1
|
Shaposhnikov M, Thakar J, Berk BC. Value of Bioinformatics Models for Predicting Translational Control of Angiogenesis. Circ Res 2025; 136:1147-1165. [PMID: 40339045 DOI: 10.1161/circresaha.125.325438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Angiogenesis, the formation of new blood vessels, is a fundamental biological process with implications for both physiological functions and pathological conditions. While the transcriptional regulation of angiogenesis, mediated by factors such as HIF-1α (hypoxia-inducible factor 1-alpha) and VEGF (vascular endothelial growth factor), is well-characterized, the translational regulation of this process remains underexplored. Bioinformatics has emerged as an indispensable tool for advancing our understanding of translational regulation, offering predictive models that leverage large data sets to guide research and optimize experimental approaches. However, a significant gap persists between bioinformatics experts and other researchers, limiting the accessibility and utility of these tools in the broader scientific community. To address this divide, user-friendly bioinformatics platforms are being developed to democratize access to predictive analytics and empower researchers across disciplines. Translational control, compared with transcriptional control, offers a more energy-efficient mechanism that facilitates rapid cellular responses to environmental changes. Furthermore, transcriptional regulators themselves are often subject to translational control, emphasizing the interconnected nature of these regulatory layers. Investigating translational regulation requires advanced, accessible bioinformatics tools to analyze RNA structures, interacting micro-RNAs, long noncoding RNAs, and RBPs (RNA-binding proteins). Predictive platforms such as RNA structure, human internal ribosome entry site Atlas, and RBPSuite enable the study of RNA motifs and RNA-protein interactions, shedding light on these critical regulatory mechanisms. This review highlights the transformative role of bioinformatics using widely accessible user-friendly tools with a Web-browser interface to elucidate translational regulation in angiogenesis. The bioinformatics tools discussed extend beyond angiogenesis, with applications in diverse fields, including clinical care. By integrating predictive models and experimental insights, researchers can streamline hypothesis generation, reduce experimental costs, and find novel translational regulators. By bridging the bioinformatics knowledge gap, this review aims to empower researchers worldwide to adopt bioinformatics tools in their work, fostering innovation and accelerating scientific discovery.
Collapse
Affiliation(s)
- Michal Shaposhnikov
- Department of Cellular and Molecular Pharmacology and Physiology (M.S., B.C.B.), University of Rochester School of Medicine and Dentistry, NY
- Department of Medicine, Aab Cardiovascular Research Institute (M.S., B.C.B.), University of Rochester School of Medicine and Dentistry, NY
| | - Juilee Thakar
- Department of Microbiology and Immunology (J.T.), University of Rochester School of Medicine and Dentistry, NY
- Department of Biomedical Genetics, Biostatistics and Computational Biology (J.T.), University of Rochester School of Medicine and Dentistry, NY
| | - Bradford C Berk
- Department of Cellular and Molecular Pharmacology and Physiology (M.S., B.C.B.), University of Rochester School of Medicine and Dentistry, NY
- Department of Medicine, Aab Cardiovascular Research Institute (M.S., B.C.B.), University of Rochester School of Medicine and Dentistry, NY
| |
Collapse
|
2
|
Aryal A, Harmon AC, Noël A, Yu Q, Varner KJ, Dugas TR. AhR Activation at the Air-Blood Barrier Alters Systemic microRNA Release After Inhalation of Particulate Matter Containing Environmentally Persistent Free Radicals. Cardiovasc Toxicol 2025; 25:651-665. [PMID: 40214911 PMCID: PMC12018632 DOI: 10.1007/s12012-025-09989-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
Particulate matter containing environmentally persistent free radicals (EPFRs) is formed when organic pollutants are incompletely burned and adsorb to the surface of particles containing redox-active metals. Our prior studies showed that in mice, EPFR inhalation impaired vascular relaxation in a dose- and endothelium-dependent manner. We also observed that activation of the aryl hydrocarbon receptor (AhR) in the alveolar type-II (AT-II) cells that form the air-blood interface stimulates the release of systemic factors that promote endothelial dysfunction in vessels peripheral to the lung. AhR is a recognized regulator of microRNA (miRNA) biogenesis, and miRNA control diverse signaling pathways. We thus hypothesized that systemic EPFR-induced vascular endothelial dysfunction is initiated via AhR activation in AT-II cells, resulting in a systemic release of miRNA. Using a combustion reactor, we generated EPFR of two free radical concentrations-EPFRlo (1016-17 radicals/g particles) and EPFR (1018-19 radicals/g)-and exposed mice by inhalation. EFPR inhalation resulted in changes in a distinct array of miRNA in the plasma, and these miRNAs are linked to multiple systemic effects, including cardiovascular diseases and dysregulation of cellular and molecular pathways associated with cardiovascular dysfunction. We identified 17 miRNA in plasma that were altered dependent upon both AhR activation in AT-II cells and ~ 280 ug/m3 EPFR exposure. Using Ingenuity Pathway Analysis, we found that 5 of these miRNAs have roles in modulating endothelin-1 and endothelial nitric oxide signaling, known regulators of endothelial function. Furthermore, EPFR exposure reduced the expression of lung adherens and gap junction proteins in control mice but not AT-II-AhR deficient mice, and reductions in barrier function may facilitate miRNA release from the lungs. In summary, our findings support that miRNA may be systemic mediators promoting endothelial dysfunction mediated via EPFR-induced AhR activation at the air-blood interface.
Collapse
Affiliation(s)
- Ankit Aryal
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Ashlyn C Harmon
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA
| | - Qingzhao Yu
- Biostatistics, School of Public Health, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Kurt J Varner
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, 70112, USA
| | - Tammy R Dugas
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
3
|
Dykxhoorn DM, Da Fonseca Ferreira A, Gomez K, Shi J, Zhu S, Zhang L, Wang H, Wei J, Zhang Q, Macon CJ, Hare JM, Marzouka GR, Wang L, Dong C. MicroRNA-29c-3p and -126a Contribute to the Decreased Angiogenic Potential of Aging Endothelial Progenitor Cells. Int J Mol Sci 2025; 26:4259. [PMID: 40362495 PMCID: PMC12072698 DOI: 10.3390/ijms26094259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/22/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
EPCs play important roles in the maintenance of vascular repair and health. Aging is associated with both reduced numbers and functional impairment of EPCs, leading to diminished angiogenic capacity, impaired cardiac repair, and increased risk for cardiovascular disease (CVD). The molecular mechanisms that govern EPC function in cardiovascular health are not fully understood, but there is increasing evidence that microRNAs (miRNAs) play key roles in modulating EPC functionality, endothelial homeostasis, and vascular repair. We aimed to determine how aging alters endothelial progenitor (EPC) health and functionality by altering key miRNA-mRNA pathways. To identify key miRNA-mRNA pathways contributing to diminished EPC functionality associated with aging, microRNA and mRNA profiling were conducted in EPCs from young and aged C57BL/6 mice. We identified a complex aging-associated regulatory network involving two miRNAs-miR-29c-3p and -126a-that acted in tandem to impair vascular endothelial growth factor signaling through targeting Klf2 and Spred1, respectively. The modulation of components of the miR-29c-3p-Klf2-miR-126a-Spred-1-Vegf signaling pathway altered EPC self-renewal capacity, vascular tube formation, and migration in vitro, as well as cardiac repair in vivo. The miR-29c-3p-Klf2-miR-126a-Spred1-Vegf signaling axis plays a critical role in regulating the aging-associated deficits in EPC-mediated vascular repair and CVD risk.
Collapse
Affiliation(s)
- Derek M. Dykxhoorn
- John T. Macdonald Foundation Department of Human Genetics and, the John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (D.M.D.)
| | - Andrea Da Fonseca Ferreira
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Karenn Gomez
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Jianjun Shi
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Shoukang Zhu
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Lukun Zhang
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Huilan Wang
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Jianqin Wei
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Qianhuan Zhang
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Conrad J. Macon
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
| | - Joshua M. Hare
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - George R. Marzouka
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Liyong Wang
- John T. Macdonald Foundation Department of Human Genetics and, the John P. Hussman Institute for Human Genomics, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (D.M.D.)
| | - Chunming Dong
- Interdisciplinary Stem Cell Institute, University of Miami Miller School of Medicine, Miami, FL 33136, USA; (A.D.F.F.)
- Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| |
Collapse
|
4
|
Vrapcea A, Pisoschi CG, Ciupeanu-Calugaru ED, Traşcă ET, Tutunaru CV, Rădulescu PM, Rădulescu D. Inflammatory Signatures and Biological Markers in Platelet-Rich Plasma Therapy for Hair Regrowth: A Comprehensive Narrative Analysis. Diagnostics (Basel) 2025; 15:1123. [PMID: 40361941 PMCID: PMC12071426 DOI: 10.3390/diagnostics15091123] [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: 01/20/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Context: Hair loss (alopecia) presents both aesthetic and psychological challenges, significantly impacting quality of life. Platelet-rich plasma (PRP) therapy has gained prominence due to its ability to deliver growth factors and modulate local inflammation. However, uncertainties remain regarding the mechanisms through which systemic inflammation, oxidative stress, and coagulation factors influence PRP's efficacy. Objectives: This narrative review explores the impact of inflammatory biomarkers (e.g., NLR, PLR, IL-6, TNF-α) and growth factors (VEGF, TGF-β, FGF) on hair regeneration in PRP therapy. It discusses how oxidative stress and vitamin status (B12, D, folate) correlate with therapeutic success. Additionally, it examines the PRP preparation protocols and combined approaches (microneedling, minoxidil, LLLT) that may amplify clinical responses. Results: The synthesized data highlight that elevated systemic inflammation (increased NLR/PLR values) can limit PRP's effectiveness, while the regulation of inflammation and optimization of antioxidant status can enhance hair density and thickness. Integrating vitamins and an anti-inflammatory diet into the therapeutic protocol is associated with more stable hair growth and reduced adverse reactions. The variability in PRP's preparation and activation methods remains a major obstacle, underscoring the need for standardization. Conclusions: Integrating inflammatory biomarkers with oxidative stress indicators provides fresh insights for tailoring PRP therapies in alopecia. Multimodal treatment strategies combined with collaborative multicenter studies-in which biological markers are embedded within rigorous protocols-could establish standardized methodologies and significantly enhance the treatment success.
Collapse
Affiliation(s)
- Adelina Vrapcea
- Doctoral School, University of Medicine and Pharmacy of Craiova, 200585 Craiova, Romania;
| | - Cătălina Gabriela Pisoschi
- Biochemistry Department, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 200585 Craiova, Romania;
| | | | - Emil-Tiberius Traşcă
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| | | | | | - Dumitru Rădulescu
- Department of Surgery, The Military Emergency Clinical Hospital ‘Dr. Stefan Odobleja’, University of Medicine and Pharmacy of Craiova, 200349 Craiova, Romania;
| |
Collapse
|
5
|
Lin K, Luo X, Du C, Zuo C, Li Z, Zhang G, Li C, Zhu L. ANRIL modulates endothelial senescence and angiogenesis through SASP-driven miR146a regulation in age-related vascular dysfunction. Mech Ageing Dev 2025; 225:112058. [PMID: 40222710 DOI: 10.1016/j.mad.2025.112058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Revised: 03/25/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Vascular aging, marked by endothelial cell (EC) dysfunction and compromised angiogenesis, is a central driver of age-related ischemic diseases. Although lncRNAs have emerged as pivotal regulators of endothelial function, their specific roles in endothelial aging remain enigmatic. In this study, we identify the lncRNA ANRIL as a crucial modulator of endothelial dysfunction during aging. By analyzing publicly available lncRNA sequencing datasets comparing young and old ECs, we pinpointed ANRIL and validated its role through a replicative senescence model in human umbilical vein ECs (HUVECs) and FACS sorting of skeletal muscle ECs from aged mice. While ANRIL showed minimal direct effects on angiogenesis, functional assays and transcriptomic analysis revealed its profound impact on the senescence-associated secretory phenotype (SASP). Remarkably, ANRIL regulates the expression of miR146a in ECs, which is transferred to macrophages, where it inhibits VEGF secretion and disrupts endothelial neovascularization. In vivo, ANRIL downregulation in a murine hindlimb ischemia model significantly enhanced neovascularization and restored blood flow, revealing its therapeutic potential for ischemic diseases. These findings position ANRIL as a novel, potent regulator of endothelial senescence, offering new insights into the molecular basis of vascular aging and suggesting ANRIL as a promising therapeutic target to mitigate age-related vascular dysfunction.
Collapse
Affiliation(s)
- Kechuan Lin
- Department of geriatric, Coronary Circulation Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China; The Third Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Xin Luo
- Department of geriatric, Coronary Circulation Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Can Du
- Department of geriatric, Coronary Circulation Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Chenzhe Zuo
- Department of geriatric, Coronary Circulation Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Zhenyu Li
- Department of geriatric, Coronary Circulation Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Guogang Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China; The Third Xiangya Hospital of Central South University, Changsha, Hunan 410008, China
| | - Chuanchang Li
- Department of geriatric, Coronary Circulation Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China
| | - Lingping Zhu
- Department of geriatric, Coronary Circulation Center, Xiangya Hospital of Central South University, Changsha, Hunan 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Changsha, Hunan 410008, China.
| |
Collapse
|
6
|
Bigagli E, Spataro E, Pasquini L, Cinci L, D'Ambrosio M, De Blasi C, Bartolini C, Petraglia F, Luceri C. Vaginal miR-210-3p as a potential biomarker for pregnancies complicated by early fetal growth restriction: A proof-of-concept case-control study. Placenta 2025; 163:8-15. [PMID: 40023010 DOI: 10.1016/j.placenta.2025.02.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 03/04/2025]
Abstract
INTRODUCTION Fetal growth restriction (FGR) is associated with increased risk of neonatal morbidity and mortality or long-term adverse outcomes. We investigated the ability of hypoxia and angiogenesis-related miR-210-3p and miR-126-5p to identify early FGR cases and their correlations with neonatal outcomes. METHODS Twenty-nine women with pregnancies complicated by early FGR diagnosis and 25 controls matched for gestational age (GA) were enrolled and their vaginal fluid (VF) and plasma were collected. MiR-210-3p and miR-126-5p were measured by RT-qPCR and their targets were identified by in-silico analysis limited only to those already experimentally validated in other contexts. RESULTS Overall, VF levels of miR-210-3p were lower in early FGR cases compared to controls (p < 0.05). miR-210-3p was lower in severe cases and in women who later developed preeclampsia (p < 0.05). VF miR-210-3p levels correlated with lower birth weight, premature birth and severe complications at birth (p < 0.05). miR-210-3p was not detected in plasma and no correlations were observed between miR-126-5p and FGR or neonatal outcomes. In silico analyses identified HIF-1α, HIF-3α, BDNF, IGFBP3, RAD52 and TWIST-1 as experimentally validated targets of miR-210-3p. Among the predicted biological pathways controlled by miR-210-3p, we found hypoxia-responsive signaling such as autophagy, oxidative stress and metabolic pathways. DISCUSSION Although validation is needed, these findings suggest that VF levels of miR-210-3p may potentially serve as biomarker for the diagnosis of early FGR; future mechanistic studies are also advisable to investigate whether pharmacological strategies based on miR-210-3p, or its downstream targets may be useful for FGR.
