1
|
Osorio-Antonio F, Diaz-González DM, Bautista-Rodríguez E, Campos-Viguri GE, Peralta-Zaragoza O, Sánchez-López JM, Cortez-Sánchez JL, Muñoz-Olivos C, Castelán F, Velázquez-Orozco V. LRG1 in cancer: mechanisms, potential therapeutic target, and use in clinical diagnosis. Mol Biol Rep 2025; 52:577. [PMID: 40493117 DOI: 10.1007/s11033-025-10669-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Accepted: 05/30/2025] [Indexed: 06/12/2025]
Abstract
Leucine-rich alpha-2 glycoprotein 1 (LRG1) is an extracellular protein whose elevated expression has been linked to cancers with poor prognosis and low survival rates. Given its significance as a biomarker, this manuscript explores the role of LRG1 in cancer, as several studies have identified it as a promoter of proliferation, angiogenesis, invasion, and metastasis in breast, ovarian, pancreatic, and neuroblastoma cancers. However, contrasting evidence suggests that LRG1 inhibits proliferation in hepatocellular carcinoma cells and suppresses migration and invasion in esophageal squamous cell carcinoma. We also examine its regulation and the associated signaling pathways, including TGF-β and PI3K/AKT/mTOR.
Collapse
Affiliation(s)
- Federico Osorio-Antonio
- Children's Hospital of Tlaxcala, Tlaxcala, 90610, Mexico
- Faculty of Biotechnology, Popular Autonomous University of the State of Puebla, Puebla City, 72410, Mexico
| | - Daniela Michel Diaz-González
- Department of Physiology, Biophysics, and Neurosciences, Center for Research and Advanced Studies of the IPN, Mexico City, 07360, Mexico
| | - Elizabeth Bautista-Rodríguez
- Clinical Chemistry, Faculty of Health Sciences, Autonomous University of Tlaxcala, Tlaxcala, 90750, Mexico.
- Facultad de Ciencias de la Salud, Universidad Autónoma de Tlaxcala, Tlaxcala, 90750, Mexico.
| | - Gabriela Elizabeth Campos-Viguri
- Center for Research in Applied Science and Advanced Technology, Morelos Unit, National Polytechnic Institute, Boulevard de la Technology, 1036 Z-1, P 2/2, Atlacholoaya, Morelos, 62790, Mexico
| | - Oscar Peralta-Zaragoza
- Direction of Chronic Infections and Cancer, Research Center in Infection Diseases, Instituto Nacional de Salud Pública, Cuernavaca, 62100, Mexico
| | | | - José Luis Cortez-Sánchez
- Environmental Epigenetics and Mental Health Laboratory, Faculty of Chemical Biological Sciences, Autonomous University of Campeche, Campeche, Mexico
| | - Cristina Muñoz-Olivos
- Faculty of Biotechnology, Popular Autonomous University of the State of Puebla, Puebla City, 72410, Mexico
- Department of Sciences and Engineering, Iberoamerican Puebla University, Puebla, 71820, Mexico
| | - Francisco Castelán
- Tlaxcala Center for Behavioral Biology, Autonomous University of Tlaxcala, Tlaxcala, 90070, Mexico
- Department of Biology and Cellular Physiology, Institute of Biomedical Research, National Autonomous University of Mexico, Federal Highway Tlaxcala-Puebla km 1.5 s/n, Tlaxcala, 90070, Mexico
| | - Verónica Velázquez-Orozco
- Clinical Chemistry, Faculty of Health Sciences, Autonomous University of Tlaxcala, Tlaxcala, 90750, Mexico
| |
Collapse
|
2
|
Mao J, Xu Y, Wang W, Deng X, Hui Y, Rui M, Tang J, Wang W, Huang Y, Wu L, Xi K, Zhu Y, Gu Y, Chen L. Topological cues of microparticles train stem cells for tissue repair via mechanotransduction. Bioact Mater 2025; 48:531-549. [PMID: 40114729 PMCID: PMC11923629 DOI: 10.1016/j.bioactmat.2025.02.032] [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: 10/22/2024] [Revised: 02/15/2025] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Microspheres (MPs) and porous microspheres (PMPs) are the two most widely used microparticles in tissue engineering and stem cell therapy. However, how stem cells perceive the topological differences between them to regulate cell function remains to be unclear. Here, we systematically studied the changes in stem cell function under the action of MPs and PMPs and elucidated the related mechanisms. Our findings show that the porous structure of PMPs can be sensed by focal adhesions (FAs), which triggers the synthesis of F-actin to inhibit the phosphorylation and degradation of Yes-associated protein (YAP), while also transmitting stress to the nucleus through the contraction of F-actin, thereby enhancing the nuclear translocation of YAP protein. The activation of YAP significantly enhances the proliferation, osteogenesis, paracrine and glucose metabolism of BMSCs, making them exhibit stronger bone repair ability in both in vivo and in vitro experiments. In summary, this study provides a comprehensive and reliable understanding of the behavior of BMSCs in response to MPs and PMPs. It also deepens our understanding of the association between microparticles' topological cues and biological functions, which will provide valuable guidance for the construction of bone tissue engineering (BTE) scaffolds.
