maintaining vascular homeostasis and mediating pathological processes such as inflammation, angiogenesis, and extracellular matrix remodeling. This article comprehensively examines the role of integrins in the pathogenesis of cardiovascular diseases, focusing on their dysfunction in endothelial cells and interactions with inflammatory mediators, such as TNF-α. Molecular mechanisms of integrin action are discussed, including their involvement in mechanotransduction, leukocyte adhesion, and signaling pathways that regulate vascular integrity. The review also highlights experimental findings, such as the use of specific integrin-targeting plasmids and immunofluorescence to elucidate integrin functions under inflammatory conditions. Additionally, potential therapeutic strategies are explored, including the development of integrin inhibitors, monoclonal antibodies, and their application in regenerative medicine. These approaches aim not only to mitigate pathological vascular remodeling but also to promote tissue repair and angiogenesis. By bridging insights from molecular studies with their translational potential, this work underscores the promise of integrin-based therapies in advancing the management and treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- Karolina Ławkowska
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Klaudia Bonowicz
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
- Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland
| | - Dominika Jerka
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
| | - Yidong Bai
- Department of Cell Systems and Anatomy, University of Texas Health San Antonio, San Antonio, TX 78229, USA;
| | - Maciej Gagat
- Department of Histology and Embryology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, 85-092 Bydgoszcz, Poland
- Collegium Medicum, Mazovian Academy in Płock, 09-402 Płock, Poland
| |
Collapse
|
27
|
Haritz JL, Pflaum M, Güntner HJ, Katsirntaki K, Hegermann J, Hehnen F, Lommel M, Kertzscher U, Arens J, Haverich A, Ruhparwar A, Wiegmann B. Citrate-Coated Iron Oxide Nanoparticles Facilitate Endothelialization of Left Ventricular Assist Device Impeller for Improved Antithrombogenicity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408976. [PMID: 39707689 PMCID: PMC11809402 DOI: 10.1002/advs.202408976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/14/2024] [Indexed: 12/23/2024]
Abstract
Although left ventricular assist devices (LVADs) are an alternative to heart transplantation, their artificial surfaces often lead to serious thrombotic complications requiring high-risk device replacement. Coating blood-contacting surfaces with antithrombogenic endothelial cells is considered an effective strategy for preventing thrombus formation. However, this concept has not yet been successfully implemented in LVADs, as severe cell loss is to be expected, especially on the impeller surface with high prothrombogenic supraphysiological shear stress. This study presents a strategy that exploits the magnetic attraction of the impeller on ECs loaded with iron oxide nanoparticles (IONPs) to minimize shear stress-induced cell detachment from the rotating magnetic impeller while ensuring antithrombogenic EC adhesion, especially as a bridge until they formed their adhesion-promoting matrix. In contrast to polyvinylpyrrolidone (PVP)-coated IONPs, more efficient and safer cell loading is achieved with sodium citrate (Cit)-stabilized IONPs, where incubation with 6.6 µg iron mL-1 Cit-IONPs for 24 h resulting in an average internalization of 23 pg iron per cell. Internalization of Cit-IONP significantly improved cell attraction to the highly magnetic impeller surface without affecting cell viability or antithrombogenic function. This protocol is key for the development of a biohybrid LVAD impeller that can prevent life-threatening thrombosis and hemorrhage in a future clinical application.
Collapse
Affiliation(s)
- Jasper L. Haritz
- Department of CardiothoracicTransplantation and Vascular SurgeryHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Lower Saxony Center for Biomedical EngineeringImplant Research and DevelopmentStadtfelddamm 3430625HannoverGermany
| | - Michael Pflaum
- Department of CardiothoracicTransplantation and Vascular SurgeryHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Lower Saxony Center for Biomedical EngineeringImplant Research and DevelopmentStadtfelddamm 3430625HannoverGermany
- German Center for Lung ResearchBREATHHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Hans J. Güntner
- Department of CardiothoracicTransplantation and Vascular SurgeryHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Lower Saxony Center for Biomedical EngineeringImplant Research and DevelopmentStadtfelddamm 3430625HannoverGermany
| | - Katherina Katsirntaki
- Department of CardiothoracicTransplantation and Vascular SurgeryHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Lower Saxony Center for Biomedical EngineeringImplant Research and DevelopmentStadtfelddamm 3430625HannoverGermany
| | - Jan Hegermann
- German Center for Lung ResearchBREATHHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Research Core Unit Electron Microscopy and Institute of Functional and Applied AnatomyHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Felix Hehnen
- Biofluid Mechanics LaboratoryInstitute of Computer‐assisted Cardiovascular MedicineCharité – Universitätsmedizin Berlin13353BerlinGermany
- Charité –Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinCharitéplatz 110117BerlinGermany
| | - Michael Lommel
- Biofluid Mechanics LaboratoryInstitute of Computer‐assisted Cardiovascular MedicineCharité – Universitätsmedizin Berlin13353BerlinGermany
- Charité –Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinCharitéplatz 110117BerlinGermany
| | - Ulrich Kertzscher
- Biofluid Mechanics LaboratoryInstitute of Computer‐assisted Cardiovascular MedicineCharité – Universitätsmedizin Berlin13353BerlinGermany
- Charité –Universitätsmedizin Berlincorporate member of Freie Universität Berlin and Humboldt‐Universität zu BerlinCharitéplatz 110117BerlinGermany
| | - Jutta Arens
- Engineering Organ Support Technologies GroupDepartment of Biomechanical EngineeringFaculty of Engineering TechnologyUniversity of TwenteEnschedeNB7522Netherlands
- Member of the DFG‐SPP201430625HannoverGermany
| | - Axel Haverich
- Department of CardiothoracicTransplantation and Vascular SurgeryHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Lower Saxony Center for Biomedical EngineeringImplant Research and DevelopmentStadtfelddamm 3430625HannoverGermany
- German Center for Lung ResearchBREATHHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Arjang Ruhparwar
- Department of CardiothoracicTransplantation and Vascular SurgeryHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Lower Saxony Center for Biomedical EngineeringImplant Research and DevelopmentStadtfelddamm 3430625HannoverGermany
- German Center for Lung ResearchBREATHHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
| | - Bettina Wiegmann
- Department of CardiothoracicTransplantation and Vascular SurgeryHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Lower Saxony Center for Biomedical EngineeringImplant Research and DevelopmentStadtfelddamm 3430625HannoverGermany
- German Center for Lung ResearchBREATHHannover Medical SchoolCarl‐Neuberg‐Str. 130625HannoverGermany
- Member of the DFG‐SPP201430625HannoverGermany
| |
Collapse
|
28
|
Li K, Zhang C, Wang LX, Wang X, Wang R. KLF4's role in regulating nitric oxide production and promoting microvascular formation following ischemic stroke. Nitric Oxide 2025; 154:86-104. [PMID: 39557151 DOI: 10.1016/j.niox.2024.11.004] [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: 08/22/2024] [Revised: 11/15/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
This study examines KLF4's role in endothelial cells (ECs), emphasizing its effects on nitric oxide (NO) production, microvascular formation, and oxidative stress regulation following ischemic stroke. Through high-throughput sequencing, we identified eight cell subpopulations in carotid artery tissues post-stroke, with KLF4 notably elevated in ECs. KLF4 overexpression in ECs promoted NO synthesis, enhanced endothelial tube formation, mitigated oxidative stress, and improved smooth muscle cells (SMCs) function, collectively boosting blood flow in ischemic regions. These findings highlight KLF4 as pivotal in vascular regeneration and oxidative stress reduction, positioning it as a promising target for cardiovascular and cerebrovascular therapies.