Collapse
Affiliation(s)
- Elisabetta Bigagli
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| | - Elisa Spataro
- Department of Experimental and Clinical Biomedical Sciences, Obstetrics and Gynecology, University of Florence, Florence, Italy; Obstetrics and Gynecology, Department of Maternal and Child Health, University of Florence, Careggi University Hospital, Florence, Italy
| | - Lucia Pasquini
- Fetal Medicine Unit, Department for Woman and Child Health, Careggi University Hospital, Florence, Italy
| | - Lorenzo Cinci
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Mario D'Ambrosio
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy
| | - Chiara De Blasi
- Department of Experimental and Clinical Biomedical Sciences, Obstetrics and Gynecology, University of Florence, Florence, Italy; Obstetrics and Gynecology, Department of Maternal and Child Health, University of Florence, Careggi University Hospital, Florence, Italy
| | - Chiara Bartolini
- Department of Experimental and Clinical Biomedical Sciences, Obstetrics and Gynecology, University of Florence, Florence, Italy; Obstetrics and Gynecology, Department of Maternal and Child Health, University of Florence, Careggi University Hospital, Florence, Italy
| | - Felice Petraglia
- Department of Experimental and Clinical Biomedical Sciences, Obstetrics and Gynecology, University of Florence, Florence, Italy; Obstetrics and Gynecology, Department of Maternal and Child Health, University of Florence, Careggi University Hospital, Florence, Italy
| | - Cristina Luceri
- Department of Neuroscience, Psychology, Drug Research and Child Health (NEUROFARBA), Section of Pharmacology and Toxicology, University of Florence, Florence, Italy.
| |
Collapse
|
7
|
Guo B, Gu J, Zhuang T, Zhang J, Fan C, Li Y, Zhao M, Chen R, Wang R, Kong Y, Xu S, Gao W, Liang L, Yu H, Han T. MicroRNA-126: From biology to therapeutics. Biomed Pharmacother 2025; 185:117953. [PMID: 40036996 DOI: 10.1016/j.biopha.2025.117953] [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: 12/30/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
MicroRNA-126 (miR-126) has emerged as one of the most extensively studied microRNAs in the context of human diseases, particularly in vascular disorders and cancer. Its high degree of conservation across vertebrates underscores its evolutionary significance and essential functional roles. Extensive research has been devoted to elucidating the molecular mechanisms through which miR-126 modulates key physiological and pathological processes, including angiogenesis, immune response, inflammation, tumor growth, and metastasis. Furthermore, miR-126 plays a causal role in the pathogenesis of various diseases, serving as potential biomarkers for disease prediction, diagnosis, prognosis and drug response, as well as a promising therapeutic target. In this review, we synthesize findings from 283 articles, focusing on the roles of miR-126 in critical biological processes such as cell development, survival, cycle regulation, proliferation, migration, invasion, communication, and metabolism. Additionally, miR-126 represents a promising candidate for miRNA-based therapeutic strategies. A comprehensive understanding and evaluation of miR-126 are crucial for advancing its clinical applications and therapeutic potential.
Collapse
Affiliation(s)
- Bei Guo
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tongtian Zhuang
- Department of Dermatology, Air Force Hospital of Northern Theater Command, Shenyang, China
| | - Jingbin Zhang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chunyang Fan
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yiyao Li
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Mengdi Zhao
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Ruoran Chen
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Rui Wang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuan Kong
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Shuang Xu
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Wei Gao
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Linlang Liang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Hao Yu
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
8
|
Villagrán-Silva F, Loren P, Sandoval C, Lanas F, Salazar LA. Circulating microRNAs as Potential Biomarkers of Overweight and Obesity in Adults: A Narrative Review. Genes (Basel) 2025; 16:349. [PMID: 40149500 PMCID: PMC11942292 DOI: 10.3390/genes16030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
In an obesogenic environment, such as the one we have been experiencing in recent decades, epigenetics provides answers to the relationship between hereditary and environmentally acquired patterns that have significantly contributed to the global rise in obesity prevalence. MicroRNA (miRNA) constitutes a diminutive non-coding small RNA molecule, 20 to 24 nucleotides in length, that functions as a regulator of gene regulation at the post-translational level. Circulating miRNAs (c-miRNAs) have been detected in multiple body fluids, including blood, plasma, serum, saliva, milk from breastfeeding mothers, and urine. These molecules hold significant therapeutic value and serve as extracellular biomarkers in metabolic diseases. They aid in the diagnosis and tracking of therapy responses, as well as dietary and physical habit modifications. Researchers have studied c-miRNAs as potential biomarkers for diagnosing and characterizing systemic diseases in people of all ages and backgrounds since then. These conditions encompass dyslipidemia, type 2 diabetes mellitus (T2DM), cardiovascular risk, metabolic syndrome, cardiovascular diseases, and obesity. This review therefore analyzes the usefulness of c-miRNAs as therapeutic markers over the past decades. It also provides an update on c-miRNAs associated with general obesity and overweight, as well as with the most prevalent pathologies in the adult population. It also examines the effect of different nutritional approaches and physical activity regarding the activity of miRNAs in circulation in adults with overweight or general obesity. All of this is done with the aim of evaluating their potential use as biomarkers in various research contexts related to overweight and obesity in adults.
Collapse
Affiliation(s)
- Francisca Villagrán-Silva
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile;
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Fernando Lanas
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| |
Collapse
|
9
|
Hajsadeghi S, Iranpour A, Mirshafiee S, Nekouian R, Mollababaei M, Motevalli H, Yasin Ahmadi SA, Dakkali MS. Impact of smoking on microRNAs in significant coronary artery disease. ROMANIAN JOURNAL OF INTERNAL MEDICINE = REVUE ROUMAINE DE MEDECINE INTERNE 2025; 63:49-59. [PMID: 39543851 DOI: 10.2478/rjim-2024-0031] [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: 09/05/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Given the importance of coronary artery disease (CAD) and the range of cardiovascular disease phenotypes in smokers, as well as the potential genetic and epigenetic factors, we were motivated to explore the impact of smoking on some selected microRNAs associated with significant CAD. METHODS A total of 60 individuals were selected in four groups including non-smoker without significant CAD (S-A-), non-smokers with significant CAD (S-A+), smokers without significant CAD (S+A-) and smokers with significant CAD (S+A+). Micro-RNA expression was investigated using real-time PCR. General linear model was used to calculate fold change (FC) considering S-A- as the reference group. RESULTS For mir-34a, down-regulation was observed in S+A- (FC =0.13, P =0.007) and S+A+ (FC =0.23, P =0.036) groups. For mir-126-3p, down-regulation was observed in S-A+ group (FC =0.05, P =0.024). For mir-199, up-regulation was observed for S+A- group (FC =9.38, P =0.007). The only significant interaction between pack-years of smoking and number of significantly narrowed vessels (≥75% stenosis) was for mir-199 which was in favor of down-regulation (P =0.006), while the main effects were in favor of up-regulation (P <0.05). CONCLUSION Mir-34a expression may be affected by smoking, whereas mir-126-3p expression may be affected by atherosclerosis, the most common reason of CAD. The significant down-regulation of mir-199 for the interaction of smoking dose and severity of CAD was a notable finding showing the harmful consequence of this interaction. Further studies are needed for this micro-RNA.
Collapse
Affiliation(s)
- Shokoufeh Hajsadeghi
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Aida Iranpour
- Research Center for Prevention of Cardiovascular Disease, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Shayan Mirshafiee
- Department of Cardiology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Nekouian
- Department of Medical Biotechnology, School of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Mollababaei
- Pediatric Growth and Development Research Center, Institute of Endocrinology and Metabolism, Iran University of Medical Sciences, Tehran, Iran
| | - Hamed Motevalli
- Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyyed Amir Yasin Ahmadi
- Preventive Medicine and Public Health Research Center, Psychosocial Health Research Institute, Iran University of Medical Sciences, Tehran, Iran
| | | |
Collapse
|
10
|
Li X, Hallajzadeh J. Circulating microRNAs and physical activity: Impact in diabetes. Clin Chim Acta 2025; 569:120178. [PMID: 39900127 DOI: 10.1016/j.cca.2025.120178] [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: 12/26/2024] [Revised: 01/28/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
The term "ci-miRNAs," or "circulating microRNAs," refers to extracellular microRNAs (miRNAs) that exist outside of cells and can be detected in various bodily fluids, including blood, saliva, urine, and breast milk. These ci-miRNAs play a role in regulating gene expression and are mainly recognized for their functions beyond the cell, serving as signaling molecules in the blood. Researchers have thoroughly investigated the roles of these circulating miRNAs in various diseases. The capacity to detect and quantify ci-miRNAs in bodily fluids suggests their potential as biomarkers for monitoring several health conditions, including cancer, heart disease, brain disorders, and metabolic disorders, where fluctuations in miRNA levels may correlate with different physiological and pathological states. Current methods enable researchers to identify and measure miRNAs in these fluids, facilitating the exploration of their roles in health maintenance and disease resistance. Although research on ci-miRNAs is ongoing, recent studies focus on uncovering their significance, assessing their viability as biomarkers, and clarifying their functions. However, our understanding of how various types, intensities, and durations of exercise influence the levels of these miRNAs in the bloodstream is still limited. This section seeks to provide an overview of the changes in ci-miRNAs in response to exercise.
Collapse
Affiliation(s)
- Xiu Li
- Shanghai Minyuan College, Shanghai 201210, China.
| | - Jamal Hallajzadeh
- Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran.
| |
Collapse
|
11
|
Enteshari-Moghadam A, Fouladi N, Pordel S, Jeddi F, Asghariazar V, Eterafi M, Safarzadeh E. Evaluation of the miRNA-126 and VCAM-1 in scleroderma patients and its association with clinical characteristics. Am J Med Sci 2025; 369:339-345. [PMID: 39326739 DOI: 10.1016/j.amjms.2024.09.007] [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: 02/16/2024] [Revised: 08/30/2024] [Accepted: 09/19/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND Systemic sclerosis (SSc) has the highest level of mortality and disability among all rheumatological diseases. Being heterogenous leads to no predictable method for clinical courses. The aim of this study was to evaluate the levels of miRNA-126 and soluble VCAM-1 protein markers in patients with SSc, and to examine the assossiation of their levels with the severity of clinical and paraclinical parameters in patients with SSc. METHOD In current study tweny six patients with SSc along with twenty-three SSc-free controls were recruited. Enzyme-linked immunosorbent assay (ELISA) was performed to measure the VCAM-1 protein. MiRNA-126 amounts in serum were detected by quantitative real-time polymerase chain reaction (PCR). RESULT SSc patients' average age was 45.42 years and control group 49.85. The mean±SD for circulating miR-126 levels were significantly lower in SSc patients compared with healthy donors (p = 0.02), 0.48 ± 0.72 vs 1.11 ± 0.61 respectively. A significant difference was also observed in the serum level of miRNA-126 in SSc patients who suffer from pulmonary artery hypertension (P = 0.03) and pulmonary fibrosis (P = 0.04). In contrast, analysis of the serum VCAM-1 levels in the study groups uncovered a significant increase in SSc patients (5.92 ± 3.52 µg/ml) compared to control group (2.62 ± 1.2 µg/ml) (P value < 0.001). CONCLUSION Significant change in circulating levels of miR-126 and VCAM-1 in the SSc patients supporting its role in the pathogenesis of the disease. It could also proposed potential role as a predictor of pulmonary complications for miRNA-126.
Collapse
Affiliation(s)
- Afsaneh Enteshari-Moghadam
- Department of Internal Medicine, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Nasrin Fouladi
- School of Medicine and Allied Medical Sciences, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Shohreh Pordel
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Farhad Jeddi
- Department of Genetics and Pathology, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Vahid Asghariazar
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Majid Eterafi
- Students Research Committee, School of Medicine, Ardabil University of Medical Sciences, Ardabil, Iran
| | - Elham Safarzadeh
- Cancer Immunology and Immunotherapy Research Center, Ardabil University of Medical Sciences, Ardabil, Iran; Department of Microbiology, Parasitology and Immunology, Ardabil University of Medical Sciences, Ardabil, Iran.
| |
Collapse
|
12
|
Liu Z, Lu J, Sha W, Lei T. Comprehensive treatment of diabetic endothelial dysfunction based on pathophysiological mechanism. Front Med (Lausanne) 2025; 12:1509884. [PMID: 40093018 PMCID: PMC11906411 DOI: 10.3389/fmed.2025.1509884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/24/2025] [Indexed: 03/19/2025] Open
Abstract
Vascular endothelium is integral to the regulation of vascular homeostasis and maintenance of normal arterial function in healthy individuals. Endothelial dysfunction is a significant contributor to the advancement of atherosclerosis, which can precipitate cardiovascular complications. A notable correlation exists between diabetes and endothelial dysfunction, wherein chronic hyperglycemia and acute fluctuations in glucose levels exacerbate oxidative stress. This results in diminished nitric oxide synthesis and heightened production of endothelin-1, ultimately leading to endothelial impairment. In clinical settings, it is imperative to implement appropriate therapeutic strategies aimed at enhancing endothelial function to prevent and manage diabetes-associated vascular complications. Various antidiabetic agents, including insulin, GLP-1 receptor agonists, sulfonylureas, DPP-4 inhibitors, SGLT2 inhibitors, α-glucosidase inhibitors, thiazolidinediones (TZDs), and metformin, are effective in mitigating blood glucose variability and improving insulin sensitivity by lowering postprandial glucose levels. Additionally, traditional Chinese medicinal compounds, such as turmeric extract, resveratrol, matrine alkaloids, tanshinone, puerarin, tanshinol, paeonol, astragaloside, berberine, and quercetin, exhibit hypoglycemic properties and enhance vascular function through diverse mechanisms. Consequently, larger randomized controlled trials involving both pharmacological and herbal interventions are essential to elucidate their impact on endothelial dysfunction in patients with diabetes. This article aims to explore a comprehensive approach to the treatment of diabetic endothelial dysfunction based on an understanding of its pathophysiology.
Collapse
Affiliation(s)
- Zhao Liu
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
- Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Lu
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenjun Sha
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tao Lei
- Department of Endocrinology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
13
|
Wilson TG, Baghel M, Kaur N, Datta I, Loveless I, Potla P, Mendez D, Hansen L, Baker K, Lynch TS, Moutzouros V, Davis J, Ali SA. Circulating miR-126-3p is a mechanistic biomarker for knee osteoarthritis. Nat Commun 2025; 16:2021. [PMID: 40016267 PMCID: PMC11868599 DOI: 10.1038/s41467-025-57308-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 02/18/2025] [Indexed: 03/01/2025] Open
Abstract
Osteoarthritis is a major contributor to pain and disability worldwide, yet there are currently no validated soluble biomarkers or disease-modifying treatments. Given that microRNAs are promising mechanistic biomarkers that can be therapeutically targeted, in this study, we aimed to identify and prioritize reproducible circulating microRNAs associated with radiographic knee osteoarthritis. Across four independent cohorts, we find circulating miR-126-3p is elevated in knee osteoarthritis versus controls. Across six primary human knee osteoarthritis tissues, miR-126-3p is highest in subchondral bone, fat pad and synovium, and lowest in cartilage. Following both intravenous and intra-articular miR-126-3p mimic treatment in a surgical mouse model of knee osteoarthritis, we show reduced disease severity in males. In human knee osteoarthritis biospecimens, miR-126-3p mimic treatment reduces genes and markers associated with angiogenesis, as well as genes linked to osteogenesis, adipogenesis, and synovitis-processes secondary to angiogenesis. Our findings indicate that miR-126-3p is elevated in knee osteoarthritis and mitigates disease severity, supporting its potential as a biomarker and therapeutic target.
Collapse
Affiliation(s)
- Thomas G Wilson
- Bone and Joint Center, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Madhu Baghel
- Bone and Joint Center, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Navdeep Kaur
- Bone and Joint Center, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Indrani Datta
- Center for Bioinformatics, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Ian Loveless
- Center for Bioinformatics, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Pratibha Potla
- Schroeder Arthritis Institute, University Health Network, Toronto, ON, Canada
| | - Devin Mendez
- Bone and Joint Center, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - Logan Hansen
- Department of Orthopedic Surgery, Henry Ford Health, Detroit, MI, USA
| | - Kevin Baker
- Bone and Joint Center, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA
| | - T Sean Lynch
- Department of Orthopedic Surgery, Henry Ford Health, Detroit, MI, USA
| | | | - Jason Davis
- Department of Orthopedic Surgery, Henry Ford Health, Detroit, MI, USA
| | - Shabana Amanda Ali
- Bone and Joint Center, Henry Ford Health + Michigan State University Health Sciences, Detroit, MI, USA.