Collapse
Affiliation(s)
- Jiannan Mao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yichang Xu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Wenbo Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Xiongwei Deng
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yujian Hui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Min Rui
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Jincheng Tang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Wei Wang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yiyang Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Liang Wu
- Division of Spine Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, PR China
| | - Kun Xi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Yunrong Zhu
- Department of Orthopaedics, Wuxi Key Laboratory of Biomaterials for Clinical Application, Department of Central Laboratory, Jiangyin Clinical College of Xuzhou Medical University, No.163 Shoushan Road, Jiang Yin, 214400, PR China
| | - Yong Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| | - Liang Chen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Orthopedic Institute, Soochow University, 188 Shizi Road, Suzhou, Jiangsu, 215006, PR China
| |
Collapse
|
3
|
Eldeen S, Ramirez AFG, Keresteci B, Chang PD, Botvinick EL. Label-Free Prediction of Fluorescently Labeled Fibrin Networks. Biomater Res 2025; 29:0211. [PMID: 40438124 PMCID: PMC12117218 DOI: 10.34133/bmr.0211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 04/07/2025] [Accepted: 04/26/2025] [Indexed: 06/01/2025] Open
Abstract
While fluorescent labeling has been the standard for visualizing fibers within fibrillar scaffold models of the extracellular matrix (ECM), the use of fluorescent dyes can compromise cell viability and photobleach prematurely. The intricate fibrillar composition of ECM is crucial for its viscoelastic properties, which regulate intracellular signaling and provide structural support for cells. Naturally derived biomaterials such as fibrin and collagen replicate these fibrillar structures, but longitudinal confocal imaging of fibers using fluorescent dyes may impact cell function and photobleach the sample long before termination of the experiment. An alternative technique is reflection confocal microscopy (RCM) that provides high-resolution images of fibers. However, RCM is sensitive to fiber orientation relative to the optical axis, and consequently, many fibers are not detected. We aim to recover these fibers. Here, we propose a deep learning tool for predicting fluorescently labeled optical sections from unlabeled image stacks. Specifically, our model is conditioned to reproduce fluorescent labeling using RCM images at 3 laser wavelengths and a single laser transmission image. The model is implemented using a fully convolutional image-to-image mapping architecture with a hybrid loss function that includes both low-dimensional statistical and high-dimensional structural components. Upon convergence, the proposed method accurately recovers 3-dimensional fibrous architecture without substantial differences in fiber length or fiber count. However, the predicted fibers were slightly wider than original fluorescent labels (0.213 ± 0.009 μm). The model can be implemented on any commercial laser scanning microscope, providing wide use in the study of ECM biology.