Collapse
Affiliation(s)
- Kuo Li
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China.
| | - Chuansuo Zhang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Li Xuan Wang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Xiaoxuan Wang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| | - Ruyue Wang
- No. 2, Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061000, China
| |
Collapse
|
29
|
Zhu Y, Verkhratsky A, Chen H, Yi C. Understanding glucose metabolism and insulin action at the blood-brain barrier: Implications for brain health and neurodegenerative diseases. Acta Physiol (Oxf) 2025; 241:e14283. [PMID: 39822067 PMCID: PMC11737474 DOI: 10.1111/apha.14283] [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: 11/11/2024] [Revised: 12/09/2024] [Accepted: 01/01/2025] [Indexed: 01/19/2025]
Abstract
The blood-brain barrier (BBB) is a highly selective, semipermeable barrier critical for maintaining brain homeostasis. The BBB regulates the transport of essential nutrients, hormones, and signaling molecules between the bloodstream and the central nervous system (CNS), while simultaneously protecting the brain from potentially harmful substances and pathogens. This selective permeability ensures that the brain is nourished and shielded from toxins. An exception to this are brain regions, such as the hypothalamus and circumventricular organs, which are irrigated by fenestrated capillaries, allowing rapid and direct response to various blood components. We overview the metabolic functions of the BBB, with an emphasis on the impact of altered glucose metabolism and insulin signaling on BBB in the pathogenesis of neurodegenerative diseases. Notably, endothelial cells constituting the BBB exhibit distinct metabolic characteristics, primarily generating ATP through aerobic glycolysis. This occurs despite their direct exposure to the abundant oxygen in the bloodstream, which typically supports oxidative phosphorylation. The effects of insulin on astrocytes, which form the glial limitans component of the BBB, show a marked sexual dimorphism. BBB nutrient sensing in the hypothalamus, along with insulin signaling, regulates systemic metabolism. Insulin modifies BBB permeability by regulating the expression of tight junction proteins, angiogenesis, and vascular remodeling, as well as modulating blood flow in the brain. The disruptions in glucose and insulin signaling are particularly evident in neurodegenerative diseases, such as Alzheimer's disease and Parkinson's disease, where BBB breakdown accelerates cognitive decline. This review highlights the critical role of normal glucose metabolism and insulin signaling in maintaining BBB functionality and investigates how disruptions in these pathways contribute to the onset and progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yiyi Zhu
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
| | - Alexei Verkhratsky
- Faculty of Biology, Medicine and HealthThe University of ManchesterManchesterUK
- Department of NeurosciencesUniversity of the Basque Country, CIBERNEDLeioaBizkaiaSpain
- IKERBASQUE Basque Foundation for ScienceBilbaoSpain
- Department of Forensic Analytical Toxicology, School of Forensic MedicineChina Medical UniversityShenyangChina
| | - Hui Chen
- School of Life Sciences, Faculty of ScienceUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Chenju Yi
- Research CenterThe Seventh Affiliated Hospital of Sun Yat‐Sen UniversityShenzhenChina
- Guangdong Provincial Key Laboratory of Brain Function and DiseaseGuangzhouChina
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational ResearchShenzhenChina
| |
Collapse
|
30
|
Kohutek ZA, Caslin HL, Fehrenbach DJ, Heimlich JB, Brown JD, Madhur MS, Ferrell PB, Doran AC. Bone Marrow Niche in Cardiometabolic Disease: Mechanisms and Therapeutic Potential. Circ Res 2025; 136:325-353. [PMID: 39883790 PMCID: PMC11790260 DOI: 10.1161/circresaha.124.323778] [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] [Indexed: 02/01/2025]
Abstract
Cardiovascular and cardiometabolic diseases are leading causes of morbidity and mortality worldwide, driven in part by chronic inflammation. Emerging research suggests that the bone marrow microenvironment, or marrow niche, plays a critical role in both immune system regulation and disease progression. The bone marrow niche is essential for maintaining hematopoietic stem cells (HSCs) and orchestrating hematopoiesis. Under normal conditions, this niche ensures a return to immune homeostasis after acute stress. However, in the setting of inflammatory conditions such as those seen in cardiometabolic diseases, it becomes dysregulated, leading to enhanced myelopoiesis and immune activation. This review explores the reciprocal relationship between the bone marrow niche and cardiometabolic diseases, highlighting how alterations in the niche contribute to disease development and progression. The niche regulates HSCs through complex interactions with stromal cells, endothelial cells, and signaling molecules. However, in the setting of chronic diseases such as hypertension, atherosclerosis, and diabetes, inflammatory signals disrupt the balance between HSC self-renewal and differentiation, promoting the excessive production of proinflammatory myeloid cells that exacerbate the disease. Key mechanisms discussed include the effects of hyperlipidemia, hyperglycemia, and sympathetic nervous system activation on HSC proliferation and differentiation. Furthermore, the review emphasizes the role of epigenetic modifications and metabolic reprogramming in creating trained immunity, a phenomenon whereby HSCs acquire long-term proinflammatory characteristics that sustain disease states. Finally, we explore therapeutic strategies aimed at targeting the bone marrow niche to mitigate chronic inflammation and its sequelae. Novel interventions that modulate hematopoiesis and restore niche homeostasis hold promise for the treatment of cardiometabolic diseases. By interrupting the vicious cycle of inflammation and marrow dysregulation, such therapies may offer new avenues for reducing cardiovascular risk and improving patient outcomes.
Collapse
Affiliation(s)
- Zachary A Kohutek
- Department of Radiation Oncology (Z.A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Heather L Caslin
- Department of Health and Human Performance, University of Houston, TX (H.L.C.)
| | - Daniel J Fehrenbach
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis (D.J.F., M.S.M.)
| | - J Brett Heimlich
- Division of Cardiovascular Medicine, Department of Medicine (J.B.H., J.D.B., A.C.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Jonathan D Brown
- Division of Cardiovascular Medicine, Department of Medicine (J.B.H., J.D.B., A.C.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Meena S Madhur
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis (D.J.F., M.S.M.)
| | - P Brent Ferrell
- Division of Hematology and Oncology, Department of Medicine (P.B.F.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN (P.B.F., A.C.D.)
| | - Amanda C Doran
- Division of Cardiovascular Medicine, Department of Medicine (J.B.H., J.D.B., A.C.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN (P.B.F., A.C.D.)
