1
|
Goyal A, Afzal M, Khan NH, Goyal K, Srinivasamurthy SK, Gupta G, Benod Kumar K, Ali H, Rana M, Wong LS, Kumarasamy V, Subramaniyan V. Targeting p53-p21 signaling to enhance mesenchymal stem cell regenerative potential. Regen Ther 2025; 29:352-363. [PMID: 40248767 PMCID: PMC12004386 DOI: 10.1016/j.reth.2025.03.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/09/2025] [Accepted: 03/18/2025] [Indexed: 04/19/2025] Open
Abstract
Mesenchymal stem cells (MSCs) are properties of self-renewal and differentiation potentials and thus are very appealing to regenerative medicine. Nevertheless, their therapeutic potential is frequently constrained by senescence, limited proliferation, and stress-induced apoptosis. The key role of the p53-p21 biology in MSC biology resides in safeguarding genomic stability while promoting senescence and limiting regenerative capacity upon over-activation demonstrated. This pathway is a key point for improving MSC function and exploiting the inherent limitations. Recent advances indicate that senescence can be delayed by targeting the p53-p21 signaling and improved MSC proliferation and differentiation capacity. PFT-α pharmacological agents transiently inhibit p53 from increasing proliferation and lineage-specific differentiation, while antioxidants such as hydrogen-rich saline and epigallocatechin 3 gallate (EGCG) suppress oxidative stress and attenuate p53 p21 signaling. Genetic tools like CRISPR-Cas9 and RNA interference also precisely modulate TP53 and CDKN1A expression to optimize MSC functionality. The interplay of p53-p21 with pathways like Wnt/β-catenin and MAPK further highlights opportunities for combinatorial therapies to enhance MSC resilience and regenerative outcomes. This review aims to offer a holistic view of how p53-p21 targeting can further the regenerative potential of MSCs, resolving senescence, proliferation, and stress resilience towards advanced therapeutics built on MSCs.
Collapse
Affiliation(s)
- Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suresh Kumar Srinivasamurthy
- Department of Pharmacology, Ras Al Khaimah College of Medical Sciences, Ras Al Khaimah Medical & Health Sciences University, P.O. Box 11172, Ras Al Khaimah, United Arab Emirates
| | - Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - K. Benod Kumar
- Department of General Surgery, Consultant Head and Neck Surgical Oncology, Dr.D.Y.Patil Medical College, Hospital and Research Centre, Pimpri, Pune, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Ling Shing Wong
- Faculty of Health and Life Sciences, INTI International University, Nilai 71800, Malaysia
| | - Vinoth Kumarasamy
- Department of Parasitology and Medical Entomology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000, Kuala Lumpur, Malaysia
| | - Vetriselvan Subramaniyan
- Division of Pharmacology, Faculty of Medical and Life Sciences, Sunway University, Bandar Sunway, 47500 Selangor Darul Ehsan, Malaysia
| |
Collapse
|
2
|
Tai J, Wang L, Xie Y, Li Y, Fu H, Ma X, Li H, Li X, Yan Z, Liu J. Research on multi-algorithm and explainable AI techniques for predictive modeling of acute spinal cord injury using multimodal data. Sci Rep 2025; 15:18832. [PMID: 40442476 PMCID: PMC12122863 DOI: 10.1038/s41598-025-93006-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 03/04/2025] [Indexed: 06/02/2025] Open
Abstract
Machine learning technology has been extensively applied in the medical field, particularly in the context of disease prediction and patient rehabilitation assessment. Acute spinal cord injury (ASCI) is a sudden trauma that frequently results in severe neurological deficits and a significant decline in quality of life. Early prediction of neurological recovery is crucial for the personalized treatment planning. While extensively explored in other medical fields, this study is the first to apply multiple machine learning methods and Shapley Additive Explanations (SHAP) analysis specifically to ASCI for predicting neurological recovery. A total of 387 ASCI patients were included, with clinical, imaging, and laboratory data collected. Key features were selected using univariate analysis, Lasso regression, and other feature selection techniques, integrating clinical, radiomics, and laboratory data. A range of machine learning models, including XGBoost, Logistic Regression, KNN, SVM, Decision Tree, Random Forest, LightGBM, ExtraTrees, Gradient Boosting, and Gaussian Naive Bayes, were evaluated, with Gaussian Naive Bayes exhibiting the best performance. Radiomics features extracted from T2-weighted fat-suppressed MRI scans, such as original_glszm_SizeZoneNonUniformity and wavelet-HLL_glcm_SumEntropy, significantly enhanced predictive accuracy. SHAP analysis identified critical clinical features, including IMLL, INR, BMI, Cys C, and RDW-CV, in the predictive model. The model was validated and demonstrated excellent performance across multiple metrics. The clinical utility and interpretability of the model were further enhanced through the application of patient clustering and nomogram analysis. This model has the potential to serve as a reliable tool for clinicians in the formulation of personalized treatment plans and prognosis assessment.