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
14
|
Al-Rawaf HA, Gabr SA, Alghadir T, Alghadir F, Iqbal A, Alghadir AH. Correlation between circulating microRNAs and vascular biomarkers in type 2 diabetes based upon physical activity: a biochemical analytic study. BMC Endocr Disord 2025; 25:55. [PMID: 40016689 PMCID: PMC11866858 DOI: 10.1186/s12902-025-01855-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/21/2025] [Indexed: 03/01/2025] Open
Abstract
BACKGROUND This research investigated how physical activity (PA) might impact the expression of several microRNAs, specifically miR-126, miR-146a, miR-34a, miR-124a, miR-155, and miR-221, in the blood of elderly individuals with type 2 diabetes (T2D). Additionally, the study examined the relationship between these microRNAs and markers of vascular endothelial dysfunction, including vascular endothelial growth factor (VEGF), apolipoprotein A-I (apoA-I), and apolipoprotein B (apoB), to assess their potential in the prevention, early detection, and treatment of diabetes. METHODS This correlational observational study involved 100 male participants, aged between 18 and 65 years, all of whom had been living with type 2 diabetes (T2D) for over six years. The participants were divided into three groups: inactive, moderate, and active, depending on their level of physical activity (PA). Real-time PCR and immunoassays were employed to measure the expression of selected miRNAs, as well as VEGF, apoA-I, apoB, and diabetic management indicators. PA levels were determined using ACTi graph GT1M accelerometer (model WAM 7164; Fort Walton Beach, FL) and energy expenditure was measured in the form of metabolic equivalent (MET) by indirect calorimetry method. RESULTS The expression levels of miR-146a, miR-34a, and miR-124a were significantly higher in patients with higher physical activity, while no such increase was observed for the other miRNAs in less active participants. Additionally, PA-active individuals showed a more pronounced decrease in fasting blood sugar (FBS), insulin resistance (IR), fasting insulin (FINS), HOMA-IR, HbA1c (%), and levels of VEGF, apoAI, apoB, and the apoB/apoA-I ratio. The alteration in miRNA expression was positively associated with physical activity, VEGF, apoAI, apoB, the apoB/apoA-I ratio, and diabetes-related metrics, while being inversely related to BMI. CONCLUSIONS In diabetic patients with higher physical activity levels, circulating miR-146a, miR-34a, and miR-124a showed elevated expression, accompanied by a notable decrease in vascular biomarkers, including apoAI, apoB, and the apoB/apoA-I ratio. The findings revealed a strong correlation between these vascular biomarkers and the physiological responses of miR-146a, miR-34a, and miR-124a, though larger studies are required to validate these results further. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Hadeel A Al-Rawaf
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, Riyadh, 11433, Saudi Arabia
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Sami A Gabr
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| | - Talal Alghadir
- College of Medicine, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Faisal Alghadir
- College of Medicine, King Saud University, Riyadh, 11433, Saudi Arabia
| | - Amir Iqbal
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia.
| | - Ahmad H Alghadir
- Rehabilitation Research Chair, Department of Rehabilitation Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh, 11433, Saudi Arabia
| |
Collapse
|
15
|
Tanashyan MM, Raskurazhev AA, Shabalina AA, Mazur AS, Annushkin VA, Kuznetsova PI, Illarioshkin SN, Piradov MA. Differential Pattern of Circulating MicroRNA Expression in Patients with Intracranial Atherosclerosis. Biomedicines 2025; 13:514. [PMID: 40002927 PMCID: PMC11853257 DOI: 10.3390/biomedicines13020514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/12/2025] [Accepted: 01/22/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Intracranial atherosclerosis (ICAS) is a major cause of ischemic stroke, yet fundamental studies regarding epigenetic regulation of ICAS are lacking. We hypothesized that, due to anatomical and/or functional differences, extracranial atherosclerosis is distinct from ICAS, which may explain the clinical variability as well. Methods: We chose a number of miRNAs involved in various steps of atherogenesis (namely, miR-712/205-5p/-3p, miR-106b-3p/-5p, miR-146a-3p/-5p, miR-100-3p/miR-5p, miR-200c-3p/-5p, miR-532-3p/-5p, and miR-126-3p/-5p) and examined their plasma levels in a cohort of patients with carotid stenosis > 50% (n = 35, mean age: 65 years, 54% male; 12 patients had ICAS). Results: A differential pattern of circulating miR expression was found in ICAS patients: there was an overexpression of miR-712/205-5p, miR-106b-5p, miR-146a-5p, miR-200c-5p, miR-532-3p, and miR-126-3p. The following miRs were underexpressed in intracranial atherosclerosis-miR-712/205-3p and miR-100-3p. These changes represent a plethora of atherogenic mechanisms: smooth muscle cell migration (miR-712/205, miR-532), foam cell formation (miR-106b, miR-146a), endothelial dysfunction (miR-200c), low-density lipoprotein-induced vascular damage (miR-100), and leukocyte recruitment (miR-126). In symptomatic ICAS patients, we observed a statistically significant upregulation of miR-712/205-3p and miR-146a-5p. Conclusions: Overall, the findings of our pilot study revealed several new and interesting associations: (1) intracranial atherosclerosis seems to have a different epigenetic profile (regarding circulating microRNA expression) than isolated extracranial vessel involvement; (2) ischemic stroke in ICAS may be potentiated by other pathophysiologic mechanisms than in extracranial-only atherosclerosis (ECAS). Certain miRs (e.g., miR-712/205) seem to have a larger impact on ICAS than on extracranial atherosclerosis; this may be potentially linked to difference between extra- and intracranial artery morphology and physiology, and/or may lead to the said differences. This underscores the importance of making a distinction in future epigenetic studies between ECAS and ICAS, as the mechanisms of atherogenesis are likely to vary.
Collapse
Affiliation(s)
| | | | | | | | | | - Polina I. Kuznetsova
- Research Center of Neurology, Moscow 125367, Russia; (M.M.T.); (A.A.R.); (A.S.M.); (V.A.A.); (S.N.I.); (M.A.P.)
| | | | | |
Collapse
|
16
|
Fan X, Yang G, Wang Y, Shi H, Nitschke K, Sattler K, Abumayyaleh M, Cyganek L, Nuhn P, Worst T, Liao B, Dobreva G, Duerschmied D, Zhou X, El-Battrawy I, Akin I. Exosomal mir-126-3p derived from endothelial cells induces ion channel dysfunction by targeting RGS3 signaling in cardiomyocytes: a novel mechanism in Takotsubo cardiomyopathy. Stem Cell Res Ther 2025; 16:36. [PMID: 39901299 PMCID: PMC11792229 DOI: 10.1186/s13287-025-04157-0] [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: 11/06/2024] [Accepted: 01/21/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND Takotsubo cardiomyopathy (TTC) is marked by an acute, transient, and reversible left ventricular systolic dysfunction triggered by stress, with endothelial dysfunction being one of its pathophysiological mechanisms. However, the precise molecular mechanism underlying the interaction between endothelial cells and cardiomyocytes during TTC remains unclear. This study reveals that exosomal miRNAs derived from endothelial cells exposed to catecholamine contribute to ion channel dysfunction in the setting of TTC. METHODS Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) were treated with epinephrine (Epi) or exosomes (Exo) from Epi-treated human cardiac microvascular endothelial cells (HCMECs) or Exo derived from HCMECs transfected with miR-126-3p. The immunofluorescence staining, flow cytometry, qPCR, single-cell contraction, intracellular calcium transients, patch-clamp, dual luciferase reporter assay and western blot were performed for the study. RESULTS Modeling TTC with high doses of epinephrine (Epi) treatment in hiPSC-CMs shows suppression of depolarization velocity (Vmax), prolongation of action potential duration (APD), and induction of arrhythmic events. Exo derived from HCMECs treated with Epi (Epi-exo) mimicked or enhanced the effects of Epi. Epi exposure led to elevated levels of miR-126-3p in both HCMECs and their exosomes. Exo enriched with miR-126-3p demonstrated similar effects as Epi-exo, establishing the crucial role of miR-126-3p in the mechanism of Epi-exo. Dual luciferase reporter assay coupled with gene mutation techniques identified that miR-126-3p was found to target the regulator of G-protein signaling 3 (RGS3) gene. Western blot and qPCR analyses confirmed that miR-126-3p-mimic reduced RGS3 expression in both HCMECs and hiPSC-CMs, indicating miR-126-3p inhibits RGS3 signaling. Additionally, miR-126-3p levels were significantly higher in the serum of TTC patients compared to healthy controls and patients who had recovered from TTC. CONCLUSIONS Our study is the first to reveal that exosomal miR-126-3p, originating from endothelial cells, contributes to ion channel dysfunction by regulating RGS3 signaling in cardiomyocytes. These findings provide new perspectives on the pathogenesis of TTC and suggest potential therapeutic targets for treatment.
Collapse
Affiliation(s)
- Xuehui Fan
- Department of Cardiology, Angiology, Hemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, Mannheim, Germany.
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China.
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany.
| | - Guoqiang Yang
- Department of Cardiology, Angiology, Hemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, Mannheim, Germany
- Acupuncture and Rehabilitation Department, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yinuo Wang
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Haojie Shi
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Katja Nitschke
- Department of Urology and Urosurgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Katherine Sattler
- Department of Cardiology, Angiology, Hemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, Mannheim, Germany
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Mohammad Abumayyaleh
- Department of Cardiology, Angiology, Hemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, Mannheim, Germany
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany
- DZHK (German Center for Cardiovascular Research), Partner Site, Göttingen, Germany
| | - Philipp Nuhn
- Department of Urology and Urosurgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Thomas Worst
- Department of Urology and Urosurgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Bin Liao
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Sichuan, 646000, China
| | - Gergana Dobreva
- Department of Cardiovascular Genomics and Epigenomics, European Center for Angioscience (ECAS), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Daniel Duerschmied
- Department of Cardiology, Angiology, Hemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, Mannheim, Germany
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Xiaobo Zhou
- Department of Cardiology, Angiology, Hemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, Mannheim, Germany.
- Key Laboratory of Medical Electrophysiology of the Ministry of Education, Medical Electrophysiological Key Laboratory of Sichuan Province, Institute of Cardiovascular Research, Southwest Medical University, Luzhou, Sichuan, China.
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany.
- Department of Cardiac Macrovascular Surgery, Affiliated Hospital of Southwest Medical University, Sichuan, 646000, China.
| | - Ibrahim El-Battrawy
- Department of Cardiology and Angiology, Bergmannsheil University Hospitals, Ruhr University of Bochum, 44789, Bochum, Germany
- Institute of Physiology, Department of Cellular and Translational Physiology, Medical Faculty and Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, Bochum, Germany
| | - Ibrahim Akin
- Department of Cardiology, Angiology, Hemostaseology and Medical Intensive Care, Medical Faculty Mannheim, University Medical Centre Mannheim (UMM), Heidelberg University, Mannheim, Germany
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| |
Collapse
|
17
|
Kostiniuk D, Marttila S, Raitoharju E. Circulatory miRNAs in essential hypertension. Atherosclerosis 2025; 401:119069. [PMID: 39645458 DOI: 10.1016/j.atherosclerosis.2024.119069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/22/2024] [Accepted: 11/22/2024] [Indexed: 12/09/2024]
Abstract
MicroRNAs (miRNAs) are short non-coding RNAs, that regulate gene-expression at post-transcriptional level. Unlike other RNA species, blood miRNAs circulate in a highly stable form, either within extracellular vesicles or bound to proteins. In recent years, circulatory miRNA profiles have been proposed as potential biomarkers for multitude of pathologies, including essential hypertension. However, the evidence of miRNA biomarker potential is limited, mainly due to the scarcity of profiling studies associating miRNA levels with hypertension. Furthermore, most of these studies have been performed with preselected miRNA pool, limiting their discovery potential. Here, we summarize the results of the unbiased profiling studies and additionally discuss findings from targeted miRNA analysis. Only miR-30e has been found to be associated with hypertension in more than one unbiased study. The targeted analyses highlight the association of miR-1, -21, -34a, -92a, -122, -126, -143, -145, -605, -623, -1299, as well as let-7 and miR-30 families with hypertension. Current literature indicates that some of these miRNAs are involved in hypertension-associated vascular dysfunction and the development of atherosclerosis, suggesting a novel mechanism for cardiovascular disease risk posed by hypertension. All in all, studies associating hypertension with circulatory miRNA profiles are scarce, with several limitations affecting the comparability of the studies. This review discusses the functions and potential mechanisms linking the identified miRNAs to hypertension and underscores the need for further research.
Collapse
Affiliation(s)
- Daria Kostiniuk
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland
| | - Saara Marttila
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland; Gerontology Research Center, Tampere University, Tampere, 33014, Finland
| | - Emma Raitoharju
- Molecular Epidemiology, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland; Tampere University Hospital, Wellbeing Services County of Pirkanmaa, Tampere, Finland; Finnish Cardiovascular Research Center Tampere, Faculty of Medicine and Health Technology, Tampere University, Arvo Ylpön katu 34, Tampere, 33520, Finland; Fimlab Laboratories, Finland.
| |
Collapse
|
18
|
Azami P, Mohammadzadeh S, Seirafi S, Razeghian-Jahromi I. A review of cutting-edge biomarkers for diagnosing coronary artery disease. Medicine (Baltimore) 2025; 104:e41377. [PMID: 39854741 PMCID: PMC11771658 DOI: 10.1097/md.0000000000041377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/26/2025] Open
Abstract
Chronic coronary artery disease (CAD) remains a significant global healthcare burden. Current risk assessment methods have notable limitations in early detection and risk stratification. Hence, there is an urgent need for innovative biomarkers that facilitate the premature CAD diagnosis, ultimately leading to reduction in associated morbidity and mortality rates. This review comprehensively examines recent advances in emerging biomarkers for CAD detection. Our analysis delves into various aspects of these biomarkers such as their mechanisms of action, roles in the pathophysiology of the disease, and different measurement techniques employed in clinical practice. Comparative assessment of biomarker performance between CAD patients and control groups was also presented relying on their sensitivity, specificity, and area under the curve at specific cutoff points. In this regard, prominent biomarkers including Tenascin-C, IL-37, PTX3, transthyretin, soluble interleukin-6 receptor α, and miR-15a are identified as having high diagnostic potential for chronic CAD that indeed showcase promising performance metrics. These findings underscore the role of novel biomarkers in enhancing CAD risk stratification and improving patient outcomes through early intervention. However, the pursuit of an ideal and inclusive biomarker continues due to the multifaceted nature of CAD. Future randomized controlled trials are essential to bridge the gap between research findings and clinical practice in order to augment the practical application of these biomarkers in routine healthcare settings.
Collapse
Affiliation(s)
- Pouria Azami
- Cardiovascular Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Soroush Seirafi
- Department of Cardiology, Shiraz University of Medical Sciences, Shiraz, Iran
| | | |
Collapse
|
19
|
Gambacurta A, Tullio V, Savini I, Mauriello A, Catani MV, Gasperi V. Identification of the EBF1/ETS2/KLF2-miR-126-Gene Feed-Forward Loop in Breast Carcinogenesis and Stemness. Int J Mol Sci 2025; 26:328. [PMID: 39796183 PMCID: PMC11719960 DOI: 10.3390/ijms26010328] [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: 11/25/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
MicroRNA (miR)-126 is frequently downregulated in malignancies, including breast cancer (BC). Despite its tumor-suppressive role, the mechanisms underlying miR-126 deregulation in BC remain elusive. Through silencing experiments, we identified Early B Cell Factor 1 (EBF1), ETS Proto-Oncogene 2 (ETS2), and Krüppel-Like Factor 2 (KLF2) as pivotal regulators of miR-126 expression. These transcription factors were found to be downregulated in BC due to epigenetic silencing or a "poised but not transcribed" promoter state, impairing miR-126 expression. Gene Ontology analysis of differentially expressed miR-126 target genes in the Cancer Genome Atlas: Breast Invasive Carcinoma (TCGA-BRCA) cohort revealed their involvement in cancer-related pathways, primarily signal transduction, chromatin remodeling/transcription, and differentiation/development. Furthermore, we defined interconnections among transcription factors, miR-126, and target genes, identifying a potential feed-forward loop (FFL) crucial in maintaining cellular identity and preventing the acquisition of stemness properties associated with cancer progression. Our findings propose that the dysregulation of the EBF1/ETS2/KLF2/miR-126 axis disrupts this FFL, promoting oncogenic transformation and progression in BC. This study provides new insights into the molecular mechanisms of miR-126 downregulation in BC and highlights potential targets for therapeutic intervention. Further research is warranted to clarify the role of this FFL in BC, and to identify novel therapeutic strategies aimed at modulating this network as a whole, rather than targeting individual signals, for cancer management.