Collapse
Affiliation(s)
- Sarah Eldeen
- Department of Mathematical, Computational, and Systems Biology,
University of California, Irvine, Irvine, CA, USA
| | - Andres Felipe Guerrero Ramirez
- Department of Mathematical, Computational, and Systems Biology,
University of California, Irvine, Irvine, CA, USA
- Department of Radiological Sciences and Computer Sciences,
University of California, Irvine, Irvine, CA, USA
| | - Bora Keresteci
- Department of Biomedical Engineering,
University of California, Irvine, Irvine, CA, USA
| | - Peter D. Chang
- Department of Radiological Sciences and Computer Sciences,
University of California, Irvine, Irvine, CA, USA
| | - Elliot L. Botvinick
- Department of Biomedical Engineering,
University of California, Irvine, Irvine, CA, USA
- Beckman Laser Institute and Medical Clinic,
University of California, Irvine, Irvine, CA, USA
- Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center,
University of California, Irvine, Irvine, CA, USA
- Department of Surgery,
University of California, Irvine, Irvine, CA, USA
| |
Collapse
|
4
|
Davidson SM, Andreadou I, Antoniades C, Bartunek J, Basso C, Brundel BJJM, Byrne RA, Chiva-Blanch G, da Costa Martins P, Evans PC, Girão H, Giricz Z, Gollmann-Tepeköylü C, Guzik T, Gyöngyösi M, Hübner N, Joner M, Kleinbongard P, Krieg T, Liehn E, Madonna R, Maguy A, Paillard M, Pesce M, Petersen SE, Schiattarella GG, Sluijter JPG, Steffens S, Streckfuss-Bömeke K, Thielmann M, Tucker A, Van Linthout S, Wijns W, Wojta J, Wu JC, Perrino C. Opportunities and challenges for the use of human samples in translational cardiovascular research: a scientific statement of the ESC Working Group on Cellular Biology of the Heart, the ESC Working Group on Cardiovascular Surgery, the ESC Council on Basic Cardiovascular Science, the ESC Scientists of Tomorrow, the European Association of Percutaneous Cardiovascular Interventions of the ESC, and the Heart Failure Association of the ESC. Cardiovasc Res 2025; 121:702-729. [PMID: 40084813 PMCID: PMC12101359 DOI: 10.1093/cvr/cvaf023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/23/2024] [Accepted: 10/21/2024] [Indexed: 03/16/2025] Open
Abstract
Animal models offer invaluable insights into disease mechanisms but cannot entirely mimic the variability and heterogeneity of human populations, nor the increasing prevalence of multi-morbidity. Consequently, employing human samples-such as whole blood or fractions, valvular and vascular tissues, myocardium, pericardium, or human-derived cells-is essential for enhancing the translational relevance of cardiovascular research. For instance, myocardial tissue slices, which preserve crucial structural and functional characteristics of the human heart, can be used in vitro to examine drug responses. Human blood serves as a rich source of biomarkers, including extracellular vesicles, various types of RNA (miRNA, lncRNA, and circRNAs), circulating inflammatory cells, and endothelial colony-forming cells, facilitating detailed studies of cardiovascular diseases. Primary cardiomyocytes and vascular cells isolated from human tissues are invaluable for mechanistic investigations in vitro. In cases where these are unavailable, human induced pluripotent stem cells serve as effective substitutes, albeit with specific limitations. However, the use of human samples presents challenges such as ethical approvals, tissue procurement and storage, variability in patient genetics and treatment regimens, and the selection of appropriate control samples. Biobanks are central to the efficient use of these scarce and valuable resources. This scientific statement discusses opportunities to implement the use of human samples for cardiovascular research within specific clinical contexts, offers a practical framework for acquiring and utilizing different human materials, and presents examples of human sample applications for specific cardiovascular diseases, providing a valuable resource for clinicians, translational and basic scientists engaged in cardiovascular research.
Collapse
Affiliation(s)
- Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, UK
| | - Ioanna Andreadou
- School of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Charalambos Antoniades
- RDM Division of Cardiovascular Medicine, Acute Multidisciplinary Imaging and Interventional Centre, University of Oxford, Headley Way, Headington, Oxford OX3 9DU, UK
| | - Jozef Bartunek
- Cardiovascular Center Aalst, OLV Hospital, Aalst, Belgium
| | - Cristina Basso
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, Cardiovascular Pathology, University of Padua, Padua, Italy
| | - Bianca J J M Brundel
- Physiology, Amsterdam UMC Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam, The Netherlands
| | - Robert A Byrne