| |
Collapse
|
31
|
Zhu Y, Wei J, Yang X, Zhu W. Molecular mechanism underlying cardioprotective effect of dehydroepiandrosterone on endoplasmic reticulum stress induced apoptosis in human vascular smooth muscle cells and human umbilical vein endothelial cells. Front Pharmacol 2025; 16:1496393. [PMID: 39936092 PMCID: PMC11810946 DOI: 10.3389/fphar.2025.1496393] [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: 09/14/2024] [Accepted: 01/10/2025] [Indexed: 02/13/2025] Open
Abstract
Introduction This study aimed to investigate the underlying mechanisms involved in the cardioprotective effects of dehydroepiandrosterone (DHEA) on endoplasmic reticulum stress (ERS) -mediated apoptosis in human vascular smooth muscle cells (HVSMCs) and human umbilical vein endothelial cells (HUVECs). Material and methods Various concentrations of Dithiothreitol (DTT) were used to induce ERS-mediated apoptosis. DHEA was utilized to inhibit the apoptotic effects of DTT, while estrogen receptor (ER) antagonists ICI 182,780 and G15, the androgen receptor (AR) antagonist flutamide and the aromatase inhibitor letrozole were used to identify the receptors activated during DHEA treatment in HVSMCs and HUVECs. Flow cytometry assessed the apoptotic rate, and Western blotting analysis evaluated the expression levels of ERS-related proteins GRP78 and PERK, and the apoptotic protein marker CHOP. Furthermore, the primary receptor signaling pathways were identified using signaling pathway blockers: LY294002 (PI3K blocker), SP600125 (JNK blocker), and U0126 (ERK1/2 blocker). Results In the DTT pretreatment group (0.8 mmol/L, for 8 h), the expressions of GRP78 and CHOP were significantly up regulated, indicating that an optimal ERS model was successfully established. Additionally, 10-4 mmol/L DHEA significantly inhibited the DTT-induced upregulation of GRP78 and CHOP. The results also demonstrated that the apoptotic rate in the DTT group was increased, while DHEA significantly reduced this rate. The addition of ER antagonists ICI 182,780 and G15 to HVSMCs reversed the effects of DHEA; however, the aromatase inhibitor letrozole and the AR antagonist flutamide did not reverse this effect. Notably, the use of the JNK inhibitor SP600125, the PI3K inhibitor LY294002, and the ERK1/2 inhibitor U0126 antagonized the protective effects of DHEA, with SP600125 showing the most significant impact on both HVSMCs and HUVECs. Conclusion Our study has identified a novel mechanism underlying the cardioprotective effects of DHEA. Specifically, DHEA may mitigate ERS-induced cell apoptosis by activating estrogen receptors ERα, ERβ, and GPER via the activated JNK pathway.
Collapse
Affiliation(s)
- Ye Zhu
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Junxiu Wei
- Department of Reproductive Medicine, Affiliated Hospital of Hebei University, Baoding, China
| | - Xin Yang
- Department of Obstetrics and Gynecology, Peking University People’s Hospital, Beijing, China
| | - Wei Zhu
- Department of Immunology, Mudanjiang Medical University, Mudanjiang, China
| |
Collapse
|
32
|
Ge R, Lin J, Feng X, Huang C, Huang J, Li C, Wen Z, Xu A, Huang M, Yuan H, Shi H, Ma G, Yi R, Liang S, Bi Y, Su S, Zhang X, Li X, Duan C. Analysis of the effect of platelet function and different doses of ticagrelor after flow diverter treatment of intracranial aneurysms. Neurosurg Rev 2025; 48:90. [PMID: 39870937 DOI: 10.1007/s10143-025-03225-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/07/2025] [Accepted: 01/14/2025] [Indexed: 01/29/2025]
Abstract
Ticagrelor has become the standard drug for the treatment of intracranial aneurysms (IAs) with flow diverters (FDs), but the dosage has not been standardized. The effect of platelet function on clinical and imaging prognosis remains unclear. This study aimed to show the effects of different doses of ticagrelor and platelet aggregation function on the clinical and imaging prognosis after FDs treatment of aneurysms. Patients with IAs and underwent FDs stenting were recruited between July 2019 and June 2023. Logistic regression was performed to assess the predictors of incomplete occlusion and in-stent stenosis (ISS). Linear regression analysis, scatter plot and violin diagram were used to investigate the predictors of maximum platelet aggregation rate induced by ADP (ADP-MPA). The study included 156 patients with 206 aneurysms. There was no significant difference in clinical prognosis and aneurysm occlusion rates between the standard-dose group (ticagrelor, 90 mg/bid) and the low-dose group (ticagrelor, 45 mg/bid). Multivariable analysis identified the ADP-MPA ≥ ADP-MPA (median) (24.2%) (p = 0.037) as an independent risk factor for incomplete occlusion of aneurysms. In addition, higher ADP-MPA (p = 0.045) was an independent risk factor for ISS. Uric acid level (p = 0.019) was negatively associated with ADP-MPA, whereas age (p = 0.031) and body mass index (BMI) (p = 0.042) were positively associated with ADP-MPA. The differences of clinical prognosis and aneurysm occlusion rates between the standard-dose group and the low-dose group were not significant for the treatment of aneurysms with FDs. Higher ADP-MPA predicted higher rates of incomplete occlusion and in-stent stenosis. Uric acid level was negatively associated with ADP-MPA, whereas age and BMI were positively associated with ADP-MPA.
Collapse
Affiliation(s)
- Runze Ge
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiancheng Lin
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Feng
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Chi Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jiwan Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Can Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zhuohua Wen
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Anqi Xu
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Mengshi Huang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hao Yuan
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongyu Shi
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Gengwu Ma
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ruizhe Yi
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shuyin Liang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yiming Bi
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Shixing Su
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xin Zhang
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Xifeng Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chuanzhi Duan
- Neurosurgery Center, Department of Cerebrovascular Surgery, Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
33
|
Haferanke J, Baumgartner L, Willinger L, Oberhoffer-Fritz R, Schulz T. Molecular Mechanisms of Vascular Tone in Exercising Pediatric Populations: A Comprehensive Overview on Endothelial, Antioxidative, Metabolic and Lipoprotein Signaling Molecules. Int J Mol Sci 2025; 26:1027. [PMID: 39940797 PMCID: PMC11817131 DOI: 10.3390/ijms26031027] [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: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/16/2025] Open
Abstract
Vasoactive molecules are central regulators of vascular tone, angiogenesis and inflammation. Key molecular agents include nitric oxide (NO), endothelin-1 (ET-1), prostacyclin, free triiodothyronine (fT3), leptin, low-density lipoprotein (LDL), high-density lipoprotein (HDL), superoxide dismutase (SOD), and glutathione peroxidase (GPX). Dysregulation of these compounds can lead to endothelial dysfunction, an early predictor of atherosclerosis and cardiovascular diseases (CVD). Maintaining endothelial health is thus essential for vascular homeostasis and cardiovascular risk prevention. Regular exercise serves as a vital protective measure against CVD and the risk of cardiovascular conditions. However, young athletes often significantly exceed recommended levels of training load, engaging in highly intensive training that leads to substantial physiological adaptations. Despite this, research on the impact of exercise on vasoactive substances in children and adolescents, particularly young athletes, is limited and inconsistent. Most studies focus on those with pre-existing conditions, like obesity or diabetes mellitus. Existing findings suggest exercise may favorably affect vascular biomarkers in youth, but methodological variations hinder consistent conclusions. This literature review examines 68 studies on the effects of exercise on vascular molecules in children and adolescents, young athletes, and children and adolescents with pre-existing conditions, offering deeper insights into how exercise may influence vascular health at the molecular level.