Collapse
Affiliation(s)
- Alessandra Gambacurta
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (A.G.); (V.T.); (I.S.); (A.M.)
- NAST Centre (Nanoscience & Nanotechnology & Innovative Instrumentation), 00133 Rome, Italy
| | - Valentina Tullio
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (A.G.); (V.T.); (I.S.); (A.M.)
| | - Isabella Savini
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (A.G.); (V.T.); (I.S.); (A.M.)
| | - Alessandro Mauriello
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (A.G.); (V.T.); (I.S.); (A.M.)
| | - Maria Valeria Catani
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (A.G.); (V.T.); (I.S.); (A.M.)
| | - Valeria Gasperi
- Department of Experimental Medicine, Tor Vergata University of Rome, 00133 Rome, Italy; (A.G.); (V.T.); (I.S.); (A.M.)
| |
Collapse
|
20
|
Al-Ward H, Chen W, Gao W, Zhang C, Yang X, Xiong Y, Wang X, Agila R, Xu H, Sun YE. Can miRNAs in MSCs-EVs Offer a Potential Treatment for Hypoxic-ischemic Encephalopathy? Stem Cell Rev Rep 2025; 21:236-253. [PMID: 39503828 DOI: 10.1007/s12015-024-10803-6] [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] [Accepted: 10/09/2024] [Indexed: 01/26/2025]
Abstract
Neonatal hypoxic-ischemic encephalopathy (HIE) is a critical condition resulting from impaired oxygen and blood flow to the brain during birth, leading to neuroinflammation, neuronal apoptosis, and long-term neurological deficits. Despite the use of therapeutic hypothermia, current treatments remain inadequate in fully preventing brain damage. Recent advances in mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) offer a novel, cell-free therapeutic approach, as these EVs can cross the blood-brain barrier (BBB) and deliver functional microRNAs (miRNAs) to modulate key pathways involved in inflammation and neuroprotection. This review examines how specific miRNAs encapsulated in MSC-EVs-including miR-21, miR-124, miR-146, and the miR-17-92 cluster-target the complex inflammatory responses that drive HIE pathology. By modulating pathways such as NF-κB, STAT3, and PI3K/Akt, these miRNAs influence neuroinflammatory processes, reduce neuronal apoptosis, and promote tissue repair. The aim is to assess the therapeutic potential of miRNA-loaded MSC-EVs in mitigating inflammation and neuronal damage, thus addressing the limitations of current therapies like therapeutic hypothermia.
Collapse
Affiliation(s)
- Hisham Al-Ward
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenxia Gao
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chunxue Zhang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xueyan Yang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yao Xiong
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xinyi Wang
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Rafeq Agila
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, China
| | - Hui Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiamusi University, Jiamusi, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China.
- Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
21
|
Wehbe Z, Wehbe M, Al Khatib A, Dakroub AH, Pintus G, Kobeissy F, Eid AH. Emerging understandings of the role of exosomes in atherosclerosis. J Cell Physiol 2025; 240:e31454. [PMID: 39370679 PMCID: PMC11730360 DOI: 10.1002/jcp.31454] [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: 01/23/2024] [Revised: 08/20/2024] [Accepted: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Atherosclerosis remains a major contributor to cardiovascular disease, the leading cause of global morbidity and mortality. Despite the elucidation of several molecular, biochemical, and cellular aspects that contribute to the etio-pathogenesis of atherosclerosis, much remains to be understood about the onset and progression of this disease. Emerging evidence supports a role for exosomes in the cellular basis of atherosclerosis. Indeed, exosomes of activated monocytes seem to accentuate a positive feedback loop that promotes recruitment of pro-inflammatory leukocytes. Moreover, in addition to their role in promoting proliferation and invasion of vascular smooth muscle cells, exosomes can also induce neovascularization within lesions and increase endothelial permeability, two important features of fibrous plaques. Depending on their sources and cargo, exosomes can also induce clot formation and contribute to other hallmarks of atherosclerosis. Taken together, it is becoming increasingly evident that a better understanding of exosome biology is integral to elucidating the pathogenesis of atherosclerosis, and may thus provide insight into a potentially new therapeutic target for this disease.
Collapse
Affiliation(s)
- Zena Wehbe
- Vascular Biology Research Centre, Molecular and Clinical Research InstituteSt. George's University of LondonLondonUnited Kingdom
| | - Maya Wehbe
- Oxford University HospitalsOxfordUnited Kingdom
| | - Ali Al Khatib
- Department of Nutrition and Food SciencesLebanese International UniversityBeirutLebanon
| | - Ali H. Dakroub
- Departments of Medicine (Cardiology) and Population Health Science and Policy, Blavatnik Family Research InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | - Gianfranco Pintus
- Department of Biomedical SciencesUniversity of Sassari, Viale San PietroSassari07100Italy
| | - Firas Kobeissy
- Department of Neurobiology, Morehouse School of MedicineCenter for Neurotrauma, Multiomics & Biomarkers (CNMB)AtlantaGAUSA
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of MedicineQU Health, Qatar UniversityDohaP.O. Box 2713Qatar
| |
Collapse
|
22
|
Goyal K, Afzal M, Altamimi ASA, Babu MA, Ballal S, Kaur I, Kumar S, Kumar MR, Chauhan AS, Ali H, Shahwan M, Gupta G. Chronic kidney disease and aging: dissecting the p53/p21 pathway as a therapeutic target. Biogerontology 2024; 26:32. [PMID: 39725742 DOI: 10.1007/s10522-024-10173-z] [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: 11/09/2024] [Accepted: 12/14/2024] [Indexed: 12/28/2024]
Abstract
Chronic kidney diseases (CKD) are a group of multi-factorial disorders that markedly impair kidney functions with progressive renal deterioration. Aging contributes to age-specific phenotypes in kidneys, which undergo several structural and functional alterations, such as a decline in regenerative capacity and increased fibrosis, inflammation, and tubular atrophy, all predisposing them to disease and increasing their susceptibility to injury while impeding their recovery. A central feature of these age-related processes is the activation of the p53/p21 pathway signaling. The pathway is a key player in cellular senescence, apoptosis, and cell cycle regulation, which are all key to maintaining the health of the kidney. P53 is a transcription factor and a tumor suppressor protein that responds to cell stress and damage. Persistent activation of cell p53 can lead to the expression of p21, an inhibitor of the cell cycle known as a cyclin-dependent kinase. This causes cells to cease dividing and leads to senescence, where cells can no longer increase. The accumulation of senescent cells in the aging kidney impairs kidney function by altering the microenvironment. As the number of senescent cells increases, the capacity of the kidney to recover from injury decreases, accelerating the progression of end-stage renal disease. This article review extensively explores the relationship between the p53/p21 pathway and cellular senescence within an aging kidney and the emerging therapeutic strategies that target it to overcome the impacts of cellular senescence on CKD.
Collapse
Affiliation(s)
- Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to Be University), Clement Town, Dehradun, 248002, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, 21442, Jeddah, Saudi Arabia
| | | | - M Arockia Babu
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Irwanjot Kaur
- Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Sachin Kumar
- NIMS Institute of Pharmacy, NIMS University Rajasthan, Jaipur, India
| | - M Ravi Kumar
- Chandigarh Pharmacy College, Chandigarh Group of College, Jhanjeri, Mohali, Punjab, 140307, India
| | - Ashish Singh Chauhan
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, 531162, India
| | - Haider Ali
- Uttaranchal Institute of Pharmaceutical Sciences, Division of Research and Innovation, Uttaranchal University, Dehradun, India
| | - Moyad Shahwan
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab, 140401, India.
| |
Collapse
|
23
|
Zheng Y, Xu C, Jin Y. The role of exosomes in the pathogenesis and management of diabetic kidney disease: a systematic review and meta-analysis. Front Endocrinol (Lausanne) 2024; 15:1398382. [PMID: 39703859 PMCID: PMC11658263 DOI: 10.3389/fendo.2024.1398382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 10/18/2024] [Indexed: 12/21/2024] Open
Abstract
Objective This systematic review and meta-analysis aimed to synthesize the role of exosomes in the pathogenesis and management of diabetic kidney disease. Methods PubMed, Embase, Cochrane Library, and Web of Science were searched for studies that compared the levels of exosomes between patients with diabetic kidney disease and controls published up to 27 November 2023. Methodological quality was assessed using the JBI Appraisal Checklist for Case-Control Studies. The methodology of the samples and the main results were summarized. A meta-analysis of the diagnostic performance of exosomes was performed using estimates of test sensitivity and specificity, and these values were summarized using summary receiver-operating characteristic curves. The results were reported following the PRISMA 2020 checklist. Results A total of 32 studies, including 1,119 patients with diabetic kidney disease and 1,328 controls, met the inclusion criteria. A total of 78 upregulated and 22 downregulated microRNAs, 2 upregulated and 4 downregulated mRNAs, 6 upregulated and 1 downregulated proteins, and 4 upregulated lipids were identified. The miR-126, miR-145, miR-150, miR-21, and WT1 mRNA dysregulation were consistently reported in at least two studies. The overall sensitivity and specificity of the exosomes in diabetic kidney disease diagnosis were 0.70 (95% CI: 0.59-0.80) and 0.79 (95% CI: 0.70-0.85), respectively. The summary receiver operating characteristic curve was plotted to assess diagnostic accuracy with the area under the curve (AUC) of 0.82 (95% CI: 0.78-0.85). Conclusion Exosomes have great potential to become effective diagnostic biomarkers for diabetic kidney disease. Panels of exosomes or the combination of exosomes with other clinical indicators seemed more accurate than single exosomes.
Collapse
Affiliation(s)
- Yan Zheng
- Department of Endocrinology, Zhoushan Hospital, Zhejiang Province, Zhoushan, Zhejiang, China
| | | | | |
Collapse
|
24
|
Wang L, Xu W, Zhang S, Gundberg GC, Zheng CR, Wan Z, Mustafina K, Caliendo F, Sandt H, Kamm R, Weiss R. Sensing and guiding cell-state transitions by using genetically encoded endoribonuclease-mediated microRNA sensors. Nat Biomed Eng 2024; 8:1730-1743. [PMID: 38982158 DOI: 10.1038/s41551-024-01229-z] [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: 04/24/2023] [Accepted: 05/11/2024] [Indexed: 07/11/2024]
Abstract
Precisely sensing and guiding cell-state transitions via the conditional genetic activation of appropriate differentiation factors is challenging. Here we show that desired cell-state transitions can be guided via genetically encoded sensors, whereby endogenous cell-state-specific miRNAs regulate the translation of a constitutively transcribed endoribonuclease, which, in turn, controls the translation of a gene of interest. We used this approach to monitor several cell-state transitions, to enrich specific cell types and to automatically guide the multistep differentiation of human induced pluripotent stem cells towards a haematopoietic lineage via endothelial cells as an intermediate state. Such conditional activation of gene expression is durable and resistant to epigenetic silencing and could facilitate the monitoring of cell-state transitions in physiological and pathological conditions and eventually the 'rewiring' of cell-state transitions for applications in organoid-based disease modelling, cellular therapies and regenerative medicine.
Collapse
Affiliation(s)
- Lei Wang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Bioengineering, Northeastern University, Boston, MA, USA.
- Department of Biology, Northeastern University, Boston, MA, USA.
| | - Wenlong Xu
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Shun Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China
- State Key Laboratory of Organ Regeneration and Reconstruction, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Gregory C Gundberg
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christine R Zheng
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Zhengpeng Wan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Kamila Mustafina
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Fabio Caliendo
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Hayden Sandt
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Roger Kamm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ron Weiss
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
25
|
Piacquadio KA, Margolis LM, Gwin JA, Leidy HJ. Preliminary Evidence Supports that Long-Term Consumption of Higher-Protein Breakfast Promotes Higher Expression of Select miRNA Associated with Cardiometabolic Health in Adolescents. J Nutr 2024; 154:3585-3591. [PMID: 39393494 DOI: 10.1016/j.tjnut.2024.10.007] [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: 06/08/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Increased dietary protein at breakfast promotes cardiometabolic health; however, whether these improvements occur at the molecular level is unknown. OBJECTIVES The objective was to examine whether long-term consumption of breakfast, varying in protein quantity, alters the expression of circulating microRNAs (miRNAs) associated with cardiometabolic health in "breakfast-skipping" adolescents. METHODS Thirty adolescents (age: 19 ± 1 y; body mass index: 25.4 ± 3 kg/m2) completed a 6-mo tightly controlled breakfast trial in which participants consumed 350 kcal normal-protein (NP, 10 g protein) or higher-protein (HP, 30 g protein) breakfasts or continued to BS for 6 mo. Fasting blood samples were collected at baseline (PRE) and 6 mo (POST) for assessment of 12 a priori circulating plasma miRNA expression levels (real-time quantitative polymerase chain reaction), glucose, insulin, IL-6, and C-reactive protein. RESULTS No main effects of group were observed for any miRNAs; however, a time-by-group interaction was detected for the expression of miR-126-3p (P = 0.05). HP breakfast tended to increase miR-126-3p expression throughout the study (POST-PRE, P = 0.09) leading to greater expression at POST compared with BS (P = 0.03), whereas NP breakfast did not. Additionally, several miRNAs predicted fasting concentrations of IL-6: miR-320a-3p, -146a-5p, -150-5p, -423-5p, -122-5p, glucose: miR-24-3p, -126-3p; insulin: miR-24-3p, -126-3p, -15b-5p; insulin sensitivity: miR-24-3p, -126-3p, -199a-5p, -15b-5p; and β-cell function: miR-15b-5p (R2 between 0.2 and 0.39; P < 0.05) from PRE and POST samples across groups. CONCLUSIONS These data support the daily consumption of a HP breakfast to promote cardiometabolic health, potentially through changes in miRNA expression, in a sensitive life-stage where early intervention strategies are critical to reduce the risk of adult-onset chronic disease. TRIAL REGISTRATION NUMBER NCT03146442.
Collapse
Affiliation(s)
- Kamille A Piacquadio
- Department of Nutritional Sciences & Department of Pediatrics, University of Texas at Austin, Austin, TX, United States
| | - Lee M Margolis
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Jess A Gwin
- Military Nutrition Division, US Army Research Institute of Environmental Medicine, Natick, MA, United States
| | - Heather J Leidy
- Department of Nutritional Sciences & Department of Pediatrics, University of Texas at Austin, Austin, TX, United States.
| |
Collapse
|
26
|
Brisebois MF, Gordon RA, Zumbro EL, Sokoloski ML, Duplanty AA, Juma S, Rigby BR. Acute Effects of Serial and Integrated Concurrent Exercise on Circulating microRNAs -126 and -222 in Sedentary Adults. INTERNATIONAL JOURNAL OF EXERCISE SCIENCE 2024; 17:1444-1460. [PMID: 39807384 PMCID: PMC11728573 DOI: 10.70252/xfjk8005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
The purpose of this study was to compare changes in circulating microRNAs -126 (c-miR-126) and -222 (c-miR-222) following acute serial concurrent exercise (SCE) and integrated concurrent exercise (ICE) sessions among young, sedentary adults. Ten males and 9 females completed the study procedures. For SCE, participants performed resistance exercise (RE) followed by aerobic exercise (AE), without mixing the two. For ICE, participants performed a brief bout of AE before each set of RE. Blood was collected before, immediately after (IP), and 1 h (1HR) after each exercise session. Expression of c-miR-126 significantly increased from baseline at IP (1.6-fold SCE, 2.1-fold ICE; p = .037) and 1HR (1.8-fold SCE, 1.7-fold ICE; p = .034) following both sessions, with no difference between the two sessions. Expression of c-miR-222 significantly increased from baseline at IP (1.7-fold SCE, 1.9-fold ICE; p = .024) and 1HR (2.0-fold SCE, 1.6-fold ICE; p = .038) following both sessions, with no difference between the two sessions. There were no differences in peak heart rate or average heart rate between the two workout sessions. Both SCE and ICE patterns appear equally effective at acutely increasing c-miR-126 and -222.