- Cardiovascular Research Institute Dublin, Mater Private Network, Dublin, Ireland
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Gemma Chiva-Blanch
- Faculty of Health Sciences, Universitat Oberta de Catalunya, Barcelona, Spain
- Department of Endocrinology and Nutrition, August Pi i Sunyer Biomedical Research Institute, Hospital Clínic of Barcelona, Barcelona, Spain
- Biomedical Network Research Centre on Obesity and Nutrition Physiopathology, Instituto de Salud Carlos III, Madrid, Spain
| | - Paula da Costa Martins
- Department of Molecular Genetics, Faculty of Sciences and Engineering, Maastricht, The Netherlands
- CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paul C Evans
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Henrique Girão
- Center for Innovative Biomedicine and Biotechnology, Clinical Academic Centre of Coimbra, Faculty of Medicine, University of Coimbra, Coimbra Institute for Clinical and Biomedical Research, Coimbra, Portugal
| | - Zoltan Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Can Gollmann-Tepeköylü
- Department for Cardiac Surgery, Cardiac Regeneration Research, Medical University of Innsbruck, Anichstraße 35 A, 6020 Innsbruck, Austria
| | - Tomasz Guzik
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Mariann Gyöngyösi
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Norbert Hübner
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
- Charite-Universitätsmedizin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
| | - Michael Joner
- Department of Cardiology, German Heart Center Munich, Technical University of Munich, Lazarettstrasse 36, 80636 Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Petra Kleinbongard
- Faculty of Medicine University of Duisburg-Essen, Institute of Pathophysiology, Duisburg-Essen, Germany
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Elisa Liehn
- Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Rosalinda Madonna
- Cardiology Division, Department of Pathology, University of Pisa, Pisa, Italy
| | - Ange Maguy
- Department of Physiology, University of Bern, Bern, Switzerland
| | - Melanie Paillard
- Laboratoire CarMeN—IRIS Team, INSERM, INRA, Université Claude Bernard Lyon-1, Univ-Lyon, 69500 Bron, France
| | - Maurizio Pesce
- Unità di Ingegneria Tissutale Cardiovascolare, Centro Cardiologico Monzino, IRCCS, Milan, Italy
- Department of Aerospace and Mechanical Engineering, Politecnico di Torino, Italy
- Department of Cell Biology, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Steffen E Petersen
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, UK
- Health Data Research UK, London, UK
- Alan Turing Institute, London, UK
| | - Gabriele G Schiattarella
- German Center for Cardiovascular Research (DZHK), partner site Berlin, Berlin, Germany
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
- Deutsches Herzzentrum der Charité (DHZC), Charité-Universitätsmedizin Berlin, Berlin, Germany
- Translational Approaches in Heart Failure and Cardiometabolic Disease, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Joost P G Sluijter
- Department of Cardiology, Laboratory of Experimental Cardiology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Sabine Steffens
- Institute for Cardiovascular Prevention, Ludwig-Maximilians-Universität, Munich, Germany
- German Center for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
| | - Katrin Streckfuss-Bömeke
- Institute of Pharmacology and Toxicology, University of Würzburg, Würzburg, Germany
- Clinic for Cardiology and Pneumology, University Medicine Göttingen, Germany and German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Matthias Thielmann
- West-German Heart and Vascular Center, University Duisburg-Essen, Essen, Germany
| | - Art Tucker
- William Harvey Research Institute, NIHR Barts Biomedical Research Centre, Queen Mary University London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, UK
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité, BIH Center for Regenerative Therapies, Universitätmedizin Berlin, Berlin, Germany
- Max Delbrück Center in the Helmholtz Association, Berlin, Germany
| | - William Wijns
- The Lambe Institute for Translational Research and Curam, University of Galway, Galway, Ireland
| | - Johann Wojta
- Department of Internal Medicine II, Medical University of Vienna, Vienna, Austria
- Core Facilities, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Cardiovascular Research, Vienna, Austria
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford, CA, USA
| | - Cinzia Perrino
- Department of Advanced Biomedical Sciences, Federico II University, Via Pansini 5, 80131 Naples, Italy
| |
Collapse
|
5
|