Collapse
Affiliation(s)
- Jonas Haferanke
- Department Health and Sport Sciences, Institute of Preventive Pediatrics, TUM School of Medicine and Health, Technical University of Munich (TUM), 80992 Munich, Germany
| | | | | | | | - Thorsten Schulz
- Department Health and Sport Sciences, Institute of Preventive Pediatrics, TUM School of Medicine and Health, Technical University of Munich (TUM), 80992 Munich, Germany
| |
Collapse
|
34
|
Liu S, Yan J, Gao M, Yang H. Research progress in the regulation of endothelial cells and smooth muscle cells using a micro-nanostructure. Biomed Eng Online 2025; 24:6. [PMID: 39849451 PMCID: PMC11760742 DOI: 10.1186/s12938-025-01337-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 01/25/2025] Open
Abstract
Recently, the incidence rate and mortality of various acute or chronic vascular occlusive diseases have increased yearly. As one of the most effective measures to treat them, vascular stents have been widely studied by researchers, and presently, the most commonly used is a drug-eluting stent, which reduces the process of rapid endothelialization because the drug is not selective. Fortunately, with the discovery and exploration of micro-nanostructures that can regulate cells selectively, reducing the incidence of "intravascular restenosis" and achieving rapid endothelialization simultaneously are possible through a special structure that cannot only improve endothelial cells (ECs), but also inhibit smooth muscle cells (SMCs). Therefore, this paper mainly introduces the preparation methods of micro-nanostructures used in the past, as well as the detection methods of EC and SMC. Then, the various functions of different dimensional structures for different cells are summarized and analyzed. Finally, the application of micro-nanostructure in future stent materials is summarized and proposed.
Collapse
Affiliation(s)
- Songhao Liu
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, China
- School of Energy and Electrical Engineering, Qinghai University, Xining, 810016, Qinghai, China
| | - Juan Yan
- Department of Pharmacy, Medical College of Qinghai University, Xining, 810016, China
| | - Mengyu Gao
- School of Energy and Electrical Engineering, Qinghai University, Xining, 810016, Qinghai, China
| | - Hongxia Yang
- Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, China.
| |
Collapse
|
35
|
Donadini MP, Calcaterra F, Romualdi E, Ciceri R, Cancellara A, Lodigiani C, Bacci M, Della Bella S, Ageno W, Mavilio D. The Link Between Venous and Arterial Thrombosis: Is There a Role for Endothelial Dysfunction? Cells 2025; 14:144. [PMID: 39851572 PMCID: PMC11763525 DOI: 10.3390/cells14020144] [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: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 01/26/2025] Open
Abstract
Venous thromboembolism (VTE) and arterial thrombosis (AT) are distinct yet closely related pathological processes. While traditionally considered separate entities, accumulating evidence suggests that they share common risk factors, such as inflammation and endothelial dysfunction (ED). This review explores the parallels and differences between venous and arterial thrombosis, with particular attention to the role of unprovoked VTE and its potential links to atherosclerosis and systemic inflammation. A key focus is the role of ED, which is emerging as a critical factor in thrombogenesis across both the venous and arterial systems. We examine the current methods for clinically detecting ED, including the use of biomarkers and advanced imaging techniques. Additionally, we discuss novel research avenues, such as the potential of endothelial colony-forming cells and other innovative methodologies, to further unravel the complex mechanisms of thrombosis. Finally, we propose future clinical scenarios where targeting endothelial health could pave the way for more effective prevention and treatment strategies in thrombosis management.
Collapse
Affiliation(s)
- Marco Paolo Donadini
- Department of Medicine and Surgery, Research Center on Thromboembolic Diseases and Antithrombotic Therapies, University of Insubria, 21100 Varese, Italy;
- Centro Trombosi e TAO, Azienda Socio Sanitaria Territoriale dei Sette Laghi, 21100 Varese, Italy;
| | - Francesca Calcaterra
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Erica Romualdi
- Centro Trombosi e TAO, Azienda Socio Sanitaria Territoriale dei Sette Laghi, 21100 Varese, Italy;
| | - Roberta Ciceri
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Assunta Cancellara
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Corrado Lodigiani
- Center for Thrombosis and Hemorrhagic Diseases, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.L.); (M.B.)
| | - Monica Bacci
- Center for Thrombosis and Hemorrhagic Diseases, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy; (C.L.); (M.B.)
| | - Silvia Della Bella
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| | - Walter Ageno
- Department of Medicine and Surgery, Research Center on Thromboembolic Diseases and Antithrombotic Therapies, University of Insubria, 21100 Varese, Italy;
- Department of Internal Medicine, Ospedale Regionale di Bellinzona e Valli, 6500 Bellinzona, Switzerland
| | - Domenico Mavilio
- Department of Medical Biotechnologies and Translational Medicine, University of Milan, 20125 Milan, Italy; (F.C.); (R.C.); (A.C.); (S.D.B.); (D.M.)
- Unit of Clinical and Experimental Immunology, IRCCS Humanitas Research Hospital, 20089 Rozzano, Italy
| |
Collapse
|
36
|
Camarda ND, Lu Q, Tesfu AF, Liu RR, Ibarrola J, Jaffe IZ. Mineralocorticoid Receptor in Endothelial Cells Contributes to Vascular Endothelial Growth Factor Receptor Inhibitor-Induced Vascular and Kidney Damage. Am J Hypertens 2025; 38:104-110. [PMID: 39514632 PMCID: PMC11735467 DOI: 10.1093/ajh/hpae140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 08/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Vascular endothelial growth factor receptor inhibitors (VEGFRis) improve cancer patient survival by inhibiting tumor angiogenesis. However, VEGFRis induce treatment-limiting hypertension which has been associated with impaired vascular endothelial cell (EC) function and kidney damage. The mineralocorticoid receptor (MR) regulates blood pressure (BP) via its effects on the vasculature and the kidney. Thus, we interrogated the role of the MR in EC dysfunction, renal impairment, and hypertension in a mouse model of VEGFRi-induced hypertension using sorafenib. METHODS EC dysfunction in mesenteric arterioles was assessed by immunoblotting for phosphorylation of endothelial nitric oxide synthase (eNOS) at serine 1177. Renal damage was measured by assessing glomerular endotheliosis histologically. BP was measured using implanted radiotelemetry. RESULTS Six days of sorafenib treatment significantly impaired mesenteric resistance vessel EC function, induced renal damage, and increased BP. Pharmacologic MR blockade with spironolactone prevented the sorafenib-induced decline in eNOS phosphorylation and renal glomerular endotheliosis, without affecting systolic BP (SBP) or diastolic BP. Mice with the MR knocked out specifically in ECs (EC-MR-KO) were protected from sorafenib-induced EC dysfunction and glomerular endotheliosis, whereas smooth muscle cell-specific MR (SMC-MR) knockout mice were not. Neither EC-MR nor SMC-MR knockout affected the degree to which sorafenib increased SBP or diastolic BP. CONCLUSIONS These results reveal that the MR, specifically in EC but not in SMCs, is necessary for VEGFRi-induced renal and vascular injury. While ineffective at lowering SBP, these data suggest potential therapeutic benefits of MR antagonists, like spironolactone, to protect the vasculature and the kidneys from VEGFRi-induced injury.
Collapse
Affiliation(s)
- Nicholas D Camarda
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
- Genetics, Molecular, and Cellular Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| | - Qing Lu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Angelina F Tesfu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Rui R Liu
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jaime Ibarrola
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
| | - Iris Z Jaffe
- Molecular Cardiology Research Institute, Tufts Medical Center, Boston, Massachusetts, USA
- Genetics, Molecular, and Cellular Biology Program, Tufts Graduate School of Biomedical Sciences, Boston, Massachusetts, USA
| |
Collapse
|
37
|
Mao M, Wu Y, He Q. Breaking Through Physiological Barriers: Nanorobotic Strategies for Active Drug Delivery. Bioconjug Chem 2025; 36:1-14. [PMID: 39729406 DOI: 10.1021/acs.bioconjchem.4c00480] [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: 12/29/2024]
Abstract
Self-propelled micro/nanomotors (MNMs) represent a groundbreaking advancement in precision drug delivery, offering potential solutions to persistent challenges such as systemic toxicity, limited bioavailability, and nonspecific distribution. By transforming various energy sources into mechanical motion, MNMs are able to autonomously navigate through complex physiological environments, facilitating targeted delivery of therapeutic agents to previously inaccessible regions. However, to achieve efficient in vivo drug delivery, biomedical MNMs must demonstrate their ability to overcome crucial physiological barriers encompassing mucosal surfaces, blood flow dynamics, vascular endothelium, and cellular membrane. This review provides a comprehensive overview of the latest strategies developed to address these obstacles while also analyzing the broader challenges and opportunities associated with clinical translation. Our objective is to establish a solid foundation for future research in medical MNMs by focusing on enhancing drug delivery efficiency and advancing precision medicine, ultimately paving the way for practical theragnostic applications and wider clinical adoption.