Collapse
Affiliation(s)
- Matthew F Brisebois
- Department of Human Performance and Health, University of South Carolina Upstate, Spartanburg, SC, USA
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX, USA
| | - Ryan A Gordon
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX, USA
- Department of Kinesiology, Missouri State University, Springfield, MO, USA
| | - Emily L Zumbro
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX, USA
- Department of Medicine, Division of Gerontology, Geriatrics and Palliative Care, the University of Alabama at Birmingham, Birmingham, AL, USA
- Center for Exercise Medicine, The University of Alabama Birmingham, Birmingham, AL, USA
| | - Matthew L Sokoloski
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX, USA
| | - Anthony A Duplanty
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX, USA
| | - Shanil Juma
- Department of Nutrition and Food Sciences, Texas Woman's University, Denton, TX, USA
| | - Brandon R Rigby
- School of Health Promotion and Kinesiology, Texas Woman's University, Denton, TX, USA
- Institute for Women's Health, Texas Woman's University, Denton, TX, USA
| |
Collapse
|
27
|
Xiao J, Xu Z. Roles of noncoding RNAs in diabetic retinopathy: Mechanisms and therapeutic implications. Life Sci 2024; 357:123092. [PMID: 39368772 DOI: 10.1016/j.lfs.2024.123092] [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: 07/09/2024] [Revised: 08/20/2024] [Accepted: 09/28/2024] [Indexed: 10/07/2024]
Abstract
Diabetic retinopathy (DR) is a microvascular complication of diabetes that leads to vision loss. The striking features of DR are hard exudate, cotton-wool spots, hemorrhage, and neovascularization. The dysregulated retinal cells, encompassing microvascular endothelial cells, pericytes, Müller cells, and adjacent retinal pigment epithelial cells, are involved in the pathological processes of DR. According to recent research, oxidative stress, inflammation, ferroptosis, pyroptosis, apoptosis, and angiogenesis contribute to DR. Recent advancements have highlighted that noncoding RNAs could regulate diverse targets in pathological processes that contribute to DR. Noncoding RNAs, including long noncoding RNAs, microRNAs (miRNA), and circular RNAs, are dysregulated in DR, and interact with miRNA, mRNA, or proteins to control the pathological processes of DR. Hence, modulation of noncoding RNAs may have therapeutic effects on DR. Small extracellular vesicles may be valuable tools for transferring noncoding RNAs and regulating the genes involved in progression of DR. However, the roles of noncoding RNA in developing DR are not fully understood; it is critical to summarize the mechanisms for noncoding RNA regulation of pathological processes and pathways related to DR. This review provides a fundamental understanding of the relationship between noncoding RNAs and DR, exploring the mechanism of how noncoding RNA modulates different signaling pathways, and pave the way for finding potential therapeutic strategies for DR.
Collapse
Affiliation(s)
- Jing Xiao
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhuping Xu
- Department of Ophthalmology, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
28
|
Zhang LJ, Hu YX, Huang RZ, Xu YY, Dong SH, Guo FH, Guo JJ, Qiu JJ, Cao ZY, Wei LJ, Mao JH, Lyu A, Liu JL, Zhao XX, Guo ZF, Jing Q. Intraplatelet miRNA-126 regulates thrombosis and its reduction contributes to platelet inhibition. Cardiovasc Res 2024; 120:1622-1635. [PMID: 38900927 DOI: 10.1093/cvr/cvae138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 07/17/2023] [Accepted: 01/08/2024] [Indexed: 06/22/2024] Open
Abstract
AIMS MicroRNA-126 (miR-126), one of the most abundant microRNAs in platelets, is involved in the regulation of platelet activity and the circulating miR-126 is reduced during antiplatelet therapy. However, whether intraplatelet miR-126 plays a role in thrombosis and platelet inhibition remains unclear. METHODS AND RESULTS Here, using tissue-specific knockout mice, we reported that the deficiency of miR-126 in platelets and vascular endothelial cells significantly prevented thrombosis and prolonged bleeding time. Using chimeric mice, we identified that the lack of intraplatelet miR-126 significantly prevented thrombosis. Ex vivo experiments further demonstrated that miR-126-deficient platelets displayed impaired platelet aggregation, spreading, and secretory functions. Next, miR-126 was confirmed to target phosphoinositol-3 kinase regulatory subunit 2 (PIK3R2) in platelet, which encodes a negative regulator of the phosphoinositide 3-kinase/protein kinase B pathway, enhancing platelet activation through activating the integrin αIIbβ3-mediated outside-in signalling. After undergoing myocardial infarction (MI), chimeric mice lacking intraplatelet miR-126 displayed reduced microvascular obstruction and prevented MI expansion in vivo. In contrast, overexpression of miR-126 by the administration of miR-126 agonist (agomiR-126) in wild-type mice aggravated microvascular obstruction and promoted MI expansion, which can be almost abolished by aspirin administration. In patients with cardiovascular diseases, antiplatelet therapies, either aspirin alone or combined with clopidogrel, decreased the level of intraplatelet miR-126. The reduction of intraplatelet miR-126 level was associated with the decrease in platelet activity. CONCLUSION Our murine and human data reveal that (i) intraplatelet miR-126 contributes to platelet activity and promotes thrombus formation, and (ii) the reduction of intraplatelet miR-126 contributes to platelet inhibition during antiplatelet therapy.
Collapse
Affiliation(s)
- Lu-Jun Zhang
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Yang-Xi Hu
- Department of Cardiology, Shanghai Changzheng Hospital, Shanghai, China
| | - Rong-Zhong Huang
- Department of Geriatrics, Second Hospital Affiliated to Chongqing Medical University, Chongqing Medical University, Chongqing, China
| | - Yan-Yan Xu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shao-Hua Dong
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Fang-Hao Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jun-Jun Guo
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Jing-Jing Qiu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Zi-Yun Cao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Li-Jiang Wei
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia-Hao Mao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| | - Ankang Lyu
- Department of Cardiology, Rui Jin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun-Ling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory of Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xian-Xian Zhao
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Zhi-Fu Guo
- Department of Cardiology, Shanghai Changhai Hospital, 168 Changhai Road, Shanghai 200433, China
| | - Qing Jing
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Innovation Center for Intervention of Chronic Disease and Promotion of Health, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue-Yang Road, Shanghai 200031, China
| |
Collapse
|
29
|
Zhang J. Non-coding RNAs and angiogenesis in cardiovascular diseases: a comprehensive review. Mol Cell Biochem 2024; 479:2921-2953. [PMID: 38306012 DOI: 10.1007/s11010-023-04919-5] [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: 10/27/2023] [Accepted: 12/18/2023] [Indexed: 02/03/2024]
Abstract
Non-coding RNAs (ncRNAs) have key roles in the etiology of many illnesses, including heart failure, myocardial infarction, stroke, and in physiological processes like angiogenesis. In transcriptional regulatory circuits that control heart growth, signaling, and stress response, as well as remodeling in cardiac disease, ncRNAs have become important players. Studies on ncRNAs and cardiovascular disease have made great progress recently. Here, we go through the functions of non-coding RNAs (ncRNAs) like circular RNAs (circRNAs), and microRNAs (miRNAs) as well as long non-coding RNAs (lncRNAs) in modulating cardiovascular disorders.
Collapse
Affiliation(s)
- Jie Zhang
- Medical School, Shandong University of Traditional Chinese Medicine, Jinan, 250355, China.
| |
Collapse
|
30
|
Nazir A, Uwishema O, Shariff S, Franco WXG, El Masri N, Ayele ND, Munyangaju I, Alzain FE, Wojtara M. A Thorough Navigation of miRNA's Blueprint in Crafting Cardiovascular Fate. Health Sci Rep 2024; 7:e70136. [PMID: 39502130 PMCID: PMC11535861 DOI: 10.1002/hsr2.70136] [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: 09/02/2023] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
Introduction Cardiovascular diseases contribute significantly to global morbidity and mortality. MicroRNAs are crucial in the development and progression of these diseases by regulating gene expression in various cells and tissues. Their roles in conditions like atherosclerosis, heart failure, myocardial infarction, and arrhythmias have been widely researched. Materials and Methods The present study provides an overview of existing evidence regarding miRNAs' role in cardiovascular disease pathogenesis. Furthermore, the study examines current state-of-the-art technologies used in the study of miRNAs in cardiovascular disease. As a final point, we examine how miRNAs may serve as disease biomarkers, therapeutic targets, and prognostic indicators. Results In cardiology, microRNAs, small noncoding RNA molecules, are crucial to the posttranscriptional regulation of genes. Their role in regulating cardiac cell differentiation and maturation is critical during the development of the heart. They maintain the cardiac function of an adult heart by contributing to its electrical and contractile activity. By binding to messenger RNA molecules, they inhibit protein translation or degrade mRNA. Several cardiovascular diseases are associated with dysregulation of miRNAs, including arrhythmias, hypertension, atherosclerosis, and heart failure. miRNAs can be used as biomarkers to diagnose and predict diseases as well as therapeutic targets. A variety of state-of-the-art technologies have aided researchers in discovering, profiling, and analyzing miRNAs, including microarray analysis, next-generation sequencing, and others. Conclusion Developing new diagnostics and therapeutic approaches is becoming more feasible as researchers refine their understanding of miRNA function. Ultimately, this will reduce the burden of cardiovascular disease around the world.
Collapse
Affiliation(s)
- Abubakar Nazir
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineKing Edward Medical UniversityPakistan
| | - Olivier Uwishema
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
| | - Sanobar Shariff
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineYerevan State Medical UniversityYerevanArmenia
| | - William Xochitun Gopar Franco
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineUniversity of GuadalajaraGuadalajaraMexico
| | - Noha El Masri
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Faculty of MedicineBeirut Arab UniversityLebanon
| | - Nitsuh Dejene Ayele
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of Internal Medicine, Faculty of MedicineWolkite UniversityWolkiteEthiopia
| | - Isabelle Munyangaju
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Barcelona Institute for Global Health—Hospital ClínicUniversitat de Barcelona
| | - Fatima Esam Alzain
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineCollege of Medicine and General Surgery—Sudan University of Science and Technology
| | - Magda Wojtara
- Department of MedicineOli Health Magazine Organization, Research and EducationKigaliRwanda
- Department of MedicineUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
31
|
Liang W, Huang B, Shi Q, Yang X, Zhang H, Chen W. Circulating MicroRNAs as potential biomarkers for cerebral collateral circulation in symptomatic carotid stenosis. Front Physiol 2024; 15:1403598. [PMID: 39552721 PMCID: PMC11563797 DOI: 10.3389/fphys.2024.1403598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 10/03/2024] [Indexed: 11/19/2024] Open
Abstract
Background Cerebral collateral circulation (CCC) considerably improves the prognosis of patients with symptomatic carotid stenosis (SCS). This study evaluated the diagnostic value of plasma microRNAs (miRNAs) in determining CCC status in patients with SCS. Methods This single-center observational study enrolled patients with ≥50% carotid artery stenosis diagnosed using Doppler ultrasound. CCC was assessed using cerebrovascular digital subtraction angiography (DSA). Quantitative reverse transcription-polymerase chain reaction was used to determine the expression levels of plasma miRNAs. A multivariate logistic regression model and receiver operating characteristic (ROC) curve were used to analyze the diagnostic value of plasma miRNA expression in determining CCC status. Results A total of 43 patients were enrolled (28 with CCC and 15 without CCC). The plasma expression levels of miR-126-3p, miR-132-3p, and miR-210-3p were significantly higher and those of miR-16-3p and miR-92-3p were significantly lower in patients with CCC. After adjusting for age, gender, drinking history, comorbidities and degree of SCS, miR-92a-3p, miR-126-3p, miR-132-3p, and miR-210-3p were found to be significantly associated with CCC establishment (p < 0.05). ROC curve analysis indicated a high diagnostic value of these miRNAs in determining CCC status [area under the curve (AUC): 0.918-0.965], with miR-126-3p exhibiting the highest predictive performance (AUC: 0.965). Subgroup analysis revealed that patients with CCC who had 50%-70% stenosis showed significantly higher expression level of miR-126-3p, whereas those with CCC who had 70%-99% stenosis showed significantly higher expression levels of miR-126-3p, miR-132-3p, and miR-210-3p as well as significantly lower expression levels of miR-15a-3p, miR-16-3p, and miR-92a-3p. Conclusion The results indicate that these six plasma miRNAs have promising diagnostic value in determining CCC status in varying degrees of SCS. These miRNAs can serve as biomarkers for CCC status following SCS, with miR-126-3p showing the strongest positive correlation.
Collapse
Affiliation(s)
- Wenwen Liang
- Department of Radiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Bingcang Huang
- Department of Radiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Qin Shi
- Department of General Practice, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Xuelian Yang
- Department of Neurology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Hanwen Zhang
- Department of Neurology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Wei Chen
- Department of Radiology, Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| |
Collapse
|
32
|
Han C, Zhai C, Li A, Ma Y, Hallajzadeh J. Exercise mediates myocardial infarction via non-coding RNAs. Front Cardiovasc Med 2024; 11:1432468. [PMID: 39553846 PMCID: PMC11563808 DOI: 10.3389/fcvm.2024.1432468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 09/29/2024] [Indexed: 11/19/2024] Open
Abstract
Myocardial infarction (MI), a widespread cardiovascular issue, mainly occurs due to blood clot formation in the coronary arteries, which reduces blood flow to the heart muscle and leads to cell death. Incorporating exercise into a lifestyle can significantly benefit recovery and reduce the risk of future cardiac events for MI patients. Non-coding RNAs (ncRNAs) play various roles in the effects of exercise on myocardial infarction (MI). ncRNAs regulate gene expression, influence cardiac remodeling, angiogenesis, inflammation, oxidative stress, apoptosis, cardioprotection, and cardiac electrophysiology. The expression of specific ncRNAs is altered by exercise, leading to beneficial changes in heart structure, function, and recovery after MI. These ncRNAs modulate molecular pathways that contribute to improved cardiac health, including reducing inflammation, enhancing angiogenesis, promoting cell survival, and mitigating oxidative stress. Furthermore, they are involved in regulating changes in cardiac remodeling, such as hypertrophy and fibrosis, and can influence the electrical properties of the heart, thereby decreasing the risk of arrhythmias. Knowledge on MI has entered a new phase, with investigations of ncRNAs in physical exercise yielding invaluable insights into the impact of this therapeutic modality. This review compiled research on ncRNAs in MI, with an emphasis on their applicability to physical activity.