Collapse
Affiliation(s)
- Jiaojiao Tai
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Linbang Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, 100191, China
| | - Yijun Xie
- Xi'an Central Hospital, Xi'an, Shaanxi, China
| | - Yang Li
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Hua Fu
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Xiaowen Ma
- Department of Magnetic Resonance Imaging, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Haiyan Li
- Department of Magnetic Resonance Imaging, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Xinying Li
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China
| | - Ziqiang Yan
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| | - Jingkun Liu
- Department of Orthopedics, Honghui Hospital, Xi'an Jiaotong University, Xi'an, 710054, Shaanxi, China.
| |
Collapse
|
3
|
Reid G, Cerino G, Melly L, Fusco D, Zhang C, Reuthebuch O, Milan G, Marsano A. Harnessing the angiogenic potential of adipose-derived stromal vascular fraction cells with perfusion cell seeding. Stem Cell Res Ther 2025; 16:220. [PMID: 40312732 PMCID: PMC12044990 DOI: 10.1186/s13287-025-04286-6] [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: 01/21/2025] [Accepted: 03/19/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND The rapid formation and long-term maintenance of functional vascular networks are crucial for the success of regenerative therapies. The stromal vascular fraction (SVF) from human adipose tissue is a readily available, heterogeneous cell source containing myeloid lineage cells, mesenchymal stromal cells, endothelial cells and their precursors, and pericytes, which are important for vascular support. Previous studies showed that seeding SVF cells under perfusion and pre-culturing them on three-dimensional (3D) collagen sponges enhances the vascular cell component in vitro while accelerating vascularization and improving human cell engraftment in vivo compared to static pre-culture. However, generating a perfusion-cultured SVF patch over a 5-day period is both costly and challenging for clinical translation. To overcome these limitations, this study explores a no-pre-culture strategy by comparing perfusion-based seeding with static cell loading on 3D sponges. The hypothesis is that perfusion-based seeding enhances in vivo cell engraftment and angiogenic potential by loading different SVF cell subpopulations onto 3D scaffolds during the seeding process. METHODS SVF-cells are seeded onto collagen scaffold using two approaches: a closed system perfusion bioreactor for 18 h or static loading onto the sponge surface. The in vitro cell distribution and baseline cytokine profiles were evaluated. Subsequently, human cell engraftment and differentiation were assessed in vivo using a nude rat subcutaneous implantation model. Analyses included the survival of transplanted human cells, the functionality and maturation of newly formed blood vessels within the SVF-patch. RESULTS Perfusion seeding significantly reduced the number of myeloid cells and achieved uniform spatial distribution across the construct. Vascular endothelial growth factor release was significantly increased following perfusion culture, whereas pro-inflammatory cytokines such as tumor necrosis factor-α and interleukin-1β were decreased. In the short term, perfusion culture enhanced uniform vascularization and SVF cell engraftment in vivo. However, the long-term differences between the perfusion-seeded and static-seeded groups diminished. CONCLUSION Eliminating the need for prolonged pre-culture offers a feasible and cost-effective strategy for advancing regenerative cell-based therapies by reducing pre-culture times while preserving therapeutic efficacy. Perfusion-based seeding offers significant short-term benefits, including enhanced vascularization and cell engraftment, though long-term differences compared to static seeding are minimal. Further investigation is needed to evaluate its potential in a diseased ischemic heart model.
Collapse
Affiliation(s)
- Gregory Reid
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
- Department of Plastic and Hand Surgery, University Hospital of Zürich, 8091, Zurich, Switzerland
| | - Giulia Cerino
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Ludovic Melly
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Deborah Fusco
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Chunyan Zhang
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Oliver Reuthebuch
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Giulia Milan
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland
- Department of Cardiac Surgery, University Hospital of Basel, 4031, Basel, Switzerland
| | - Anna Marsano
- Cardiac Surgery and Engineering Group, Department of Biomedicine, University of Basel and University Hospital of Basel, 4031, Basel, Switzerland.
| |
Collapse
|
4
|
Tang C. Immunologic cell deaths: involvement in the pathogenesis and intervention therapy of periodontitis. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:4957-4968. [PMID: 39718617 DOI: 10.1007/s00210-024-03732-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/12/2024] [Indexed: 12/25/2024]
Abstract
Periodontitis is one of the most common diseases and primary causes of tooth loss. The main factor that causes periodontitis is an overactive host immunological response. An in-depth investigation into the molecular pathways that cause periodontitis can aid in creating novel therapeutic approaches for periodontitis and its related systemic disorders. Several immunologic cell death (ICD) pathways have been implicated in advancing periodontitis. Nevertheless, there is still a substantial lack of understanding surrounding the precise molecular mechanisms of ICD in periodontitis. Additionally, the beneficial feature of ICD in periodontitis, which involves its ability to eliminate pathogens, needs further confirmation. According to this, a comprehensive literature search utilizing the Web of Science™, PubMed®, and Scopus® databases was conducted. Only items published in the English language up until October 2024 were taken into account, and finally, 65 relevant papers were selected to be included in this review. In this article, we present a comprehensive analysis of the processes and outcomes of ICD activation in the progression of periodontitis. Lastly, the present difficulties linked to ICDs as a viable treatment option for periodontitis are emphasized.