Collapse
Affiliation(s)
- Meng Mao
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China
| | - Yingjie Wu
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China
| | - Qiang He
- School of Medicine and Health, Harbin Institute of Technology, Harbin 150080, China
| |
Collapse
|
38
|
Xie M, Li X, Chen L, Zhang Y, Chen L, Hua H, Qi J. The crosstalks between vascular endothelial cells, vascular smooth muscle cells, and adventitial fibroblasts in vascular remodeling. Life Sci 2025; 361:123319. [PMID: 39701178 DOI: 10.1016/j.lfs.2024.123319] [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: 09/22/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 12/21/2024]
Abstract
Pathological vascular remodeling (VR) is characterized by structural and functional alterations in the vascular wall resulting from injury, which significantly contribute to the development of cardiovascular diseases (CVDs). The vascular wall consists primarily of endothelial cells (ECs), vascular smooth muscle cells (VSMCs), and adventitial fibroblasts (AFs), whose interactions are crucial for both the formation of the vascular system and the maintenance of mature blood vessels. Disruptions in the communication between these cell types have been implicated in the progression of VR. This review examines the complex interactions between ECs, VSMCs, and AFs in the context of CVD development, emphasizing a relatively underexplored yet potentially critical mechanism. This interaction framework likely extends to the broader cellular dialogue in the pathogenesis of CVDs, suggesting novel therapeutic strategies for intervention.
Collapse
Affiliation(s)
- Ming Xie
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; Department of Pharmacy, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214499, China
| | - Xiandeng Li
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China; College of Pharmacy, Chongqing Medical University, Chongqing 400016, China
| | - Lun Chen
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Yufeng Zhang
- Department of Vascular Surgery, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, Shangdong 271000, China; Postdoctoral Workstation, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shangdong 250117, China; Department of Pulmonary and Critical Care Medicine, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214499, China
| | - Long Chen
- Institute of Chinese Medicine of Taizhou China Medical City, Taizhou, Jiangsu 225316, China; International Centre for Genetic Engineering and Biotechnology, Taizhou, Jiangsu 225300, China
| | - Haibing Hua
- Department of Gastroenterology, Jiangyin Hospital of Traditional Chinese Medicine, Jiangyin Hospital Affiliated to Nanjing University of Chinese Medicine, Jiangyin, Jiangsu 214499, China.
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai 200092, China.
| |
Collapse
|
39
|
Flores J, Nugent K. Sodium, the Vascular Endothelium, and Hypertension: A Narrative Review of Literature. Cardiol Rev 2025:00045415-990000000-00402. [PMID: 39807866 DOI: 10.1097/crd.0000000000000854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The vascular endothelium and its endothelial glycocalyx contribute to the protection of the endothelial cells from exposure to high levels of sodium and help these structures maintain normal function by regulating vascular permeability due to its buffering effect. The endothelial glycocalyx has negative surface charges that bind sodium and limit sodium entry into cells and the interstitial space. High sodium levels can disrupt this barrier and allow the movement of sodium into cells and extravascular fluid. This can generate reactive oxygen species that inhibit nitric oxide production. This leads to vasospasm and increases intravascular pressures. Overtime vascular remodeling occurs, and this changes the anatomy of blood vessels, their intrinsic stiffness, and their response to vasodilators and results in hypertension. Patients with increased salt sensitivity are potentially at more risk for this sequence of events. Studies on the degradation of the glycocalyx provide insight into the pathogenesis of clinical disorders with vascular involvement, but there is limited information available in the context of higher concentrations of sodium. Data on higher intake of sodium and the imbalance between nitric oxide and reactive oxygen species have been obtained in experimental studies and provide insights into possible outcomes in humans. The current western diet with sodium intake above recommended levels has led to the assessment of sodium sensitivity, which has been used in different populations and could become a practical tool to evaluate patients. This would potentially allow more focused recommendations regarding salt intake. This review will consider the structure of the vascular endothelium, its components, the effect of sodium on it, and the use of the salt blood test mini.
Collapse
Affiliation(s)
- Jackeline Flores
- From the Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX
| | | |
Collapse
|
40
|
Rajala R, Cleuren ACA, Griffin CT. Acetaminophen Overdose Reveals PAR4 as a Low-Expressing but Potent Receptor on the Hepatic Endothelium in Mice. Arterioscler Thromb Vasc Biol 2025; 45:53-71. [PMID: 39360412 DOI: 10.1161/atvbaha.124.321353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024]
Abstract
BACKGROUND The protease thrombin, which elicits multiple physiological and pathological effects on vascular endothelial cells (ECs), can signal through PARs (protease-activated receptors) 1 and 4. PAR1 is a high-affinity thrombin receptor known to signal on ECs, whereas PAR4 is a low-affinity thrombin receptor, and evidence for its expression and function on ECs is mixed. This study aims to exploit the high levels of thrombin generation and hepatic vascular dysfunction that occur during acetaminophen (APAP) overdose to determine (1) whether hepatic endothelial PAR4 is a functional receptor, and (2) the endothelial-specific functions for PAR1 and PAR4 in a high thrombin and pathological setting. METHODS We generated mice with conditional deletion of Par1/Par4 in ECs and overdosed them with APAP. Hepatic vascular permeability, erythrocyte accumulation in the liver, thrombin generation, and liver function were assessed following overdose. Additionally, we investigated the expression levels of endothelial PARs and how they influence transcription in APAP-overdosed liver ECs using endothelial translating ribosome affinity purification followed by next-generation sequencing. RESULTS We found that mice deficient in high-expressing endothelial Par1 or low-expressing Par4 had equivalent reductions in APAP-induced hepatic vascular instability, although mice deficient for both receptors had lower vascular permeability at an earlier timepoint after APAP overdose than either of the single mutants. Additionally, mice with loss of both endothelial Par1 and Par4 had reduced thrombin generation after APAP overdose, suggesting decreased hypercoagulability. Last, we found that endothelial PAR1-but not PAR4-can regulate transcription in hepatic ECs. CONCLUSIONS Low-expressing PAR4 can react similarly to high-expressing PAR1 in APAP-overdosed hepatic ECs, demonstrating that PAR4 is a potent thrombin receptor. Additionally, these receptors are functionally redundant but act divergently in their expression and ability to influence transcription in hepatic ECs.