Collapse
Affiliation(s)
| | - Cuili Zhai
- College of Chinese Martial Arts, Beijing Sport University, Beijing, China
| | - Ailing Li
- City University of Malyasia, Kuala Lumpur, Malaysia
| | - Yongzhi Ma
- Division of Sports Science and Physical Education, Tsinghua University, Beijing, China
| | - Jamal Hallajzadeh
- Department of Biochemistry and Nutrition, Research Center for Evidence-Based Health Management, Maragheh University of Medical Sciences, Maragheh, Iran
| |
Collapse
|
33
|
Prajapat M, Sala L, Vidigal JA. The small noncoding RNA Vaultrc5 is dispensable to mouse development. RNA (NEW YORK, N.Y.) 2024; 30:1465-1476. [PMID: 39209555 PMCID: PMC11482604 DOI: 10.1261/rna.080161.124] [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: 06/25/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024]
Abstract
Vault RNAs (vtRNAs) are evolutionarily conserved small noncoding RNAs transcribed by RNA polymerase III. Vault RNAs were initially described as components of the vault particle, but have since been assigned multiple vault-independent functions, including regulation of PKR activity, apoptosis, autophagy, lysosome biogenesis, and viral particle trafficking. The full-length transcript has also been described as a noncanonical source of miRNAs, which are processed in a DICER-dependent manner. As central molecules in vault-dependent and independent processes, vtRNAs have been attributed numerous biological roles, including regulation of cell proliferation and survival, response to viral infections, drug resistance, and animal development. Yet, their impact to mammalian physiology remains largely unexplored. To study vault RNAs in vivo, we generated a mouse line with a conditional Vaultrc5 loss-of-function allele. Because Vaultrc5 is the sole murine vtRNA, this allele enables the characterization of the physiological requirements of this conserved class of small regulatory RNAs in mammals. Using this strain, we show that mice constitutively null for Vaultrc5 are viable and histologically normal but have a slight reduction in platelet counts, pointing to a potential role for vtRNAs in hematopoiesis. This work paves the way for further in vivo characterizations of this abundant but mysterious RNA molecule. Specifically, it enables the study of the biological consequences of constitutive or lineage-specific Vaultrc5 deletion and of the physiological requirements for an intact Vaultrc5 during normal hematopoiesis or in response to cellular stresses such as oncogene expression, viral infection, or drug treatment.
Collapse
Affiliation(s)
- Mahendra Prajapat
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Laura Sala
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Joana A Vidigal
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, The National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
34
|
Osaki T, Wan Z, Haratani K, Jin Y, Campisi M, Barbie DA, Kamm R, Sur M. miR126-mediated impaired vascular integrity in Rett syndrome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.11.617929. [PMID: 39415995 PMCID: PMC11482880 DOI: 10.1101/2024.10.11.617929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Rett syndrome (RTT) is a neurodevelopmental disorder that is caused by mutations in melty-CpG binding protein 2 (MeCP2). MeCP2 is a non-cell type-specific DNA binding protein, and its mutation influences not only neural cells but also non-neural cells in the brain, including vasculature associated with endothelial cells. Vascular integrity is crucial for maintaining brain homeostasis, and its alteration may be linked to the pathology of neurodegenerative disease, but a non-neurogenic effect, especially the relationship between vascular alternation and Rett syndrome pathogenesis, has not been shown. Here, we recapitulate a microvascular network using Rett syndrome patient-derived induced pluripotent stem (iPS) cells that carry MeCP2[R306C] mutation to investigate early developmental vascular impact. To expedite endothelial cell differentiation, doxycycline (DOX)-inducible ETV2 expression vectors were inserted into the AAVS1 locus of Rett syndrome patient-derived iPS cells and its isogenic control by CRISPR/Cas9. With these endothelial cells, we established a disease microvascular network (Rett-dMVNs) and observed higher permeability in the Rett-dMVNs compared to isogenic controls, indicating altered barrier function by MeCP2 mutation. Furthermore, we unveiled that hyperpermeability is involved in the upregulation of miR126-3p in Rett syndrome patient-derived endothelial cells by microRNA profiling and RNAseq, and rescue of miR126-3p level can recover their phenotype. We discover miR126-3p-mediated vascular impairment in Rett syndrome patients and suggest the potential application of these findings for translational medicine.
Collapse
Affiliation(s)
- Tatsuya Osaki
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
- Whitehead Institute for Biomedical Research, Massachusetts Institute of Technology, Cambridge, United States
| | - Zhengpeng Wan
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Koji Haratani
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ylliah Jin
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Marco Campisi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - David A. Barbie
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Roger Kamm
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, United States
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Mriganka Sur
- Picower Institute of Learning and Memory, Massachusetts Institute of Technology, Cambridge, United States
| |
Collapse
|
35
|
Tariq Z, Abusnana S, Mussa BM, Zakaria H. New insights on genetic background of major diabetic vascular complications. Diabetol Metab Syndr 2024; 16:243. [PMID: 39375805 PMCID: PMC11457557 DOI: 10.1186/s13098-024-01473-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 09/21/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND By 2045, it is expected that 693 million individuals worldwide will have diabetes and with greater risk of morbidity, mortality, loss of vision, renal failure, and a decreased quality of life due to the devastating effects of macro- and microvascular complications. As such, clinical variables and glycemic control alone cannot predict the onset of vascular problems. An increasing body of research points to the importance of genetic predisposition in the onset of both diabetes and diabetic vascular complications. OBJECTIVES Purpose of this article is to review these approaches and narrow down genetic findings for Diabetic Mellitus and its consequences, highlighting the gaps in the literature necessary to further genomic discovery. MATERIAL AND METHODS In the past, studies looking for genetic risk factors for diabetes complications relied on methods such as candidate gene studies, which were rife with false positives, and underpowered genome-wide association studies, which were constrained by small sample sizes. RESULTS The number of genetic findings for diabetes and diabetic complications has over doubled due to the discovery of novel genomics data, including bioinformatics and the aggregation of global cohort studies. Using genetic analysis to determine whether diabetes individuals are at the most risk for developing diabetic vascular complications (DVC) might lead to the development of more accurate early diagnostic biomarkers and the customization of care plans. CONCLUSIONS A newer method that uses extensive evaluation of single nucleotide polymorphisms (SNP) in big datasets is Genome-Wide Association Studies (GWAS).
Collapse
Affiliation(s)
- Zuira Tariq
- Diabetes and Endocrinology Department, University Hospital Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates
| | - Salah Abusnana
- Diabetes and Endocrinology Department, University Hospital Sharjah, P.O. Box: 27272, Sharjah, United Arab Emirates.
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.
| | - Bashair M Mussa
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Hala Zakaria
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
36
|
Amin W, Enam SA, Sufiyan S, Ghias K, Bajwa MH, Ilyas S, Laghari AA, Naeem S, Abidi SH, Mughal N. Autophagy-associated biomarkers ULK2, UVRAG, and miRNAs miR-21, miR-126, and miR-374: Prognostic significance in glioma patients. PLoS One 2024; 19:e0311308. [PMID: 39348350 PMCID: PMC11441661 DOI: 10.1371/journal.pone.0311308] [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: 02/21/2024] [Accepted: 09/06/2024] [Indexed: 10/02/2024] Open
Abstract
As the pioneering study from Pakistan, our research distinctly focuses on validating the roles of autophagy-associated genes and MicroRNAs (miRs) in the unique context of our population for glioma prognosis. The study delves into the nuanced interplay of autophagy within a miR-modulated environment, prompting an exploration of its potential impact on glioma development and survival. Employing real-time PCR (qPCR), we meticulously assessed the expression profiles of autophagy genes and miRs in glioma tissues, complemented by immunohistochemistry on Formalin-fixed paraffin-embedded tissues from the same patients. Our comprehensive statistical analyses, including the data normality hypothesis Shapiro-Wilk test, the Mann-Whitney U-test, Spearman correlation test, and Kaplan-Meier survival analysis, were tailored to unravel the intricate associations specific to low- and high-grade glioma within our population. Clinicopathological analysis revealed a predominance of male patients (66%) with a median age of 35 years. Glioblastoma (32%) and Astrocytoma (36%) were the most prevalent histopathological subtypes. Molecular analysis showed significant correlations between prognostic markers (Ki-67, IDH-1, p53) and clinicopathological factors, including age, histological type, radiotherapy, and chemotherapy. In high-grade glioma, increased expression of AKT and miR-21, coupled with reduced ULK2 and LC3 expression was distinctly observed. While correlation analysis identified a strong positive correlation between ULK2 and UVRAG, PTEN, miR-7, and miR-100 in low-grade glioma, unveiling distinctive molecular signatures unique to our study. Furthermore, a moderate positive correlation emerged between ULK2 and mTOR, miR-7, miR-30, miR-100, miR-204, and miR-374, also between miR-21 and miR-126. Similarly, a positive correlation appeared between ULK2 and AKT, LC3, PI3K, PTEN, ULK1, VPS34, mTOR, Beclin1, UVRAG, miR-7 and miR-374. AKT positively correlated with LC3, PI3K, PTEN, ULK1, VPS34, mTOR, Beclin1, UVRAG, miR-7, miR-30, miR-204, miR-374, miR-126 and miR-21 weakly correlated with AKT and miR-30 in high-grade glioma, providing further insights into the autophagy pathway within our population. The enrichment analysis for miR-21, miR-126, and miR-374 showed MAPK pathway as a common pathway along with Ras, PI3K, and mTOR pathway. The low ULK2, UVRAG, and miR-374 expression group exhibited significantly poor overall survival in glioma, while miR-21 over-expression indicated a poor prognosis in glioma patients, validating it in our population. This study provides comprehensive insights into the molecular landscape of gliomas, highlighting the dysregulation of autophagy genes ULK2, and UVRAG and the associated miR-21, miR-126 and miR-374 as potential prognostic biomarkers and emphasizing their unique significance in shaping survival outcomes in gliomas within the specific context of the Pakistani population.
Collapse
Affiliation(s)
- Wajiha Amin
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Syed Ather Enam
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan
| | - Sufiyan Sufiyan
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Kulsoom Ghias
- Department of Biological & Biomedical Science, Aga Khan University Hospital, Karachi, Pakistan
| | | | - Sahar Ilyas
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan
| | - Altaf Ali Laghari
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
| | - Sana Naeem
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan
| | - Syed Hani Abidi
- Department of Biological & Biomedical Science, Aga Khan University Hospital, Karachi, Pakistan
- Department of Biomedical Sciences, Nazarbayev School of Medicine, Nazarbayev University, Astana, Kazakhstan
- Department of Life Sciences, Western Caspian University, Baku, Azerbaijan
| | - Nouman Mughal
- Department of Surgery, Aga Khan University Hospital, Karachi, Pakistan
- Center of Oncological Research in Surgery, Aga Khan University, Karachi, Pakistan
- Department of Biological & Biomedical Science, Aga Khan University Hospital, Karachi, Pakistan
| |
Collapse
|
37
|
Domínguez-Mozo MI, Casanova I, Monreal E, Costa-Frossard L, Sainz-de-la-Maza S, Sainz-Amo R, Aladro-Benito Y, Lopez-Ruiz P, De-Torres L, Abellán S, Garcia-Martinez MA, De-la-Cuesta D, Lourido D, Torrado-Carvajal A, Gomez-Barbosa C, Linares-Villavicencio C, Villar LM, López-De-Silanes C, Arroyo R, Alvarez-Lafuente R. Association of MicroRNA Expression and Serum Neurofilament Light Chain Levels with Clinical and Radiological Findings in Multiple Sclerosis. Int J Mol Sci 2024; 25:10012. [PMID: 39337499 PMCID: PMC11432459 DOI: 10.3390/ijms251810012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/06/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
microRNAs (miRNAs) are promising biomarkers for many diseases, including multiple sclerosis (MS). The neurofilament light chain (NfL) is a biomarker that can detect axonal damage in different neurological diseases. The objective of this study was to evaluate the association of the expression profile of pre-selected miRNAs and NfL levels with clinical and radiological variables in MS patients. We conducted a 1-year longitudinal prospective study in MS patients with different clinical forms. We measured clinical disability using the expanded disability status scale (EDSS), the magnetic resonance imaging (MRI) volumetry baseline, and cognitive functioning using the processing speed test (PST) at baseline and 1 year later. Selected serum miRNAs and serum NfL (sNfL) levels were quantified. Seventy-three patients were recruited. MiR-126.3p correlated with EDSS and cognitive status at baseline and miR-126.3p and miR-9p correlated with cognitive deterioration at 1 year. Correlations with regional brain volumes were observed between miR-126.3p and the cortical gray matter, cerebellum, putamen, and pallidum; miR-146a.5p with the cerebellum and pallidum; miR-29b.3p with white matter and the pallidum; miR-138.5p with the pallidum; and miR-9.5p with the thalamus. sNfL was correlated with miR-9.5p. miR-146a.5p was also associated with the MS phenotype. These data justify future studies to further explore the utility of miRNAs (mirR-126.3p, miR-146.5p, and miR.9-5p) and sNfL levels as biomarkers of MS.
Collapse
Affiliation(s)
- María Inmaculada Domínguez-Mozo
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| | - Ignacio Casanova
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
- School of Medicine, Universidad Francisco de Vitoria, 28223 Madrid, Spain
- Department of Neurology, Hospital Universitario QuironSalud Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (P.L.-R.); (R.A.)
| | - Enric Monreal
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | - Lucienne Costa-Frossard
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | - Susana Sainz-de-la-Maza
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | - Raquel Sainz-Amo
- Department of Neurology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain; (E.M.); (L.C.-F.); (S.S.-d.-l.-M.); (R.S.-A.)
| | | | - Pedro Lopez-Ruiz
- Department of Neurology, Hospital Universitario QuironSalud Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (P.L.-R.); (R.A.)
| | - Laura De-Torres
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
| | - Sara Abellán
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
| | - Maria Angel Garcia-Martinez
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| | - David De-la-Cuesta
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| | - Daniel Lourido
- Department of Radiology, Hospital Universitario Ramón y Cajal, Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain;
| | - Angel Torrado-Carvajal
- Medical Image Analysis and Biometry Laboratory, Universidad Rey Juan Carlos, Móstoles, 28933 Madrid, Spain;
| | - Carol Gomez-Barbosa
- Department of Radiology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (C.G.-B.); (C.L.-V.)
| | | | - Luisa Maria Villar
- Department of Immunology, Hospital Universitario Ramón y Cajal, Red de Enfermedades Inflamatorias (REI), Instituto Ramón y Cajal de Investigación Sanitaria, Universidad de Alcalá, 28034 Madrid, Spain;
| | - Carlos López-De-Silanes
- Department of Neurology, Hospital Universitario de Torrejón, 28850 Madrid, Spain; (I.C.); (L.D.-T.); (S.A.); (C.L.-D.-S.)
| | - Rafael Arroyo
- Department of Neurology, Hospital Universitario QuironSalud Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (P.L.-R.); (R.A.)
| | - Roberto Alvarez-Lafuente
- Research Group in Environmental Factors of Neurodegenerative Diseases, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Red de Enfermedades Inflamatorias (REI), 28040 Madrid, Spain; (M.A.G.-M.); (D.D.-l.-C.); (R.A.-L.)
| |
Collapse
|
38
|
Pattnaik B, Pb S, Bhatraju N, Mittal S, Arava S, Jain D, Nayak B, Tiwari P, Hadda V, Mohan A, Agrawal A, Guleria R, Madan K. Utility of microRNA analysis in exhaled breath condensate of sarcoidosis and mediastinal tuberculosis patients: a pilot study. ERJ Open Res 2024; 10:00078-2024. [PMID: 39351385 PMCID: PMC11440382 DOI: 10.1183/23120541.00078-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 05/02/2024] [Indexed: 10/04/2024] Open
Abstract
Background Sarcoidosis and tuberculosis (TB) are the two most common causes of granulomatous mediastinal lymphadenopathy. These often exhibit overlapping clinical and radiological characteristics, rendering accurate diagnosis difficult. MicroRNA (miRNA) analysis is increasingly utilised as a potential biomarker for various diseases. Exhaled breath condensate (EBC) is a noninvasive technique for biomarker evaluation in different respiratory conditions. We attempted to identify differentially expressed miRNAs in the EBC of sarcoidosis and mediastinal TB patients. Methods EBC was obtained from subjects with a definitive diagnosis of sarcoidosis and mediastinal TB. EBC was also obtained from age- and sex-matched control subjects. From EBC, miRNA isolation, cDNA preparation and qPCR array were performed. Differentially expressed miRNAs were shortlisted. Further validation was conducted in the EBC of a new subset. Results Subjects with a definitive diagnosis of sarcoidosis (50) and TB (50), and control subjects (50) were included. qPCR array from EBC (20 subjects from each group) shortlisted eight differentially expressed miRNAs (miR-126, miR-132, miR-139-3p, miR-139-5p, miR-181c, miR-454, miR-512-3p and miR-362-5p). In the validation set (EBC of 30 subjects from each group), miR-126 and miR-132 were differentially expressed significantly. The miR-126 and miR-132 expression ratio could differentiate sarcoidosis from mediastinal TB with an AUC of 0.618 (82% specificity and 41% sensitivity). Conclusion While EBC miRNA expression is significantly and differently altered in sarcoidosis and mediastinal TB, a simple ratiometric approach failed to provide clinically useful signatures for differentiating between the two in patients with mediastinal lymphadenopathy.