Stefanelli V, Lombardi J, Ferrer J, Gardocki-Sandor M. Vascularization of Human Acellular Dermal Matrices: A Comparative Study in a Nonhuman Primate Model. Tissue Eng Part A 2025; 31:419-432. [PMID: 39041614 DOI: 10.1089/ten.tea.2024.0059] [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: 07/24/2024] Open
Abstract
Four human acellular dermal matrices (hADMs) were characterized in a nonhuman primate abdominal wall repair model by evaluating host immune response, vascularization, and incorporation into host tissues. AlloDerm™ (electron beam-sterilized hADM [e-hADM]), AlloMax™ (gamma beam-sterilized hADM, freeze-dried [g-hADM-FD]), DermaMatrix™ (hADM, freeze-dried [hADM-FD]), and FlexHD™ (ethanol-treated hADM [EtOH-hADM]) were each implanted in an abdominal wall-bridging defect in nonhuman primates (n = 3 animals/time point, n = 36 animals). Immunohistochemical and histological assessments were conducted on biopsies from each hADM at 1-, 3-, and 6-months postimplantation to assess vascularization (hematoxylin and eosin [H&E], CD31, alpha smooth muscle actin [αSMA], collagen IV), inflammatory/immune response (H&E, CD3, CD20, CD68), and collagen turnover (H&E, matrix metalloproteinase-9 [MMP-9]). MMP-9 immunolabeling was similar among different hADMs at 1 month; however, hADM-FD and EtOH-hADM showed higher total mean MMP-9-immunopositive areas at approximately 16% compared with <1% for e-hADM and g-hADM at 6 months postimplantation. Cells that stained positively for CD68, CD3, and CD20 were generally higher for hADM-FD and EtOH-hADM compared with other hADMs. The mean CD31-immunopositive area, CD31 vessel density, CD31 vessel diameter, and collagen IV-immunopositive area increased over time. Among all the hADM types, e-hADM had the highest mean (±standard deviation [SD]) CD31-immunopositive area at 1.54% ± 1.01%, vessel density at 7.86 × 10-5 ± 3.96 × 10-5 vessels/µm2, and collagen IV-immunopositive area at 2.55% ± 0.73% 1-month postimplantation. The pattern of αSMA immunolabeling varied among the hADMs. Histology showed that overall inflammation was mild at 1 month. Overall fibroblast repopulation and collagen remodeling increased over time from 1 to 6 months postimplantation. Fibroblast infiltration was minimal to mild at 1 month, with e-hADM showing the highest mean (±SD) score at 2.00 ± 0.00 compared with other hADMs. Only hADM-FD was not completely replaced by neotissue formation at 6 months postimplantation. All hADMs promoted vascularization, cell infiltration, and incorporation into host tissue, which were associated with acute inflammation and immune responses, within a 6-month period. A trend toward relatively enhanced early vascularization in e-hADM compared with other hADMs was observed. Immunogenic responses among the hADMs in the present study showed a slight distinction toward more quiescent terminally sterilized hADMs (e-hADM, g-hADM-FD) versus aseptically processed hADMs (EtOH-hADM, hADM-FD).
Collapse
Affiliation(s)
| | - Jared Lombardi
- Allergan Aesthetics, an AbbVie Company, Branchburg, New Jersey, USA
| | - Joselito Ferrer
- Allergan Aesthetics, an AbbVie Company, Branchburg, New Jersey, USA
| | | |
Collapse
|
6
|
He B, Wood KH, Li ZJ, Ermer JA, Li J, Bastow ER, Sakaram S, Darcy PK, Spalding LJ, Redfern CT, Canes J, Oliveira M, Prat A, Cortes J, Thompson EW, Littlefield BA, Redfern A, Ganss R. Selective tubulin-binding drugs induce pericyte phenotype switching and anti-cancer immunity. EMBO Mol Med 2025; 17:1071-1100. [PMID: 40140727 DOI: 10.1038/s44321-025-00222-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
The intratumoral immune milieu is crucial for the success of anti-cancer immunotherapy. We show here that stromal modulation by the tubulin-binding anti-cancer drugs combretastatin A4 (CA-4) and eribulin improved tumor perfusion and anti-tumor immunity. This was achieved by reverting highly proliferative, angiogenic pericytes into a quiescent, contractile state which durably normalized the vascular bed and reduced hypoxia in mouse models of pancreatic neuroendocrine cancer, breast cancer and melanoma. The crucial event in pericyte phenotype switching was RhoA kinase activation, which distinguished CA-4 and eribulin effects from other anti-mitotic drugs such as paclitaxel and vinorelbine. Importantly, eribulin pre-treatment sensitized tumors for adoptive T cell therapy or checkpoint inhibition resulting in effector cell infiltration and better survival outcomes in mice. In breast cancer patients, eribulin neoadjuvant treatment induced pericyte maturity and RhoA kinase activity indicating similar vessel remodeling effects as seen in mice. Moreover, a contractile pericyte signature was associated with overall better survival outcome in two independent breast cancer cohorts. This underscores the potential of re-purposing specific anti-cancer drugs to enable synergistic complementation with emerging immunotherapies.