Collapse
MESH Headings
- Animals
- Acetaminophen/toxicity
- Receptors, Thrombin/metabolism
- Receptors, Thrombin/genetics
- Liver/metabolism
- Liver/drug effects
- Liver/pathology
- Receptor, PAR-1/metabolism
- Receptor, PAR-1/genetics
- Receptor, PAR-1/deficiency
- Thrombin/metabolism
- Endothelial Cells/metabolism
- Endothelial Cells/drug effects
- Endothelial Cells/pathology
- Mice, Knockout
- Mice, Inbred C57BL
- Disease Models, Animal
- Capillary Permeability/drug effects
- Male
- Drug Overdose/metabolism
- Signal Transduction
- Mice
- Chemical and Drug Induced Liver Injury/metabolism
- Chemical and Drug Induced Liver Injury/pathology
- Chemical and Drug Induced Liver Injury/genetics
- Chemical and Drug Induced Liver Injury/etiology
- Cells, Cultured
- Receptors, Proteinase-Activated
Collapse
Affiliation(s)
- Rahul Rajala
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Harold Hamm Diabetes Center, Oklahoma City, OK (R.R.)
| | - Audrey C A Cleuren
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (R.R., A.C.A.C., C.T.G.)
| | - Courtney T Griffin
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation, Oklahoma City (R.R., A.C.A.C., C.T.G.)
- Department of Cell Biology, University of Oklahoma Health Sciences Center, Oklahoma City (R.R., A.C.A.C., C.T.G.)
| |
Collapse
|
41
|
Yaita N, Maruyama K, Hiroyasu K, Sato S. Immunogenic effects of enamel matrix derivative on human alveolar ridge mucosa-derived vascular endothelial cells under lipopolysaccharide stimulation. Odontology 2025; 113:180-190. [PMID: 38839677 DOI: 10.1007/s10266-024-00959-5] [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: 02/08/2024] [Accepted: 05/28/2024] [Indexed: 06/07/2024]
Abstract
Early peri-implant disease detection remains difficult. Enamel matrix derivative (EMD), which is used for periodontal tissue regeneration, promotes leukocyte chemotactic factor and adhesion molecule expression in vascular endothelial cells. We hypothesized that stimulating vascular endothelial cells with EMD would induce an inflammatory response in the peri-implant mucosa, enabling early peri-implant infection detection. To verify this hypothesis, we assessed the intercellular adhesion between human alveolar ridge mucosa-derived vascular endothelial cells (ARMEC) stimulated with lipopolysaccharide (LPS) and EMD and human periodontal ligament-derived vascular endothelial cells (PDLEC). Leukocyte chemotactic factors and cell adhesion molecules were investigated and we established an experimental model of peri-implant disease by stimulating ARMEC (representing the peri-implant mucosa) with Porphyromonas gingivalis-derived LPS. ARMEC and PDLEC were obtained from patients (n = 6) who visited the Nippon Dental University Niigata Hospital. The cells were divided into four subcategories, each cultured with: LPS (1 µg/mL), EMD (100 µg/mL), LPS + EMD, and pure medium. Cell viability, leukocyte chemotactic factor (interleukin-8: IL-8), adhesion molecules (intercellular adhesion molecule-1: ICAM-1), tight junction protein gene expression (zonula occludens-1: ZO-1 and Occludin), and transendothelial electrical resistance (TEER) was then determined. LPS reduced ARMEC viability, whereas simultaneous stimulation with EMD improved it. LPS and EMD stimulation enhanced IL-8 and ICAM-1 gene expression, suppressed TEER, and decreased ZO-1 and Occludin expression levels compared to that with stimulation with LPS alone. EMD stimulates leukocyte migration, increase vascular permeability, and trigger an immune response in the peri-implant mucosa, thus facilitating the early detection and treatment of peri-implant disease.
Collapse
Affiliation(s)
- Naomichi Yaita
- Field of Advanced Conservative Dentistry and Periodontology, Periodontology, Course of Clinical Science, The Nippon Dental University Graduate School Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan.
| | - Kosuke Maruyama
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Kazuhiko Hiroyasu
- Oral Implant Care Unit Niigata Hospital, The Nippon Dental University, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| | - Soh Sato
- Field of Advanced Conservative Dentistry and Periodontology, Periodontology, Course of Clinical Science, The Nippon Dental University Graduate School Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
- Department of Periodontology, The Nippon Dental University School of Life Dentistry at Niigata, 1-8 Hamaura-cho, Chuo-ku, Niigata, 951-8580, Japan
| |
Collapse
|
42
|
Li G, Gao J, Ding P, Gao Y. The role of endothelial cell-pericyte interactions in vascularization and diseases. J Adv Res 2025; 67:269-288. [PMID: 38246244 PMCID: PMC11725166 DOI: 10.1016/j.jare.2024.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Endothelial cells (ECs) and pericytes (PCs) are crucial components of the vascular system, with ECs lining the inner layer of blood vessels and PCs surrounding capillaries to regulate blood flow and angiogenesis. Intercellular communication between ECs and PCs is vital for the formation, stability, and function of blood vessels. Various signaling pathways, such as the vascular endothelial growth factor/vascular endothelial growth factor receptor pathway and the platelet-derived growth factor-B/platelet-derived growth factor receptor-β pathway, play roles in communication between ECs and PCs. Dysfunctional communication between these cells is associated with various diseases, including vascular diseases, central nervous system disorders, and certain types of cancers. AIM OF REVIEW This review aimed to explore the diverse roles of ECs and PCs in the formation and reshaping of blood vessels. This review focused on the essential signaling pathways that facilitate communication between these cells and investigated how disruptions in these pathways may contribute to disease. Additionally, the review explored potential therapeutic targets, future research directions, and innovative approaches, such as investigating the impact of EC-PCs in novel systemic diseases, addressing resistance to antiangiogenic drugs, and developing novel antiangiogenic medications to enhance therapeutic efficacy. KEY SCIENTIFIC CONCEPTS OF REVIEW Disordered EC-PC intercellular signaling plays a role in abnormal blood vessel formation, thus contributing to the progression of various diseases and the development of resistance to antiangiogenic drugs. Therefore, studies on EC-PC intercellular interactions have high clinical relevance.
Collapse
Affiliation(s)
- Gan Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China
| | - Peng Ding
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| | - Youshui Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China; Institute of Microsurgery on Extremities, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, China.
| |
Collapse
|
43
|
Xiang Z, Chen H, Wu F, Pan H. Polyamino Acid Based Zwitterionic Coating can Inhibit Coagulation and Inflammation Through Anti-Fouling and Restoring Microenvironment. Macromol Biosci 2025; 25:e2400336. [PMID: 39513645 DOI: 10.1002/mabi.202400336] [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: 07/16/2024] [Revised: 09/20/2024] [Indexed: 11/15/2024]
Abstract
Protein adhesion and thrombosis formation caused by limited surface properties pose great challenges to biomedical implants. Although various hydrophilic coating or drug release coatings are reported, the single coating cannot cope with cases under the condition of complex physiological environment, which causes the coating effect is limited. In this study, a polyamino acid-derived zwitterionic coating is constructed to eliminate reactive oxygen species (ROS) in the microenvironment. It is demonstrated that the coating has excellent hydrophilicity, stability, and lubricity, and can obviously prevent protein adhesion. At the same time, the coating can eliminate hydrogen peroxide and maintain the stability of the microenvironment. The in vivo and in vitro experiments show that the coating has good biocompatibility, and inhibits thrombus. Amino acid zwitterion coating prevents protein deposition, alleviates the inflammatory process, inhibit of thrombosis, reduces the risk of implantable medical devices, and prolongs their service time. Hence, the work paves a new way to develop amino acid based zwitterionic polymer coating that can reduce the implant complications.