Collapse
Affiliation(s)
- Chenyao Tang
- Department of Stomatology, Yongzhou Central Hospital, Yongzhou, Hunan Province, 425000, China.
| |
Collapse
|
5
|
Zhang X, Liu T, Ma Z, Li G, Ding N, Wang Z, Guan Y, Zhang Y, Liu L, Chen X. VEGF secreted by human dental pulp stem cell promotes spinal cord injury repair by inhibiting microglial pyroptosis through the PI3K/AKT pathway. J Transl Med 2025; 23:437. [PMID: 40221710 PMCID: PMC11992863 DOI: 10.1186/s12967-025-06388-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 03/15/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Spinal cord injury (SCI) remains a devastating central nervous system disorder. The complex pathological microenvironment following SCI, particularly the imbalance in neuroinflammation, contributes to its therapeutic challenges. Microglial pyroptosis, a type of programmed cell death, is pivotal in exacerbating neuroinflammation and secondary tissue damage after SCI. Our previous study demonstrated the inhibitory efficacy of conditioned medium (CM) derived from human dental pulp stem cells (DPSCs) on the microglial pyroptosis and its positive effects on the functional recovery in SCI models. However, the major secretory product in CM responsible for inhibiting microglial pyroptosis remains unclear. OBJECTIVE We aim to investigate whether vascular endothelial growth factor (VEGF) secreted by human DPSCs can alleviate microglial pyroptosis through the PI3K/AKT signaling pathway and promote motor and electrophysiological function recovery in SCI mice. METHODS Human DPSCs were isolated and cultured, and CM was collected for VEGF detection and further treatment. The BV2 cell line was established as a microglial pyroptosis model through the administration of lipopolysaccharide (LPS). SCI was induced in mice. Molecular and histological techniques were employed to evaluate pyroptosis and explore the underlying mechanisms both in vivo and vitro. RESULTS Human DPSC-derived VEGF significantly inhibited microglial pyroptosis both in vitro and vivo, as evidenced by the decreased expression of pyroptosis-related markers, such as caspase-1 and IL-1β. The anti-pyroptotic effects of VEGF were closely associated with the activation of the PI3K/AKT signaling pathway, which was identified as a key regulatory mechanism. Importantly, treatment with DPSC-CM improved the recovery of motor function and electrophysiological conduction in SCI mice. CONCLUSION Human DPSC-derived VEGF alleviates microglial pyroptosis via the PI3K/AKT signaling pathway, thereby contributing to the repair of SCI. Our study provides new insights into the potential for therapy of DPSCs and their secreted factors, particularly VEGF, offering new perspectives on the treatment of SCI.
Collapse
Affiliation(s)
- Xinwei Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China
| | - Tao Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China
| | - Ziqian Ma
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China
| | - Guanghao Li
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China
| | - Nan Ding
- Department of Stomatology, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China
| | - Zihang Wang
- Clinical Medical College, Fujian Medical University, Fujian, 350108, China
| | - Yun Guan
- Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Neurological Surgery, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Yan Zhang
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China.
| | - Liang Liu
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China.
| | - Xueming Chen
- Department of Orthopedics Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, 101149, China.
| |
Collapse
|
6
|
Saadh MJ, Muhammad FA, Albadr RJ, Sanghvi G, Jyothi SR, Kundlas M, Joshi KK, Rakhmatullaev A, Taher WM, Alwan M, Jawad MJ, Ali Al-Nuaimi AM. Inflammasomes and Cardiovascular Disease: Linking Inflammation to Cardiovascular Pathophysiology. Scand J Immunol 2025; 101:e70020. [PMID: 40170223 DOI: 10.1111/sji.70020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 03/15/2025] [Accepted: 03/22/2025] [Indexed: 04/03/2025]
Abstract
Cardiovascular diseases (CVDs) remain a leading cause of global mortality, driven by risk factors such as dyslipidemia, hypertension and diabetes. Recent research has highlighted the critical role of inflammasomes, particularly the NLRP3 inflammasome, in the pathogenesis of various CVDs, including hypertension, atherosclerosis, myocardial infarction and heart failure. Inflammasomes are intracellular protein complexes that activate inflammatory responses through the production of pro-inflammatory cytokines such as IL-1β and IL-18, contributing to endothelial dysfunction, plaque formation and myocardial injury. This review provides a comprehensive overview of the structure, activation mechanisms and pathways of inflammasomes, with a focus on their involvement in cardiovascular pathology. Key activation pathways include ion fluxes (K+ efflux and Ca2+ signalling), endoplasmic reticulum (ER) stress, mitochondrial dysfunction and lysosomal destabilisation. The review also explores the therapeutic potential of targeting inflammasomes to mitigate inflammation and improve outcomes in CVDs. Emerging strategies include small-molecule inhibitors, biologics and RNA-based therapeutics, with a particular emphasis on NLRP3 inhibition. Additionally, the integration of artificial intelligence (AI) in cardiovascular research offers promising avenues for identifying novel biomarkers, predicting disease risk and developing personalised treatment strategies. Future research directions should focus on understanding the interactions between inflammasomes and other immune components, as well as genetic regulators, to uncover new therapeutic targets. By elucidating the complex role of inflammasomes in CVDs, this review underscores the potential for innovative therapies to address inflammation-driven cardiovascular pathology, ultimately improving patient outcomes.