Collapse
Affiliation(s)
- Bijay Pattnaik
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Sryma Pb
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Naveen Bhatraju
- Trivedi School of Biosciences, Ashoka University, Sonipat, India
| | - Saurabh Mittal
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | | | - Deepali Jain
- Department of Pathology, AIIMS, New Delhi, India
| | - Baibaswata Nayak
- Department of Gastroenterology and Human Nutrition Unit, AIIMS, New Delhi, India
| | - Pavan Tiwari
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Vijay Hadda
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anant Mohan
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Anurag Agrawal
- Trivedi School of Biosciences, Ashoka University, Sonipat, India
| | - Randeep Guleria
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| | - Karan Madan
- Department of Pulmonary, Critical Care and Sleep Medicine, All India Institute of Medical Sciences (AIIMS), New Delhi, India
| |
Collapse
|
39
|
Hasdemir PS, Celikcekic D, Oztatlici M, Ozbilgin K. Placental expressions of Anti-Mullerian hormone/Receptor, vascular endothelial growth factor and related microRNAs in patients with preeclampsia: a case control study. Biotech Histochem 2024; 99:320-329. [PMID: 39167058 DOI: 10.1080/10520295.2024.2389516] [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: 08/23/2024] Open
Abstract
Anti-Mullerian hormone (AMH) has been implicated in the pathogenesis of preeclampsia. The present study was primarily designed to determine the placental tissue AMH, Anti-Mullerian hormone Receptor II (AMHRII), vascular endothelial growth factor (VEGF) and microRNA (miRNA) 26a/126/155/210 expressions and serum miRNA 26a/126/155/210 levels in patients with preeclampsia to examine their potential role in the pathogenesis of preeclampsia. Placental tissue samples from patients with preeclampsia (n = 20) and control subjects (n = 20) were examined by immunohistochemical staining and quantitative polymerase chain reaction (qPCR) for AMH, AMHRII, VEGF mRNA expression levels and miRNA 26a/126/155/210 expressions. Serum levels of miRNA 26a/126/155/210 were measured by qPCR. Patients with preeclampsia had lower AMH/AMHRII immunostaining, particularly in syncytiotrophoblastic cells compared to control subjects (p < 0.05). The relative mRNA expressions of AMH/AMHRII were increased (1.535 ± 0.121 and 1.155 ± 0.049 fold, p < 0.0002 and p < 0.033, respectively) and the relative mRNA expression of VEGF was decreased (4.878 ± 0.331 fold, p < 0.0002) in patients with preeclampsia compared to control subjects. The miR-26a expression was increased and miR-126 expression was decreased in serum samples of patients with preeclampsia compared to control subjects (p < 0.0002). miR-155 and miR-210 expressions were increased in serum and placental tissue samples of patients with preeclampsia compared to control subjects (p < 0.0002). In conclusion, reduced placental tissue immunostaining of AMH/AMHRII along with increased AMH/AMHRII mRNA expressions may indicate posttranscriptional dysregulation. Robust increase in expressions of hypoxia/inflammation-related miRNAs particularly miR-155 and miR-210 might have a role in this mechanistic pathway. Increased serum levels of miR 26a, 155 and 210 are potential early diagnostic markers for preeclampsia.
Collapse
Affiliation(s)
- Pinar Solmaz Hasdemir
- Department of Obstetrics and Gynecology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Didem Celikcekic
- Department of Obstetrics and Gynecology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Mustafa Oztatlici
- Department of Histology and Embryology, Celal Bayar University School of Medicine, Manisa, Turkey
| | - Kemal Ozbilgin
- Department of Histology and Embryology, Celal Bayar University School of Medicine, Manisa, Turkey
| |
Collapse
|
40
|
Fujita K, Yamada M, Morishita A, Ono M, Himoto T, Kobara H, Masaki T. Cabozantinib inhibits the growth of lenvatinib-resistant hepatoma cells restoring FTCD expression. Biochem Pharmacol 2024; 226:116321. [PMID: 38815631 DOI: 10.1016/j.bcp.2024.116321] [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: 02/27/2024] [Revised: 05/18/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Cabozantinib is a newly developed tyrosine kinase inhibitor, which is applied on patients with hepatocellular carcinoma (HCC) unresponsive to conventional tyrosine kinase inhibitors, including lenvatinib. However, the mechanism of cabozantinib efficacy for lenvatinib-resistant tumor cells has not been well established in basic studies. The purpose of this study is to elucidate the mechanisms by which cabozantinib inhibits tumor growth of lenvatinib-resistant hepatocellular carcinoma cell lines in vitro and in vivo. We established a lenvatinib-resistant Hep3B cell line (Hep3B-LR) and evaluated the inhibitory effect of cabozantinib on the growth of Hep3B-LR cells. Hep3B-LR exhibited approximately 20 times greater IC50 for lenvatinib than the wild type. Compared with wild-type Hep3B, Hep3B-LR was characterized by enhanced expression of EGFR, MET and ErbB2. Cabozantinib suppressed tumor growth of Hep3B-LR in vitro and in vivo. Microarray analysis and real-time qPCR using the xenografts revealed cabozantinib downregulated miR-126-3p, a tumor suppressor miRNA, suggesting that miR-126-3p did not contribute to tumor inhibitory effect of cabozantinib. Proteome analysis using xenograft tissues demonstrated an upregulation of FTCD, a tumor suppressor gene, by cabozantinib administration. The enhanced expression of FTCD by cabozantinib was confirmed by western blot and immunohistochemistry analysis. Furthermore, FTCD expression in Hep3B-LR before cabozantinib administration was weaker than that in wild-type Hep3B. FTCD expression was weakened along with acquisition of lenvatinib-resistance, and was restored by cabozantinib administration. FTCD may be a novel therapeutic target of cabozantinib in case of lenvatinib treatment failure.
Collapse
Affiliation(s)
- Koji Fujita
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa, Japan.
| | - Mari Yamada
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa, Japan
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa, Japan
| | - Masafumi Ono
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa, Japan
| | - Takashi Himoto
- Clinical Laboratory Medicine, Kagawa Prefectural University of Health Sciences, Kagawa, Japan
| | - Hideki Kobara
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa, Japan
| | - Tsutomu Masaki
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa, Japan
| |
Collapse
|
41
|
Luo J, Wang L, Cui C, Chen H, Zeng W, Li X. MicroRNA-19a-3p inhibits endothelial dysfunction in atherosclerosis by targeting JCAD. BMC Cardiovasc Disord 2024; 24:394. [PMID: 39080547 PMCID: PMC11287888 DOI: 10.1186/s12872-024-04063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Accepted: 07/19/2024] [Indexed: 08/03/2024] Open
Abstract
OBJECTIVE To examine the influences and mechanisms of MicroRNA-19a-3p (miR-19a-3p) on endothelial dysfunction in atherosclerosis. METHODS An analysis of miR-19a expression was carried out using the Gene Expression Omnibus (GEO) database. The effect of miR-19a-3p on endothelial function in HUVECs was evaluated by miR-19a-3p overexpression under TNF-α treatment. Luciferase assays were performed to explore the potential target genes. Overexpression of junctional protein associated with coronary artery disease (JCAD) was used to examine the effects of miR-19a-3p on cell adhesion, and proliferation. RESULTS MiR-19a-3p expression in endothelial cells decreased after exposure to TNF-α and/or oscillatory flow, consistent with the expression change of miR-19a-3p found in atherosclerotic plaques. Additionally, endothelial cell dysfunction and inflammation were significantly diminished by miR-19a-3p overexpression but markedly exacerbated by miR-19a-3p inhibition. MiR-19a-3p transfection significantly decreased the expression of JCAD by binding to the 3'-UTR of JCAD mRNA. Furthermore, the protective effect of miR-19a-3p against endothelial cell dysfunction and inflammation was achieved by regulating JCAD and was closely linked to the Hippo/YAP signaling pathway. CONCLUSION MiR-19a-3p expression is a crucial molecular switch in the onset of atherosclerosis and miR-19a-3p overexpression is a possible pharmacological therapeutic strategy for reversing the development of atherosclerosis.
Collapse
Affiliation(s)
- Jinque Luo
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
- College of Pharmacy, Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, 410219, Hunan, China
| | - Ling Wang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
- College of Pharmacy, Hunan Provincial University Key Laboratory of the Fundamental and Clinical Research on Functional Nucleic Acid, Changsha Medical University, Changsha, 410219, Hunan, China
| | - Chaoyue Cui
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Hongyu Chen
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Wanli Zeng
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China
| | - Xin Li
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, "The 14th Five-Year Plan" Application Characteristic Discipline of Hunan Province (Pharmaceutical Science), College of Pharmacy, Changsha Medical University, 1501 Leifeng Avenue, Changsha, 410219, Hunan, China.
| |
Collapse
|
42
|
Terriaca S, Scioli MG, Bertoldo F, Pisano C, Nardi P, Balistreri CR, Magro D, Belmonte B, Savino L, Ferlosio A, Orlandi A. miRNA-Driven Regulation of Endothelial-to-Mesenchymal Transition Differs among Thoracic Aortic Aneurysms. Cells 2024; 13:1252. [PMID: 39120283 PMCID: PMC11312012 DOI: 10.3390/cells13151252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/16/2024] [Accepted: 07/24/2024] [Indexed: 08/10/2024] Open
Abstract
Thoracic aortic aneurysms (TAAs) represent a serious health concern, as they are associated with early aortic dissection and rupture. TAA formation is triggered by genetic conditions, in particular Marfan syndrome (MFS) and bicuspid aortic valve (BAV). During the aneurysmatic process, aortic endothelial cells can undergo endothelial-to-mesenchymal transition (End-MT) with consequent phenotypic and functional alterations. We previously documented that MFS TAA is characterized by miR-632-driven End-MT exacerbation, whereas in BAV aortopathy, the occurrence of this process remains still controversial. We investigated the End-MT process and the underlined regulatory mechanisms in BAV, TAV and MFS TAA tissues. Gene expression and immunohistochemical analysis were performed in order to analyze some important miRNAs and genes characterizing End-MT. We documented that BAV endothelium maintains the expression of the endothelial homeostasis markers, such as ERG, CD31 and miR-126-5p, while it shows lower levels of miR-632 and mesenchymal markers compared with MFS. Interestingly, we also found higher levels of miR-632 in MFS patients' blood. Our findings definitively demonstrate that the End-MT process does not characterize BAV that, among the other TAAs, better maintains the endothelial features. In addition, our results suggest miR-632 as a promising diagnostic/prognostic factor in MFS aortopathy.
Collapse
Affiliation(s)
- Sonia Terriaca
- Anatomic Pathology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (S.T.); (L.S.)
| | - Maria Giovanna Scioli
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (A.F.); (A.O.)
| | - Fabio Bertoldo
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy; (F.B.); (C.P.); (P.N.)
| | - Calogera Pisano
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy; (F.B.); (C.P.); (P.N.)
| | - Paolo Nardi
- Cardiac Surgery Unit, Department of Surgery, Tor Vergata University, 00133 Rome, Italy; (F.B.); (C.P.); (P.N.)
| | - Carmela Rita Balistreri
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy; (C.R.B.); (D.M.)
| | - Daniele Magro
- Cellular and Molecular Laboratory, Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), University of Palermo, 90134 Palermo, Italy; (C.R.B.); (D.M.)
| | - Beatrice Belmonte
- Tumor Immunology Unit, Department of Health Sciences, University of Palermo, 90134 Palermo, Italy;
- Azienda sanitaria Provinciale di Catania (ASP), 95124 Catania, Italy
| | - Luca Savino
- Anatomic Pathology, Fondazione Policlinico Tor Vergata, 00133 Rome, Italy; (S.T.); (L.S.)
| | - Amedeo Ferlosio
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (A.F.); (A.O.)
| | - Augusto Orlandi
- Anatomic Pathology, Department of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy; (A.F.); (A.O.)
- Department of Biomedical Sciences, Catholic University Our Lady of Good Counsel, 1001 Tirana, Albania
| |
Collapse
|
43
|
Mohsin F, Javaid S, Tariq M, Mustafa M. Molecular immunological mechanisms of impaired wound healing in diabetic foot ulcers (DFU), current therapeutic strategies and future directions. Int Immunopharmacol 2024; 139:112713. [PMID: 39047451 DOI: 10.1016/j.intimp.2024.112713] [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: 04/04/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
Diabetic foot ulcer (DFU) is a foremost cause of amputation in diabetic patients. Consequences of DFU include infections, decline in limb function, hospitalization, amputation, and in severe cases, death. Immune cells including macrophages, regulatory T cells, fibroblasts and other damage repair cells work in sync for effective healing and in establishment of a healthy skin barrier post-injury. Immune dysregulation during the healing of wounds can result in wound chronicity. Hyperglycemic conditions in diabetic patients influence the pathophysiology of wounds by disrupting the immune system as well as promoting neuropathy and ischemic conditions, making them difficult to heal. Chronic wound microenvironment is characterized by increased expression of matrix metalloproteinases, reactive oxygen species as well as pro-inflammatory cytokines, resulting in persistent inflammation and delayed healing. Novel treatment modalities including growth factor therapies, nano formulations, microRNA based treatments and skin grafting approaches have significantly augmented treatment efficiency, demonstrating creditable efficacy in clinical practices. Advancements in local treatments as well as invasive methodologies, for instance formulated wound dressings, stem cell applications and immunomodulatory therapies have been successful in targeting the complex pathophysiology of chronic wounds. This review focuses on elucidating the intricacies of emerging physical and non-physical therapeutic interventions, delving into the realm of advanced wound care and comprehensively summarizing efficacy of evidence-based therapies for DFU currently available.