Collapse
Affiliation(s)
- Zehong Xiang
- Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong, 519000, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
| | - Honghong Chen
- Chen, State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin, 130022, China
| | - Feng Wu
- Zhuhai Institute of Advanced Technology, Chinese Academy of Sciences, Zhuhai, Guangdong, 519000, China
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
| | - Haobo Pan
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, 518000, China
| |
Collapse
|
44
|
Wei W, Tang M, Wang Q, Li X. Circ_HECW2 regulates ox-LDL-induced dysfunction of cardiovascular endothelial cells by miR-942-5p/TLR4 axis. Clin Hemorheol Microcirc 2025; 89:1-14. [PMID: 36213989 DOI: 10.3233/ch-221550] [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: 11/15/2022]
Abstract
BACKGROUND Coronary artery disease (CAD) is a common coronary artery disease. The functional mechanism of circular RNA (circRNA) HECT, C2 and WW domain containing E3 ubiquitin protein ligase 2 (circ_HECW2, hsa_circ_0057583) in ox-LDL-treated human cardiac microvascular endothelial cells (hCMECs) is still unclear. METHODS Expression levels of circ_HECW2, microRNA (miR)-942-5p, and toll-like receptor 4 (TLR4) were analyzed by quantitative real-time PCR (qRT-PCR) and western blot assays. Cell proliferation and apoptosis were analyzed by 5-ethynyl-2'-deoxyuridine (EdU) assay, cell counting kit-8 (CCK8) assay, and flow cytometry, respectively. Tube formation assay was performed to analyze the angiogenesis of cells. Luciferase reporter and RNA pull-down assays were performed to analyze the target relationship among circ_HECW2, miR-942-5p and TLR4. RESULTS Circ_HECW2 and TLR4 expression levels were up-regulated and miR-942-5p expression was decreased in the serum of CAD patients and oxidized low-density lipoprotein (ox-LDL)-induced hCMECs. Knockdown of circ_HECW2 enhanced cell proliferation and inhibited cell apoptosis in ox-LDL-treated hCMECs. MiR-942-5p was the target of circ_HECW2 and directly targeted TLR4. Moreover, the effect of circ_HECW2 knockdown could be weakened by anti-miR-942-5p, and TLR4 could restore the function of miR-942-5p on cell damage of ox-LDL-induced hCMECs. CONCLUSION Circ_HECW2 could regulate ox-LDL-induced cardiovascular endothelial cell dysfunction through targeting miR-942-5p/TLR4 axis.
Collapse
Affiliation(s)
- Wenbo Wei
- Department of Cardiology, Nanjing Jiangning Hospital of Traditional Chinese Medicine, Nanjing City, Jiangsu, China
| | - Min Tang
- Department of Cardiology, Nanjing Tongren Hospital Affiliated to Southeast University School of Medicine, Nanjing City, Jiangsu, China
| | - Qi Wang
- Department of Cardiology, Nanjing Tongren Hospital Affiliated to Southeast University School of Medicine, Nanjing City, Jiangsu, China
| | - Xiaoming Li
- Emergency Department, Ben Q Hospital Affiliated to Nanjing Medical University, Nanjing City, Jiangsu, China
| |
Collapse
|
45
|
Li WW, Guo ZM, Wang BC, Liu QQ, Zhao WA, Wei XL. PCSK9 induces endothelial cell autophagy by regulating the PI3K/ATK pathway in atherosclerotic coronary heart disease. Clin Hemorheol Microcirc 2025; 89:55-67. [PMID: 38728182 DOI: 10.3233/ch-242172] [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: 05/12/2024]
Abstract
OBJECTIVE Atherosclerosis is a chronic inflammatory disease of the arteries, and its pathogenesis is related to endothelial dysfunction. It has been found that the protein convertase subtilin/kexin9 type (PCSK9) plays an important role in AS, but its specific mechanism is still unclear. METHODS In this study, we first cultured human umbilical vein endothelial cells (HUVECs) with 50 or 100μg/ml oxidized low-density lipoprotein (ox-LDL) for 24 hours to establish a coronary atherosclerosis cell model. RESULTS The results showed that ox-LDL induced HUVEC injury and autophagy and upregulated PCSK9 protein expression in HUVECs in a concentration-dependent manner. Silencing PCSK9 expression with siRNA inhibited ox-LDL-induced HUVEC endothelial dysfunction, inhibited the release of inflammatory factors, promoted HUVEC proliferation and inhibited apoptosis. In addition, ox-LDL increased the expression of LC3B-I and LC3B-II and decreased the expression of p62. However, these processes are reversed by sh-PCSK9. In addition, sh-PCSK9 can inhibit PI3K, AKT and mTOR phosphorylation and promote autophagy. CONCLUSION Taken together, our research shows that silencing PCSK9 inhibits the PI3K/ATK/mTOR pathway to activate ox-LDL-induced autophagy in vascular endothelial cells, alleviating endothelial cell injury and inflammation.
Collapse
Affiliation(s)
- Wei-Wei Li
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Ze-Ming Guo
- Department of Neurology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Bing-Cai Wang
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Qing-Quan Liu
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Wen-An Zhao
- Department of Cardiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Xiao-Lan Wei
- Department of Neurology, Quanzhou First Hospital Affiliated to Fujian Medical University, Quanzhou, China
| |
Collapse
|
46
|
Campos OA, Garcia-Herreros A, Sánchez AL, Fineman JR, Pawlak G. A Multichamber Pulsating-Flow Device With Optimized Spatial Shear Stress and Pressure for Endothelial Cell Testing. J Biomech Eng 2025; 147:011006. [PMID: 39382480 PMCID: PMC11625645 DOI: 10.1115/1.4066800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/03/2024] [Accepted: 10/04/2024] [Indexed: 10/10/2024]
Abstract
Design and analysis are presented for a new device to test the response of endothelial cells to the simultaneous action of cyclic shear stresses and pressure fluctuations. The design consists of four pulsatile-flow chambers connected in series, where shear stress is identical in all four chambers and pressure amplitude decreases in successive chambers. Each flow chamber is bounded above and below by two parallel plates separated by a small gap. The design of the chamber planform must ensure that cells within the testing region experience spatially uniform time-periodic shear stress. For conditions typically encountered in applications, the viscous unsteady flow exhibits order-unity values of the associated Womersley number. The corresponding solution to the unsteady lubrication problem, with general nonsinusoidal flowrate, is formulated in terms of a stream function satisfying Laplace's equation, which can be integrated numerically to determine the spatial distribution of shear stresses for chambers of general planform. The results are used to optimize the design of a device with a hexagonal planform. Accompanying experiments using particle tracking velocimetry (PTV) in a fabricated chamber were conducted to validate theoretical predictions. Pressure readings indicate that intrachamber pressure variations associated with viscous pressure losses and acoustic fluctuations are relatively small, so that all cells in a given testing region experience nearly equal pressure forces.