Collapse
Affiliation(s)
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - S Renuka Jyothi
- Department of Biotechnology and Genetics School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Mayank Kundlas
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, Uttarakhand, India
- Graphic Era Deemed to Be University, Dehradun, Uttarakhand, India
| | - Akmal Rakhmatullaev
- Department of Faculty Pediatric Surgery, Tashkent Pediatric Medical Institute, Tashkent, Uzbekistan
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Nasiriyah, Dhi Qar, Iraq
| | - Mariem Alwan
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| | | | | |
Collapse
|
7
|
Agrawal N, Afzal M, Khan NH, Ganesan S, Kumari M, Sunitha S, Dash A, Goyal K, Kushwaha B, Rekha A, Rana M, Ali H. The role of VEGF in vascular dementia: impact of aging and cellular senescence. Biogerontology 2025; 26:77. [PMID: 40119956 DOI: 10.1007/s10522-025-10219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Vascular Endothelial Growth Factor (VEGF) is a critical element in vascular dementia (VD) pathogenesis and therapeutic development while remaining strongly influenced by aging processes and cellular aging mechanisms. VEGF's multiple effects comprise neuroprotective functions, its role in vascular development, and its ability to regulate brain blood flow systems, all leading to cognitive preservation. The prefrontal cortex exhibits elevated VEGF gene levels, which directly matches the advancement of cognitive deficits in patients with Alzheimer's disease and VD. These patients exhibit higher VEGF levels in their CSF fluid, demonstrating that disease pathology includes multiple inseparable factors. Aging dramatically worsens VEGF regulation because endothelial dysfunction combines with chronic inflammation and oxidative stress to generate adverse vascular symptoms that include atherosclerosis and stroke. Cellular senescence convolutes these processes by causing damaging inflammatory reactions alongside impaired vascular healing abilities. The secretion of pro-inflammatory cytokines from senescent cells (SCs) disrupts VEGF signaling and produces harmful consequences for both vascular health and cognitive well-being. The neuroprotective properties of VEGF-A165a and VEGF-A165b variants demonstrate their ability to lessen β-amyloid and tau protein toxicity. The protective mechanisms of VEGF depend heavily on VEGF expression levels and receptor functionality, both of which decrease with aging. The combination of approaches that modulate VEGF signaling and SC accumulation shows potential for designing treatments against VD. People can sustain BBB functionality over a longer period through Mediterranean diet consumption together with aerobic exercise along with developing therapies, including senolytics and senomorphics, which delay neurodegenerative progression. Future investigative efforts must improve VEGF delivery methods while studying cellular senescence mechanisms and developing advanced methods to detect SC cells. A three-dimensional healthcare approach combining VEGF-targeted treatments with anti-ageing interventions and detailed diagnostic techniques shows the potential for effective VD management to achieve better patient results.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - S Sunitha
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Aniruddh Dash
- Department of Orthopaedics, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Brajgopal Kushwaha
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, 462044, India
| | - A Rekha
- Hospital and Research Centre, Dr. D. Y. Patil Medical College, Pimpri, Pune, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College,, Saveetha University, Chennai, India
| |
Collapse
|
8
|
Xiao Y, Hassani M, Moghaddam MB, Fazilat A, Ojarudi M, Valilo M. Contribution of tumor microenvironment (TME) to tumor apoptosis, angiogenesis, metastasis, and drug resistance. Med Oncol 2025; 42:108. [PMID: 40087196 DOI: 10.1007/s12032-025-02675-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
The tumor microenvironment (TME) contains tumor cells, surrounding cells, and secreted factors. It provides a favorable environment for the maintenance of cancer stem cells (CSCs), the spread of cancer cells to metastatic sites, angiogenesis, and apoptosis, as well as the growth, proliferation, invasion, and drug resistance of cancer cells. Cancer cells rely on the activation of oncogenes, inactivation of tumor suppressors, and the support of a normal stroma for their growth, proliferation, and survival, all of which are provided by the TME. The TME is characterized by the presence of various cells, including cancer-associated fibroblasts (CAFs), tumor-associated macrophages (TAMs), CD8 + cytotoxic T cells (CTLs), regulatory T cells (Tregs), myeloid-derived suppressor cells (MDSCs), mesenchymal stem cells (MSCs), endothelial cells, adipocytes, and neuroendocrine (NE) cells. The high expression of inflammatory cytokines, angiogenic factors, and anti-apoptotic factors, as well as drug resistance mechanisms in the TME, contributes to the poor therapeutic efficacy of anticancer drugs and tumor progression. Hence, this review describes the mechanisms through which the TME is involved in apoptosis, angiogenesis, metastasis, and drug resistance in tumor cells.