Collapse
Affiliation(s)
- Bo He
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Kira H Wood
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Zhi-Jie Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Guangdong, P. R. China
| | - Judith A Ermer
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Ji Li
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Edward R Bastow
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | | | - Phillip K Darcy
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia. Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, VIC, Australia
| | - Lisa J Spalding
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Cameron T Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
| | - Jordi Canes
- SOLTI Cancer Research Group, Barcelona, Spain
| | - Mafalda Oliveira
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Vall d'Hebron University Hospital, Barcelona, Spain
- Breast Cancer Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Aleix Prat
- SOLTI Cancer Research Group, Barcelona, Spain
- Medical Oncology Department, Hospital Clinic of Barcelona, Barcelona, Spain
- Translational Genomics and Targeted Therapies in Solid Tumors, August Pi i Sunyer Biomedical Research Institute (IDIBAPS), Barcelona, Spain
- Faculty of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Javier Cortes
- SOLTI Cancer Research Group, Barcelona, Spain
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Jersey City, NJ, USA
- Medica Scientia Innovation Research (MEDSIR)-Oncoclínicas&Co, Sao Paulo, Brazil
- International Breast Cancer Center (IBCC), Pangaea Oncology, Quiron Group, Barcelona, Spain
- Faculty of Biomedical and Health Sciences, Department of Medicine, Universidad Europea de Madrid, Madrid, Spain
- IOB Madrid, Institute of Oncology, Hospital Beata María Ana, Madrid, Spain
| | - Erik W Thompson
- School of Biomedical Sciences and Centre for Genomics and Personalised Health, Faculty of Health, Queensland University of Technology (QUT) and Translational Research Institute, Brisbane, Australia
| | | | - Andrew Redfern
- Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia
- Fiona Stanley Hospital, Perth, WA, Australia
| | - Ruth Ganss
- Cancer Microenvironment Laboratory, Harry Perkins Institute of Medical Research, Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.
| |
Collapse
|
7
|
Gao R, Zhang X, Ju H, Zhou Y, Yin L, Yang L, Wu P, Sun X, Fang H. Telocyte-derived exosomes promote angiogenesis and alleviate acute respiratory distress syndrome via JAK/STAT-miR-221-E2F2 axis. MOLECULAR BIOMEDICINE 2025; 6:21. [PMID: 40198510 PMCID: PMC11979044 DOI: 10.1186/s43556-025-00259-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 02/27/2025] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
Acute respiratory distress syndrome (ARDS) is characterized by severe respiratory failure and significant inflammation, leading to vascular and epithelial cell damage. The absence of effective pharmacologic treatments underscores the need for novel therapeutic approaches. Telocytes (TCs), a newly identified type of interstitial cells, have shown potential in tissue repair and angiogenesis, particularly through the release of exosomal microRNAs (miRNAs). Exosomes were isolated from LPS (lipopolysaccharide)-stimulated TCs and characterized using western blotting and nanoparticle tracking analysis. The role of exosomal miR-221 in angiogenesis was assessed through tube formation, migration, and proliferation assays in mouse vascular endothelial cells (MVECs). The JAK/STAT pathway's involvement in miR-221 regulation was determined using western blotting and qRT-PCR. A dual-luciferase assay confirmed E2F2 as a direct target of miR-221. ARDS mouse model was established via LPS instillation, and the therapeutic effects of TCs-derived exosomes were evaluated by histopathological scoring, cytokine analysis, and endothelial barrier integrity assays. Our findings demonstrated that exosomes from LPS-stimulated TCs significantly promoted angiogenesis, proliferation, and migration in MVECs. These effects were mediated by miR-221, which downregulated E2F2 expression, an important regulator of endothelial cell functions. The JAK/STAT pathway played a crucial role in miR-221 production, with pathway inhibition reducing miR-221 levels and attenuating its pro-angiogenic effects. In vivo, TCs-derived exosomes reduced lung inflammation and tissue damage in ARDS mice, effects that were reversed by miR-221 inhibition. These results suggested that TCs-derived exosomes promoted angiogenesis and alleviated ARDS through the JAK/STAT-miR-221-E2F2 axis.
Collapse
Affiliation(s)
- Rongrong Gao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Clinical Center for Biotherapy at Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xu Zhang
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Huihui Ju
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yile Zhou
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Luoyue Yin
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Liuke Yang
- College of Plant Protection, Nanjing Agricultural University, Nanjing, 210000, China
| | - Pinwen Wu
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
| | - Xia Sun
- Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai, China.