Collapse
Affiliation(s)
- Fatima Mohsin
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| | - Sheza Javaid
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| | - Mishal Tariq
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| | - Muhammad Mustafa
- KAM School of Life Sciences, Forman Christian College (A Chartered University), Lahore, Pakistan.
| |
Collapse
|
44
|
Raju S, Turner ME, Cao C, Abdul-Samad M, Punwasi N, Blaser MC, Cahalane RM, Botts SR, Prajapati K, Patel S, Wu R, Gustafson D, Galant NJ, Fiddes L, Chemaly M, Hedin U, Matic L, Seidman M, Subasri V, Singh SA, Aikawa E, Fish JE, Howe KL. Multiomics unveils extracellular vesicle-driven mechanisms of endothelial communication in human carotid atherosclerosis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.21.599781. [PMID: 38979218 PMCID: PMC11230219 DOI: 10.1101/2024.06.21.599781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Background: Carotid atherosclerosis is orchestrated by cell-cell communication that drives progression along a clinical continuum (asymptomatic to symptomatic). Extracellular vesicles (EVs) are cell-derived nanoparticles representing a new paradigm in cellular communication. Little is known about their biological cargo, cellular origin/destination, and functional roles in human atherosclerotic plaque. Methods: EVs were enriched via size exclusion chromatography from human carotid endarterectomy samples dissected into paired plaque and marginal zones (symptomatic n=16, asymptomatic n=13). EV cargos were assessed via whole transcriptome miRNA sequencing and mass spectrometry-based proteomics. EV multi-omics were integrated with bulk and single cell RNA-sequencing (scRNA-seq) datasets to predict EV cellular origin and ligand-receptor interactions, and multi-modal biological network integration of EV-cargo was completed. EV functional impact was assessed with endothelial angiogenesis assays. Results: Carotid plaques contained more EVs than adjacent marginal zones, with differential enrichment for EV-miRNAs and EV-proteins in key atherogenic pathways. EV cellular origin analysis suggested that tissue EV signatures originated from endothelial cells (EC), smooth muscle cells (SMC), and immune cells. Integrated tissue vesiculomics and scRNA-seq indicated complex EV-vascular cell communication that changed with disease progression and plaque vulnerability (i.e., symptomatic disease). Plaques from symptomatic patients, but not asymptomatic patients, were characterized by increased involvement of endothelial pathways and more complex ligand-receptor interactions, relative to their marginal zones. Plaque-EVs were predicted to mediate communication with ECs. Pathway enrichment analysis delineated an endothelial signature with roles in angiogenesis and neovascularization - well-known indices of plaque instability. This was validated functionally, wherein human carotid symptomatic plaque EVs induced sprouting angiogenesis in comparison to their matched marginal zones. Conclusion: Our findings indicate that EVs may drive dynamic changes in plaques through EV- vascular cell communication and effector functions that typify vulnerability to rupture, precipitating symptomatic disease. The discovery of endothelial-directed angiogenic processes mediated by EVs creates new therapeutic avenues for atherosclerosis.
Collapse
|
45
|
Talevi V, Melas K, Pehlivan G, Imtiaz MA, Krüger DM, Centeno TP, Aziz NA, Fischer A, Breteler MMB. Peripheral whole blood microRNA expression in relation to vascular function: a population-based study. J Transl Med 2024; 22:670. [PMID: 39030538 PMCID: PMC11264787 DOI: 10.1186/s12967-024-05407-0] [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: 02/13/2024] [Accepted: 06/15/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND As key regulators of gene expression, microRNAs affect many cardiovascular mechanisms and have been associated with several cardiovascular diseases. In this study, we aimed to investigate the relation of whole blood microRNAs with several quantitative measurements of vascular function, and explore their biological role through an integrative microRNA-gene expression analysis. METHODS Peripheral whole blood microRNA expression was assessed through RNA-Seq in 2606 participants (45.8% men, mean age: 53.93, age range: 30 to 95 years) from the Rhineland Study, an ongoing population-based cohort study in Bonn, Germany. Weighted gene co-expression network analysis was used to cluster microRNAs with highly correlated expression levels into 14 modules. Through linear regression models, we investigated the association between each module's expression and quantitative markers of vascular health, including pulse wave velocity, total arterial compliance index, cardiac index, stroke index, systemic vascular resistance index, reactive skin hyperemia and white matter hyperintensity burden. For each module associated with at least one trait, one or more hub-microRNAs driving the association were defined. Hub-microRNAs were further characterized through mapping to putative target genes followed by gene ontology pathway analysis. RESULTS Four modules, represented by hub-microRNAs miR-320 family, miR-378 family, miR-3605-3p, miR-6747-3p, miR-6786-3p, and miR-330-5p, were associated with total arterial compliance index. Importantly, the miR-320 family module was also associated with white matter hyperintensity burden, an effect partially mediated through arterial compliance. Furthermore, hub-microRNA miR-192-5p was related to cardiac index. Functional analysis corroborated the relevance of the identified microRNAs for vascular function by revealing, among others, enrichment for pathways involved in blood vessel morphogenesis and development, angiogenesis, telomere organization and maintenance, and insulin secretion. CONCLUSIONS We identified several microRNAs robustly associated with cardiovascular function, especially arterial compliance and cardiac output. Moreover, our results highlight miR-320 as a regulator of cerebrovascular damage, partly through modulation of vascular function. As many of these microRNAs were involved in biological processes related to vasculature development and aging, our results contribute to the understanding of vascular physiology and provide putative targets for cardiovascular disease prevention.
Collapse
Affiliation(s)
- Valentina Talevi
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127, Bonn, Germany
| | - Konstantinos Melas
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127, Bonn, Germany
| | - Gökhan Pehlivan
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127, Bonn, Germany
| | - Mohammed A Imtiaz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127, Bonn, Germany
| | - Dennis Manfred Krüger
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Bioinformatics Unit, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - Tonatiuh Pena Centeno
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Bioinformatics Unit, German Center for Neurodegenerative Diseases, Göttingen, Germany
| | - N Ahmad Aziz
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127, Bonn, Germany
- Department of Neurology, Faculty of Medicine, University of Bonn, Bonn, Germany
| | - Andre Fischer
- Department for Epigenetics and Systems Medicine in Neurodegenerative Diseases, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Bioinformatics Unit, German Center for Neurodegenerative Diseases, Göttingen, Germany
- Department for Psychiatry and Psychotherapy, University Medical Center Göttingen, Göttingen, Germany
- Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, Göttingen, Germany
| | - Monique M B Breteler
- Population Health Sciences, German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Building 99, 53127, Bonn, Germany.
- Institute for Medical Biometry, Informatics and Epidemiology (IMBIE), Faculty of Medicine, University of Bonn, Bonn, Germany.
| |
Collapse
|
46
|
Li L, Wang Y, Xu Y, Xu J, Zhao Y, Cheng Z, Fang Y, Miao Y, Zhang X. ROS-scavenging lipid-based liquid crystalline as a favorable stem cell extracellular vesicles delivery vector to promote wound healing. J Control Release 2024; 371:298-312. [PMID: 38815703 DOI: 10.1016/j.jconrel.2024.05.048] [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: 02/20/2024] [Revised: 05/23/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
Wound management is a critical clinical challenge due to the dynamic and complex pathological characteristics of inflammation, proliferation, and matrix remodeling. To address this challenge, the regulation and management of this multi-stage pathological microenvironment may provide a feasible approach to wound healing. In this work, we synthesized a new lipid material (DA) with reactive oxygen species (ROS) scavenging effect to prepare DA-based liquid crystalline (DALC). Then, DALC was incorporated with adipose mesenchymal stem cells-derived extracellular vesicles (AMSC-EVs) to fabricate a novel scaffold dressing (EVs@DALC) for the treatment of the wound. DALC not only endowed EVs@DALC with ROS scavenging sites for relieving the oxidative stress and inflammation in the microenvironment of the wound site, but also facilitated cellular uptake and transfection of microRNA and growth factors contained in AMSC-EVs. Benefiting from DALC, AMSC-EVs effectively transferred microRNA and growth factors into the skin cells to induce cell proliferation and migration and accelerate angiogenesis. The results of wound healing effect in vivo indicate EVs@DALC achieved multi-stage pathological modulation for accelerating wound healing through alleviating inflammation, promoting cell proliferation and migration, and angiogenesis. Taken together, this work provides an effective strategy based on antioxidant lipid liquid crystalline delivering extracellular vesicles in treating skin wounds and paves a way for stem cell extracellular vesicles clinical translation.
Collapse
Affiliation(s)
- Lijun Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ying Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuqi Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jian Xu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yanqi Zhao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zijian Cheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuelin Fang
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China
| | - Yunqiu Miao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xinxin Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China; University of Chinese Academy of Sciences, Beijing 100049, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong 264117, China.
| |
Collapse
|
47
|
Rodrigues LDS, Felix TF, Minutentag IW, Reis PP, Bertanha M. Deciphering Key microRNA Regulated Pathways in Tissue-Engineered Blood Vessels: Implications for Vascular Scaffold Production. Int J Mol Sci 2024; 25:6762. [PMID: 38928467 PMCID: PMC11203763 DOI: 10.3390/ijms25126762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/10/2024] [Accepted: 06/16/2024] [Indexed: 06/28/2024] Open
Abstract
MicroRNAs (miRNAs) are non-coding RNAs involved in the regulation of gene expression associated with cell differentiation, proliferation, adhesion, and important biological functions such as inflammation. miRNAs play roles associated with the pathogenesis of chronic degenerative disorders including cardiovascular diseases. Understanding the influence of miRNAs and their target genes can effectively streamline the identification of key biologically active pathways that are important in the development of vascular grafts through the tissue engineering of blood vessels. To determine miRNA expression levels and identify miRNA target genes and pathways with biological roles in scaffolds that have been repopulated with adipose-derived stem cells (ASCs) generated through tissue engineering for the construction of blood vessels. miRNA quantification assays were performed in triplicate to determine miRNA expression in a total of 20 samples: five controls (natural inferior vena cava), five scaffolds recellularized with ASCs and differentiated into the endothelium (luminal layer), five samples of complete scaffolds seeded with ASCs differentiated into the endothelium (luminal layer) and smooth muscle (extraluminal layer), and five samples of ASC without cell differentiation. Several differentially expressed miRNAs were identified and predicted to modulate target genes with roles in key pathways associated with angiogenesis, vascular system control, and endothelial and smooth muscle regulation, including migration, proliferation, and growth. These findings underscore the involvement of these pathways in the regulatory mechanisms that are essential for vascular scaffold production through tissue engineering. Our research contributes to the knowledge of miRNA-regulated mechanisms, which may impact the design of vascular substitutes, and provide valuable insights for enhancing clinical practice. The molecular pathways regulated by miRNAs in tissue engineering of blood vessels (TEBV) allowed us to elucidate the main phenomena involved in cellular differentiation to constitute a blood vessel, with the main pathways being essential for angiogenesis, cellular differentiation, and differentiation into vascular smooth muscle.
Collapse
Affiliation(s)
- Lenize da Silva Rodrigues
- Department of Surgery and Orthopedics, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
- Applied Biotechnology Laboratory, Clinical Hospital of Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
- Experimental Research Unit, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (T.F.F.); (I.W.M.); (P.P.R.)
| | - Tainara Francini Felix
- Experimental Research Unit, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (T.F.F.); (I.W.M.); (P.P.R.)
| | - Iael Weissberg Minutentag
- Experimental Research Unit, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (T.F.F.); (I.W.M.); (P.P.R.)
| | - Patricia Pintor Reis
- Experimental Research Unit, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (T.F.F.); (I.W.M.); (P.P.R.)
| | - Matheus Bertanha
- Department of Surgery and Orthopedics, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil;
- Applied Biotechnology Laboratory, Clinical Hospital of Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
- Experimental Research Unit, Botucatu Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil; (T.F.F.); (I.W.M.); (P.P.R.)
| |
Collapse
|
48
|
Prajapat M, Sala L, Vidigal JA. The small non-coding RNA Vaultrc5 is dispensable to mouse development. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.01.596958. [PMID: 38895289 PMCID: PMC11185573 DOI: 10.1101/2024.06.01.596958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Vault RNAs (vRNAs) are evolutionarily conserved small non-coding RNAs transcribed by RNA polymerase lll. Initially described as components of the vault particle, they have since also been described as noncanonical miRNA precursors and as riboregulators of autophagy. As central molecules in these processes, vRNAs have been attributed numerous biological roles including regulation of cell proliferation and survival, response to viral infections, drug resistance, and animal development. Yet, their impact to mammalian physiology remains largely unexplored. To study vault RNAs in vivo, we generated a mouse line with a conditional Vaultrc5 loss of function allele. Because Vaultrc5 is the sole murine vRNA, this allele enables the characterization of the physiological requirements of this conserved class of small regulatory RNAs in mammals. Using this strain, we show that mice constitutively null for Vaultrc5 are viable and histologically normal but have a slight reduction in platelet counts pointing to a potential role for vRNAs in hematopoiesis. This work paves the way for further in vivo characterizations of this abundant but mysterious RNA molecule. Specifically, it enables the study of the biological consequences of constitutive or lineage-specific Vaultrc5 deletion and of the physiological requirements for an intact Vaultrc5 during normal hematopoiesis or in response to cellular stresses such as oncogene expression, viral infection, or drug treatment.
Collapse
Affiliation(s)
- Mahendra Prajapat
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, The National Institutes of Health, Bethesda, MD, USA
| | - Laura Sala
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, The National Institutes of Health, Bethesda, MD, USA
| | - Joana A. Vidigal
- Laboratory of Biochemistry and Molecular Biology, National Cancer Institute, The National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
49
|
Adegbola PI, Adetutu A. Genetic and epigenetic modulations in toxicity: The two-sided roles of heavy metals and polycyclic aromatic hydrocarbons from the environment. Toxicol Rep 2024; 12:502-519. [PMID: 38774476 PMCID: PMC11106787 DOI: 10.1016/j.toxrep.2024.04.010] [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: 01/05/2024] [Revised: 04/27/2024] [Accepted: 04/27/2024] [Indexed: 05/24/2024] Open
Abstract
This study emphasizes the importance of considering the metabolic and toxicity mechanisms of environmental concern chemicals in real-life exposure scenarios. Furthermore, environmental chemicals may require metabolic activation to become toxic, and competition for binding sites on receptors can affect the severity of toxicity. The multicomplex process of chemical toxicity is reflected in the activation of multiple pathways during toxicity of which AhR activation is major. Real-life exposure to a mixture of concern chemicals is common, and the composition of these chemicals determines the severity of toxicity. Nutritional essential elements can mitigate the toxicity of toxic heavy metals, while the types and ratio of composition of PAH can either increase or decrease toxicity. The epigenetic mechanisms of heavy metals and PAH toxicity involves either down-regulation or up-regulation of some non-coding RNAs (ncRNAs) whereas specific small RNAs (sRNAs) may have dual role depending on the tissue and circumstance of expression. Similarly, decrease DNA methylation and histone modification are major players in heavy metals and PAH mediated toxicity and FLT1 hypermethylation is a major process in PAH induced carcinogenesis. Overall, this review provides the understanding of the metabolism of environmental concern chemicals, emphasizing the importance of considering mixed compositions and real-life exposure scenarios in assessing their potential effects on human health and diseases development as well as the dual mechanism of toxicity via genetic or epigenetic axis.
Collapse
Affiliation(s)
- Peter Ifeoluwa Adegbola
- Department of Biochemistry and Forensic Science, First Technical University, Ibadan, Nigeria
| | - Adewale Adetutu
- Department of Biochemistry, Faculty of Basic Medical Sciences, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| |
Collapse
|
50
|
Liao HJ, Yang YP, Liu YH, Tseng HC, Huo TI, Chiou SH, Chang CH. Harnessing the potential of mesenchymal stem cells-derived exosomes in degenerative diseases. Regen Ther 2024; 26:599-610. [PMID: 39253597 PMCID: PMC11382214 DOI: 10.1016/j.reth.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 09/11/2024] Open
Abstract
Mesenchymal stem cells (MSCs) have gained attention as a promising therapeutic approach in both preclinical and clinical osteoarthritis (OA) settings. Various joint cell types, such as chondrocytes, synovial fibroblasts, osteoblasts, and tenocytes, can produce and release extracellular vesicles (EVs), which subsequently influence the biological activities of recipient cells. Recently, extracellular vesicles derived from mesenchymal stem cells (MSC-EVs) have shown the potential to modulate various physiological and pathological processes through the modulation of cellular differentiation, immune responses, and tissue repair. This review explores the roles and therapeutic potential of MSC-EVs in OA and rheumatoid arthritis, cardiovascular disease, age-related macular degeneration, Alzheimer's disease, and other degenerative diseases. Notably, we provide a comprehensive summary of exosome biogenesis, microRNA composition, mechanisms of intercellular transfer, and their evolving role in the highlight of exosome-based treatments in both preclinical and clinical avenues.
Collapse
Affiliation(s)
- Hsiu-Jung Liao
- Department of Medical Research, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Institute of Biopharmaceutical Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Ping Yang
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Yu-Hao Liu
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Huan-Chin Tseng
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Teh-Ia Huo
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
- Institute of Pharmacology, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Chih-Hung Chang
- Department of Orthopedic Surgery, Far Eastern Memorial Hospital, New Taipei City, Taiwan
- Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan City, Taiwan
| |
Collapse
|