Collapse
Affiliation(s)
- Yanhong Xiao
- Harbin Medical University Cancer Hospital, Harbin, 150006, Heilongjiang Province, China
| | - Mahan Hassani
- Faculty of Pharmacy, Near East University, Nicosia, North Cyprus
| | | | - Ahmad Fazilat
- Department of Genetics, Motamed Cancer Institute, Breast Cancer Research Center, ACECR, Tehran, Iran
| | - Masoud Ojarudi
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Valilo
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
9
|
Yao Z, Wen H. Impact of aneurysm sac size on the effectiveness of endovascular coiling in patient-specific middle cerebral artery aneurysms: a computational study. Sci Rep 2025; 15:8825. [PMID: 40087333 PMCID: PMC11909214 DOI: 10.1038/s41598-025-92298-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 02/26/2025] [Indexed: 03/17/2025] Open
Abstract
This study investigates the hemodynamic efficiency of endovascular coiling in the treatment of patient-specific middle cerebral artery (MCA) aneurysms using computational models of both original and scaled-down geometries. Computational fluid dynamic (CFD) is used for the blood flow modelling inside cerebral aneurysms. The Casson non-Newtonian model is employed to simulate blood flow dynamics, while a porous condition represents the coiling within the aneurysm sac. Key hemodynamic factors, including wall shear stress (WSS) and oscillatory shear index (OSI), are analyzed to evaluate the effectiveness of coiling across different aneurysm sizes. Results indicate that coiling significantly reduces rupture risk, with larger sac volumes demonstrating a more pronounced decrease in high-risk hemodynamic zones. These findings offer insights into optimizing endovascular treatments for varying aneurysm morphologies.
Collapse
Affiliation(s)
- Zhichao Yao
- Department of General Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325024, China
| | - Hao Wen
- Department of Emergency, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325024, China.
| |
Collapse
|
10
|
Alaqel SI, Imran M, Khan A, Nayeem N. Aging, vascular dysfunction, and the blood-brain barrier: unveiling the pathophysiology of stroke in older adults. Biogerontology 2025; 26:67. [PMID: 40044939 DOI: 10.1007/s10522-025-10209-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: 01/23/2025] [Accepted: 02/18/2025] [Indexed: 05/09/2025]
Abstract
The progressive decline of vascular integrity and blood-brain barrier (BBB) function is associated with aging, a major risk factor for stroke. This review describes the cellular and molecular changes in the brain microvasculature of the neurovascular unit (NVU) that contribute to the development of BBB dysfunction in aging, such as endothelial cell senescence, oxidative stress, and degradation of tight junction proteins. Stroke severity and recovery are exacerbated by BBB breakdown, leading to neuroinflammation, neurotoxicity, and cerebral oedema while identifying molecular mechanisms such as the NLRP3 inflammasome, matrix metalloproteinases (MMPs), and non-coding RNAs (e.g., miRNAs and circRNAs) that drive BBB disruption in aging and stroke. Real-time assessment of BBB permeability in stroke pathophysiology is made possible using advanced imaging techniques, such as dynamic contrast-enhanced MRI and positron emission tomography. Furthermore, biomarkers, including claudin-5, PDGFRβ, or albumin concentration, serve as markers of BBB integrity and vascular health. Restoration of BBB function and stroke recovery with emerging therapeutic strategies, including sirtuin modulators (SIRT1 and SIRT3 activators to enhance endothelial function and mitochondrial health), stem cell-derived extracellular vesicles (iPSC-sEVs for BBB repair and neuroprotection), NLRP3 inflammasome inhibitors (MCC950 to attenuate endothelial pyroptosis and inflammation), hydrogen-rich water therapy (to counteract oxidative stress-induced BBB damage), and neuropeptides such as cortistatin (to regulate neuroinflammation and BBB stability), is promising. This review explores the pathophysiological mechanisms of BBB dysfunction in aging and stroke, their relation to potential therapeutic targets, and novel approaches to improve vascular health and neuroprotection.
Collapse
Affiliation(s)
- Saleh I Alaqel
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- King Salman Center for Disability Research, 11614, Riyadh, Saudi Arabia.
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center For Health Research, Northern Border University, Arar, Saudi Arabia
| | - Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center For Health Research, Northern Border University, Arar, Saudi Arabia
| | - Naira Nayeem
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
| |
Collapse
|
11
|
Zhang T, Liu M. Cytokines in age-related eye diseases: pathogenesis and potential targets for innovative therapies. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03926-1. [PMID: 40021512 DOI: 10.1007/s00210-025-03926-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 02/12/2025] [Indexed: 03/03/2025]
Abstract
Age-related eye diseases (AREDs), such as dry eye disease (DED), age-related macular degeneration (AMD), glaucoma, and diabetic retinopathy (DR), are significant worldwide health concerns due to their rising prevalence and debilitating effects. Despite substantial research on the pathobiology of AREDs, the impact of immune-related alterations caused by aging is still not well understood. Tissue-resident cells and invading immune cells in the eye control innate responses in the event of damage or infection. However, as cells age, they gradually lose their ability to perform their protective duties and develop abnormal characteristics. Therefore, the disrupted regulation of immune responses in the eyes of older individuals enhances their vulnerability to and the intensity of eye disorders. Cytokines, immune system components, have a role in developing AREDs by contributing to inflammation. This paper examines the deficiencies in the pathogenic and therapeutic aspects of pro-inflammatory cytokines in AREDs that require further investigation in future studies.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China
| | - Ming Liu
- Department of Gynecology and Obstetrics, The Second Affiliated Hospital of Xi'an Jiaotong University (Xibei Hospital), Xi'an, 710004, China.