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
- Department of Anesthesiology, Shanghai Geriatric Medical Center, Shanghai, China.
| |
Collapse
|
8
|
Vergroesen TM, Vermeulen V, Merks RMH. Falsifying computational models of endothelial cell network formation through quantitative comparison with in vitro models. PLoS Comput Biol 2025; 21:e1012965. [PMID: 40305554 PMCID: PMC12074657 DOI: 10.1371/journal.pcbi.1012965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 05/13/2025] [Accepted: 03/14/2025] [Indexed: 05/02/2025] Open
Abstract
During angiogenesis, endothelial cells expand the vasculature by migrating from existing blood vessels, proliferating and collectively organizing into new capillaries. In vitro and in vivo experimentation is instrumental for identifying the molecular players and cell behaviour that regulate angiogenesis. Alongside experimental work, computational and mathematical models of endothelial cell network formation have helped to analyse if the current molecular and cellular understanding of endothelial cell behaviour is sufficient to explain the formation of endothelial cell networks. As input, the models take (a subset of) the current knowledge or hypotheses of single cell behaviour and capture it into a dynamical, mathematical description. As output, they predict the multicellular behaviour following from the actions of many individual cells, i.e., formation of a vascular-like network. Paradoxically, computational modelling based on different assumptions, i.e., completely different, sometimes non-intersecting sets of observed single cell behaviour, can reproduce the same angiogenesis-like multicellular behaviour, making it practically impossible to decide which, if any, of these models is correct. Here we present dynamical analyses of time-lapses of in vitro endothelial cell network formation experiments and compare these with dynamic analyses of three mathematical models: (1) the cell elongation model; (2) the contact-inhibited chemotaxis model; and (3) the mechanical cell-cell communication model. We extract a variety of dynamical characteristics of endothelial cell network formation using a custom time-lapse video analysis pipeline in ImageJ. We compare the dynamical network characteristics of the in vitro experiments to those of the cellular networks produced by the computational models. We test the response of the in silico dynamical cell network characteristics to changes in cell density and make related changes in the in vitro experiments. Of the three computational models that we have considered, the cell elongation model best captures the remodelling phase of in vitro endothelial cell network formation. Furthermore, in the in vitro model, the final size and number of lacunae in the network are independent of the initial cell density. This observation is also reproduced in the cell elongation model, but not in the other two models that we have considered. Altogether, we present an approach to model validation based on comparisons of time-resolved data and variations of model conditions.
Collapse
Affiliation(s)
| | - Vincent Vermeulen
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
| | - Roeland M. H. Merks
- Institute of Biology Leiden, Leiden University, Leiden, The Netherlands
- Mathematical Institute, Leiden University, Leiden, The Netherlands
| |
Collapse
|
9
|
Kretschmer M, Vollmar AM, Zahler S. Endothelial Sprout Formation Is Regulated by Substrate Stiffness and Notch Signaling. Int J Mol Sci 2025; 26:3155. [PMID: 40243923 PMCID: PMC11988845 DOI: 10.3390/ijms26073155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/18/2025] Open
Abstract
Angiogenesis, the process of vessel formation from pre-existing ones, is modulated by the local stiffness of the extracelluar matrix. We have previously shown that Notch signaling, a key pathway in angiogenesis, responds to substrate stiffness in endothelial cells. In the current work, we investigate the contribution of Notch signaling in angiogenesis-related in vitro assays by using VEGF and Notch inhibitors as perturbations. In addition, we investigate Notch signaling in relation to the stiffness of the respective endothelial microenvironment. While the tube formation assay on Matrigel is clearly influenced by substrate stiffness, Notch signaling seems to play no major role in this context. In contrast, spheroid sprouting is influenced by stiffness as well as Notch signaling; with decreasing stiffness, both sprouting and Notch signaling are increased. This finding adds a functional aspect to the mechanosensitivity of Notch signaling.