Collapse
Affiliation(s)
- Obed A. Campos
- Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093-0411
| | - Antoni Garcia-Herreros
- Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093-0411
| | - Antonio L. Sánchez
- Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093-0411
| | - Jeffrey R. Fineman
- Department of Pediatrics, University of San Francisco, San Francisco, CA 94158; Cardiovascular Research Institute, University of San Francisco, San Francisco, CA 94158
| | - Geno Pawlak
- Mechanical and Aerospace Engineering, University of California San Diego, La Jolla, CA 92093-0411
| |
Collapse
|
47
|
Zhang R, Sun L, Li Y, Li C, Zheng X, Hao Y. Risk Factors for Penetrating Aortic Ulcer Progression. Ann Vasc Surg 2025; 110:23-31. [PMID: 39419321 DOI: 10.1016/j.avsg.2024.08.034] [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: 06/21/2024] [Revised: 08/09/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024]
Abstract
BACKGROUND This study aimed to explore risk factors leading to asymptomatic penetrating aortic ulcer (PAU) progression. METHODS This retrospective study reviewed the clinical data of patients who were diagnosed with asymptomatic PAU through incidental imaging findings and underwent imaging follow-up between August 2018 and July 2022. Patients were grouped according to ulcer progression. The risk factors for PAU progression were also analyzed. RESULTS Among 60 patients with PAU, 32 (53.33%) experienced PAU progression. The mean follow-up time was 555.72 ± 407.60 days. Although there was no statistically significant difference in cancer incidence between the PAU progression group and nonprogression group (24 [75%] vs. 18 [64.28%], P = 0.409), the difference in antineoplastic therapy use between the progression and nonprogression groups was significant (19 [59.38%] vs. 7 [25.00%], P = 0.010). There was no difference in the aortic diameter at the PAU (20.68 ± 4.16 mm vs. 20.70 ± 5.28 mm, P = 0.990), PAU width (7.32 ± 2.53 mm vs. 7.11 ± 2.29 mm, P = 0.741), and PAU depth (4.13 ± 1.26 mm vs. 4.08 ± 1.41 mm, P = 0.880) between the 2 groups. In the progression group, the progression rates of aortic diameter at PAU, PAU width, and PAU depth were 2.16 ± 4.28 mm/year, 5.91 ± 14.49 mm/year and 2.87 ± 5.87 mm/year, respectively. Binary logistic regression analysis showed that antineoplastic therapy was an independent predictor of PAU progression (P = 0.017; odds ratio, 4.144; 95% confidence interval, 1.290-13.316). CONCLUSIONS Antineoplastic therapy may contribute to the progression of asymptomatic PAU in this retrospective study with small number of patients. Patients with asymptomatic PAU who are receiving or have completed antineoplastic therapy should be more vigilant regarding PAU progression.
Collapse
Affiliation(s)
- Rongjie Zhang
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Long Sun
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Yu Li
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Chao Li
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Xichuan Zheng
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, PR China
| | - Yingxue Hao
- Department of Vascular Surgery, Southwest Hospital, Army Medical University, Chongqing, PR China.
| |
Collapse
|
48
|
Wang E, Feng B, Chen S, Su Z, Chakrabarti S. Differential microvascular endothelial cell responses in the retina in diabetes compared to the heart and kidneys, a spatial transcriptomic analysis. PLoS One 2024; 19:e0310949. [PMID: 39739865 DOI: 10.1371/journal.pone.0310949] [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: 06/22/2024] [Accepted: 09/10/2024] [Indexed: 01/02/2025] Open
Abstract
Endothelial cells and high glucose-induced endothelial dysfunction are the common origin of chronic diabetic complications such as retinopathy, nephropathy, and cardiomyopathy. Yet their common origins, the vascular manifestations of such complications are different. We examined the basal heterogeneity between microvascular endothelial cells(MECs) from the retina, kidneys, and heart, as well as their differential responses to hyperglycemia in diabetes. To this extent, we used a spatial transcriptomic approach to investigate gene expression differences across retinal, renal, and cardiac MECs in diabetic and non-diabetic mouse models. We validated MEC heterogeneity in vitro using human retinal and cardiac MECs. The spatial transcriptomic approach was also used to explore potential similarities in retinal MECs and neuronal cells in response to hyperglycemia. We found that MECs from different target organs of major diabetic complications were transcriptomically distinct at the basal state and respond differently to hyperglycemia. These findings were recapitulated in cell culture, with selected analytes. We found minimal similarities between retinal MECs and neuronal cells. Our findings show considerable heterogeneity across retinal, renal, and cardiac MECs, both at the basal state and in their responses to hyperglycemia in diabetes. These findings show that organ specific MEC heterogeneity can influence differential development of pathological changes across various target organs of chronic diabetic complications, and suggest that MEC heterogeneity may influence treatment target(s) and drug development.
Collapse
Affiliation(s)
- Eric Wang
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Biao Feng
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Shali Chen
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| | - Zhaoliang Su
- International Genome Center, Jiangsu University, Zhenjiang, China
| | - Subrata Chakrabarti
- Department of Pathology and Laboratory Medicine, Western University, London, Ontario, Canada
| |
Collapse
|
49
|
Yan M, Wang Z, Qiu Z, Cui Y, Xiang Q. Platelet signaling in immune landscape: comprehensive mechanism and clinical therapy. Biomark Res 2024; 12:164. [PMID: 39736771 DOI: 10.1186/s40364-024-00700-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 11/28/2024] [Indexed: 01/01/2025] Open
Abstract
Platelets are essential for blood clotting and maintaining normal hemostasis. In pathological conditions, platelets are increasingly recognized as crucial regulatory factors in various immune-mediated inflammatory diseases. Resting platelets are induced by various factors such as immune complexes through Fc receptors, platelet-targeting autoantibodies and other platelet-activating stimuli. Platelet activation in immunological processes involves the release of immune activation stimuli, antigen presentation and interaction with immune cells. Platelets participate in both the innate immune system (neutrophils, monocytes/macrophages, dendritic cells (DCs) and Natural Killer (NK) cells and the adaptive immune system (T and B cells). Clinical therapeutic strategies include targeting platelet activation, platelet-immune cell interaction and platelet-endothelial cell interaction, which display positive development prospects. Understanding the mechanisms of platelets in immunity is important, and developing targeted modulations of these mechanisms will pave the way for promising therapeutic strategies.
Collapse
Affiliation(s)
- Mengyao Yan
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhe Wang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Zhiwei Qiu
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China
| | - Yimin Cui
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| | - Qian Xiang
- Institute of Clinical Pharmacology, Peking University First Hospital, Beijing, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University Health Science Center, Beijing, China.
| |
Collapse
|
50
|
Panda P, Mohanty S, Gouda SR, Mohapatra R. Advances in nanomedicine for retinal drug delivery: overcoming barriers and enhancing therapeutic outcomes. J Drug Target 2024:1-25. [PMID: 39694681 DOI: 10.1080/1061186x.2024.2443144] [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: 10/22/2024] [Revised: 11/16/2024] [Accepted: 12/07/2024] [Indexed: 12/20/2024]
Abstract
Nanomedicine offers a promising avenue for improving retinal drug delivery, effectively addressing challenges associated with ocular diseases like age-related macular degeneration and diabetic retinopathy. Nanoparticles, with their submicron size and customisable surface properties, enable enhanced permeability and retention within retinal tissues, supporting sustained drug release and minimising systemic side effects. Nanostructured scaffolds further provide a supportive environment for retinal cell growth and tissue regeneration, crucial for treating degenerative conditions. Additionally, advanced nanodevices facilitate real-time monitoring and controlled drug release, marking significant progress in retinal therapy. This study reviews recent advancements in nanomedicine for retinal drug delivery, critically analysing design innovations, therapeutic benefits, and limitations of these systems. By advancing nanotechnology integration in ocular therapies, this field holds strong potential for overcoming current barriers, ultimately improving patient outcomes and quality of life.
Collapse
Affiliation(s)
- Pratikeswar Panda
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Shreyashree Mohanty
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Sangita Ranee Gouda
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| | - Rajaram Mohapatra
- Department of Pharmaceutics, School of Pharmaceutical Science, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, India
| |
Collapse
|