| |
Collapse
|
12
|
Amin A, Saadatakhtar M, Mohajerian A, Marashi SM, Zamanifard S, Keshavarzian A, Molaee P, Keshmiri MS, Nikdoust F. Mercury-Mediated Cardiovascular Toxicity: Mechanisms and Remedies. Cardiovasc Toxicol 2025; 25:507-522. [PMID: 39904862 DOI: 10.1007/s12012-025-09966-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/22/2025] [Indexed: 02/06/2025]
Abstract
Mercury is a significant environmental pollutant and public health threat, primarily recognized for its neurotoxic effects. Increasing evidence also highlights its harmful impact on the cardiovascular system, particularly in adults. Exposure to mercury through contaminated soil, air, or water initiates a cascade of pathological events that lead to organ damage, including platelet activation, oxidative stress, enhanced inflammation, and direct injury to critical cells such as cardiomyocytes and endothelial cells. Endothelial activation triggers the upregulation of adhesion molecules, promoting the recruitment of leukocytes and platelets to vascular sites. These interactions activate both platelets and immune cells, creating a pro-inflammatory, prothrombotic environment. A key outcome is the formation of platelet-leukocyte aggregates (PLAs), which exacerbate thromboinflammation and endothelial dysfunction. These processes significantly elevate cardiovascular risks, including thrombosis and vascular inflammation. This study offers a comprehensive analysis of the mechanisms underlying mercury-induced cardiotoxicity, focusing on oxidative stress, inflammation, and cellular dysfunction.
Collapse
Affiliation(s)
- Arash Amin
- Department of Cardiology, School of Medicine, Shahid Madani Hospital, Lorestan University of Medical Sciences, Khoramabad, Iran
| | | | - Ahmad Mohajerian
- Department of Emergency Medicine, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | | | - Somayeh Zamanifard
- Department of Cardiology, School of Medicine, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | | | - Mohammad Sadegh Keshmiri
- Lung Transplant Research Center, National Research Institute of Tuberculosis and Lung Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Farahnaz Nikdoust
- Department of Cardiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, IR, Iran.
| |
Collapse
|
13
|
Jin Y, Lai Y, Hoshyar AN, Innab N, Shutaywi M, Deebani W, Swathi A. An ideally designed deep trust network model for heart disease prediction based on seagull optimization and Ruzzo Tompa algorithm. Sci Rep 2025; 15:6035. [PMID: 39971944 PMCID: PMC11839990 DOI: 10.1038/s41598-025-89348-8] [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: 03/30/2024] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
Diet, stress, genetics, and a sedentary lifestyle may all contribute to heart disease rates. Although recent studies propose comprehensive automated diagnostic systems, these systems tend to focus on one aspect, such as feature selection, prioritization, or predictive accuracy. A more complete approach that considers all of these factors can improve the efficiency of a cardiac prediction system. This study uses an appropriate strategy to overcome potential network design problems, design challenges, overfitting, and lack of robustness that can interfere with system performance. The research introduces an ideally designed deep trust network called ID-DTN to improve system performance. The Ruzzo-Tompa method is used to eliminate noncontributory features. The Seagull Optimization Algorithm (SOA) is introduced to optimize the trust depth network to achieve optimal network design. The study scrutinizes the deep trust network (ID-DTN) and the restricted Boltzmann machine (RBM) and sheds light on the system's operation. This proposal can optimize both network architecture and feature selection, which is the main novelty. The proposed method is analyzed using the below-mentioned metrics: Matthew's correlation coefficient, F1 score, accuracy, sensitivity, specificity, and accuracy. ID-DTN performs well compared to other state-of-the-art methods. The validation results confirm that the proposed method improves the prediction accuracy to 97.11% and provides reliable recommendations for patients with cardiovascular disease.
Collapse
Affiliation(s)
- Yuan Jin
- Department of Cardiovascular Medicine, The Fifth People's Hospital of Ganzhou, Ganzhou, 341000, Jiangxi, China.
- Department of Internal Medicine, Xingguo County People's Hospital, Ganzhou, 341000, Jiangxi, China.
| | - Yunliang Lai
- Department of Cardiovascular Medicine, The Fifth People's Hospital of Ganzhou, Ganzhou, 341000, Jiangxi, China
- Department of Internal Medicine, Xingguo County People's Hospital, Ganzhou, 341000, Jiangxi, China
| | - Azadeh Noori Hoshyar
- Institute of Innovation, Science and Sustainability, Federation University Australia, Berwick, VIC, 3806, Australia
| | - Nisreen Innab
- Department of Computer Science and Information Systems, College of Applied Sciences, AlMaarefa University, 13713, Diriyah, Riyadh, Saudi Arabia
| | - Meshal Shutaywi
- Department of Mathematics, College of Science and Arts, King Abdul Aziz University, Rabigh, Saudi Arabia
| | - Wejdan Deebani
- Department of Mathematics, College of Science and Arts, King Abdul Aziz University, Rabigh, Saudi Arabia
| | - A Swathi
- Department of Mathematics, Faculty of Science, University of Hradec Kralove, 50003, Hradec Kralove, Czech Republic
| |
Collapse
|
14
|
Imran M, Altamimi ASA, Afzal M, Babu MA, Goyal K, Ballal S, Sharma P, Alanazi FJ, Alruwaili AN, Aldhafeeri NA, Ali H. Targeting senescence and GATA4 in age-related cardiovascular disease: a comprehensive approach. Biogerontology 2025; 26:45. [PMID: 39831933 DOI: 10.1007/s10522-025-10189-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: 10/31/2024] [Accepted: 01/03/2025] [Indexed: 01/22/2025]
Abstract
The growing prevalence of age-related cardiovascular diseases (CVDs) poses significant health challenges, necessitating the formulation of novel treatment approaches. GATA4, a vital transcription factor identified for modulating cardiovascular biology and cellular senescence, is recognized for its critical involvement in CVD pathogenesis. This review collected relevant studies from PubMed, Google Scholar, and Science Direct using search terms like 'GATA4,' 'cellular senescence,' 'coronary artery diseases,' 'hypertension,' 'heart failure,' 'arrhythmias,' 'congenital heart diseases,' 'cardiomyopathy,' and 'cardiovascular disease.' Additionally, studies investigating the molecular mechanisms underlying GATA4-mediated regulation of GATA4 and senescence in CVDs were analyzed to provide comprehensive insights into this critical aspect of potential treatment targeting. Dysregulation of GATA4 is involved in a variety of CVDs, as demonstrated by both experimental and clinical research, comprising CAD, hypertension, congenital heart diseases, cardiomyopathy, arrhythmias, and cardiac insufficiency. Furthermore, cellular senescence enhances the advancement of age-related CVDs. These observations suggested that therapies targeting GATA4, senescence pathways, or both as necessary may be an effective intervention in CVD progression and prognosis. Addressing age-related CVDs by targeting GATA4 and senescence is a broad mechanism approach. It implies further investigation of the molecular nature of these processes and elaboration of an effective therapeutic strategy. This review highlights the importance of GATA4 and senescence in CVD pathogenesis, emphasizing their potential as therapeutic targets for age-related CVDs.
Collapse
Affiliation(s)
- Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, Rafha, 91911, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, Saudi Arabia.
| | - Abdulmalik S A Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam bin Abdulaziz University, Alkharj 11942, Saudi Arabia
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, 281406, UP, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India
| | - Fadiyah Jadid Alanazi
- Center for Health Research, Northern Border University, Arar, Saudi Arabia
- Public Health Nursing Department, College of Nursing, Northern Border University, Arar, Saudi Arabia
| | - Abeer Nuwayfi Alruwaili
- Department of Nursing Administration and Education, College of Nursing, Jouf University, Sakaka 72388, Saudi Arabia
| | - Nouf Afit Aldhafeeri
- College of Nursing, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center, Riyadh, Saudi Arabia
| | - Haider Ali
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Kyrgyz State Medical College, Bishkek, Kyrgyzstan
| |
Collapse
|
15
|
Amin A, Nikdoust F, Khorram S, Marashi SM, Ghanavati P, Ameri F, Akbarzadeh A, Hasanvand A, Khodakarim N. Dengue virus infection: how platelet-leukocyte crosstalk shapes thrombotic events and inflammation. Mol Biol Rep 2025; 52:119. [PMID: 39804486 DOI: 10.1007/s11033-025-10222-x] [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: 11/02/2024] [Accepted: 01/02/2025] [Indexed: 05/02/2025]
Abstract
Dengue virus (DENV) poses a considerable threat to public health on a global scale, since about two-thirds of the world's population is currently at risk of contracting this arbovirus. Being transmitted by mosquitoes, this virus is associated with a range of illnesses and a small percentage of infected individuals might suffer from severe vascular leakage. This leakage leads to hypovolemic shock syndrome, generally known as dengue shock syndrome, organ failure, and bleeding complications. The severe form of this disease is believed to be, at least partially, associated with inflammatory and thrombotic states. These issues are significantly affected by the activation of platelets and leukocytes, as well as their interactions, which may influence its prognosis. The platelets present in a thrombus are able to attract leukocytes to the site of injury. The intricate process leads to the significant accumulation, activation, and migration of leukocytes, thereby promoting thrombotic events and triggering inflammatory responses. The occurrence of these events, combined with the direct viral infection of endothelial cells, leads to vascular endothelialitis, the disruption of cellular membranes, and the subsequent release of DAMPs. As a result, considerable damage occurs in the endothelium, which activates neutrophils and platelets; thisleads to their interaction and initiates the process of Netosis. Collectively, these processes exacerbate inflammatory and thrombotic conditions. In this respect, current research has focused on understanding whether effective anti-inflammatory protocols can prevent thrombotic events or, conversely, if efficient anticoagulant regimens may lead to a reduction in cytokine storms and tissue damage. This review article aims to illuminate the platelet leukocyte crosstalk, detailing the mechanisms through which platelets may play a role in the pathogenesis of DENV. The research outputs are particularly important in severe cases, in which case their interactions with leukocytes can exacerbate both inflammation and thrombosis in a mutually reinforcing manner.
Collapse
Affiliation(s)
- Arash Amin
- Department of Cardiology, School of Medicine, Shahid Madani Hospital, Lorestan University of Medical Sciences, Lorestan, Iran
| | - Farahnaz Nikdoust
- Department of Cardiology, Shariati Hospital, Tehran University of Medical Sciences, Tehran, IR, Iran
| | | | | | - Pedram Ghanavati
- Department of Neurosurgery, Firouzgar Hospital, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Farshid Ameri
- Department of Infection, School of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Ashkan Akbarzadeh
- Department of Internal Medicine, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Afshin Hasanvand
- Department of General Surgery, Lorestan University of Medical Science, Khorramabad, Iran
- Student Research Committe, Lorestan University of Medical Science, Khorramabad, Iran
| | - Nastaran Khodakarim
- Department of Internal Medicine, School of Medicine, Hazrat-e Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
- Department of medical oncology and hematology, Hazrat Rasoul Hospital, School of Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Pei J, Xu X, Zhao LC. Hemodynamic evaluation of endovascular techniques of stenting and coiling for the treatment of internal carotid artery aneurysm: a computational study. Sci Rep 2024; 14:29117. [PMID: 39582080 PMCID: PMC11586413 DOI: 10.1038/s41598-024-80311-7] [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: 09/26/2024] [Accepted: 11/18/2024] [Indexed: 11/26/2024] Open
Abstract
The selection optimum endovascular method for the treatment of different cerebral aneurysms has been the main challenge for surgeons. In the present article, the computational technique is used for the hemodynamic evaluation of two main endovascular techniques of stent and coiling for the reduction of the hemorrhage risk of ICA aneurysms. Comprehensive analyses of the blood flow are performed to detect potential regions with high risk at critical stages of the cardiac cycle. Two coiling porosity conditions and stent deformation have been investigated via computational study to reveal the main change related to hemodynamics when these techniques are implemented. The hemodynamic results of this study show that the endovascular technique is more efficient in small aneurysms rather than giant ones. Meanwhile, the stent treatment of the giant saccular aneurysm is effective when the parent vessel of this type of aneurysm is fully aligned and limited blood flow enters the sac area.
Collapse
Affiliation(s)
- Jian Pei
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, 063000, HeBei, China
| | - Xiang Xu
- Department of Neurosurgery, Tangshan Gongren Hospital, Tangshan, 063000, HeBei, China.
| | - Li-Cai Zhao
- The Third Engineering Co., Ltd., China Railway 19th Bureau Group Co., Ltd, Shenyang, China
- School of Engineering, Islamic azad university, Tehran, Iran
| |
Collapse
|
17
|
Jolibois J, Domingues A, El Hamaoui D, Awaida R, Berger-de-Gaillardo M, Guérin D, Smadja DM, Marquet-DeRougé P, Margaill I, Rossi E, Nivet-Antoine V. Targeting TXNIP in endothelial progenitors mitigates IL-8-induced neutrophil recruitment under metabolic stress. Stem Cell Res Ther 2024; 15:225. [PMID: 39075518 PMCID: PMC11287885 DOI: 10.1186/s13287-024-03850-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/12/2024] [Indexed: 07/31/2024] Open
Abstract
BACKGROUND This study explores the potential role of Thioredoxin-interacting protein (TXNIP) silencing in endothelial colony-forming cells (ECFCs) within the scope of age-related comorbidities and impaired vascular repair. We aim to elucidate the effects of TXNIP silencing on vasculogenic properties, paracrine secretion, and neutrophil recruitment under conditions of metabolic stress. METHODS ECFCs, isolated from human blood cord, were transfected with TXNIP siRNA and exposed to a high glucose and β-hydroxybutyrate (BHB) medium to simulate metabolic stress. We evaluated the effects of TXNIP silencing on ECFCs' functional and secretory responses under these conditions. Assessments included analyses of gene and protein expression profiles, vasculogenic properties, cytokine secretion and neutrophil recruitment both in vitro and in vivo. The in vivo effects were examined using a murine model of hindlimb ischemia to observe the physiological relevance of TXNIP modulation under metabolic disorders. RESULTS TXNIP silencing did not mitigate the adverse effects on cell recruitment, vasculogenic properties, or senescence induced by metabolic stress in ECFCs. However, it significantly reduced IL-8 secretion and consequent neutrophil recruitment under these conditions. In a mouse model of hindlimb ischemia, endothelial deletion of TXNIP reduced MIP-2 secretion and prevented increased neutrophil recruitment induced by age-related comorbidities. CONCLUSIONS Our findings suggest that targeting TXNIP in ECFCs may alleviate ischemic complications exacerbated by metabolic stress, offering potential clinical benefits for patients suffering from age-related comorbidities.
Collapse
Affiliation(s)
- Julia Jolibois
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
| | - Alison Domingues
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France.
| | - Divina El Hamaoui
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
| | - Raphaël Awaida
- Laboratoire de Biochimie générale, AP-HP, Hôpital Necker Enfants Malades, Paris, F-75015, France
| | | | - Daniel Guérin
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
| | - David M Smadja
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
- Laboratoire d'Hématologie, AP-HP, Hôpital Européen Georges Pompidou, Paris, F-75015, France
| | - Perrine Marquet-DeRougé
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
| | - Isabelle Margaill
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
| | - Elisa Rossi
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
| | - Valérie Nivet-Antoine
- Université Paris Cité, INSERM, Innovations thérapeutiques en hémostase, Paris, F-75006, France
- Laboratoire de Biochimie générale, AP-HP, Hôpital Necker Enfants Malades, Paris, F-75015, France
| |
Collapse
|