Collapse
Affiliation(s)
| | | | - Stefan Zahler
- Department of Pharmacy, Ludwig-Maximilians-University, Butenandtstr. 5, 81377 Munich, Germany; (M.K.)
| |
Collapse
|
10
|
Janani G, Girigoswami A, Deepika B, Udayakumar S, Mercy DJ, Girigoswami K. Dual Mechanism of Amphiroa anceps: Antiangiogenic and Anticancer Effects in Skin Cancer. Chem Biodivers 2025:e202500626. [PMID: 40106265 DOI: 10.1002/cbdv.202500626] [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: 02/20/2025] [Revised: 03/19/2025] [Accepted: 03/19/2025] [Indexed: 03/22/2025]
Abstract
Skin cancer is diagnosed usually at the last stage and the available conventional treatments are costly and have various side-effects. It is therefore necessary to design a biocompatible alternative treatment approach. In our present study, the liposomal formulation of marine red algae Amphiroa anceps is investigated for its anticancer activity against skin cancer. The aqueous extract (HA) and the liposomal formulated aqueous extract (NHA) of A. anceps are characterized using Raman spectroscopy, high-performance liquid chromatography (HPLC) and scanning electron microscopy (SEM). SEM reveals the size of NHA to be around 150-230 nm. In HPLC, two prominent peaks at the retention time of 5.233 min and 5.775 min are observed. NHA exhibits anticancer activity in skin cancer cells (A375) dose-dependently, and the cell viability 100 µg/mL is 16% ± 2%. The in vitro biocompatibility assays using fibroblast cell viability and haemolysis assay, show that the NHA is safe and highly biocompatible. A step forward, the Chorioallantoic Membrane Assay assay reveals HA and NHA have anti-angiogenesis activity. Our study elucidates that NHA induces anticancer activity by cell necrosis and reducing new blood vessel formation, and can be a safe and promising therapeutic agent for cancer management.
Collapse
Affiliation(s)
- Gopalarethinam Janani
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Chennai, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Chennai, India
| | - Balasubramanian Deepika
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Chennai, India
| | - Saranya Udayakumar
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Chennai, India
| | - Devadass Jessy Mercy
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Chettinad Health City, Chennai, India
| | - Koyeli Girigoswami
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| |
Collapse
|
11
|
Dasgupta S, Barui A. Coculture to vascularization transition in bioengineered skin grafts through VEGF-associated pathways tracked by exosomal biomarkers. Biomater Sci 2025; 13:1464-1481. [PMID: 39902980 DOI: 10.1039/d4bm01544d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Inadequate vasculature poses a significant challenge in the clinical translation of tissue engineering constructs. Current strategies for vascularization typically recruit short-lived endothelial cells or induce mesenchymal stem cells (MSC) to differentiate into the endothelial lineage, often in combination with supporting pericytes or fibroblasts. However, endothelial-associated cocultures lack adaptive ability and form limited vasculature. In this study, we investigated the endothelial transdifferentiation of an MSC-fibroblast coculture loaded on a bioengineered graft and utilized the exosomes released by the coculture model as a biomarker to monitor the progress of vascularization inside the graft. To develop the pre-vascularized skin graft, dermal fibroblasts and MSC were seeded on a biocomposite chitosan/collagen/fibrinogen/D3 (CCF-D3) scaffold. The cocultured graft facilitated the differentiation of MSC to endothelial cells (MEnDoT). Additionally, it promoted vasculogenic sprouting through the VEGF-eNOS pathways, as evidenced by the expression of F-actin, VEGF-A, and downstream transcriptomic markers (CD31, CD34, eNOS, VEGF-A, VEGF-R2, PI3 K, and PLC-γ). Exosomes (∼130 nm diameter) were isolated from the coculture, and their spectral analysis revealed significant differences (p < 0.05) in the intensity ratio of nucleotides (952 cm-1), polysaccharides (1071 cm-1) and lipoproteins (1417 cm-1), corresponding to vasculogenesis. The activation of the VEGF-associated pathway in the coculture model was validated using an inhibitor (dexamethasone), which was used to treat the coculture graft as a control. Thus, this study elucidated the vascularization of coculture constructs via the VEGF-associated pathway. It demonstrated the potential of exosome spectral fingerprints as promising biomarkers to monitor the vascularization progression inside the graft, paving the way for the development of standardized grafts for full-thickness skin tissue regeneration.
Collapse
Affiliation(s)
- Shalini Dasgupta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, India.
| | - Ananya Barui
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology, Shibpur, India.
| |
Collapse
|
12
|
Lu X, Wang X, Li B, Wang X, Duan X, Liu D. Monocyte-Derived cxcl12 Guides a Directional Migration of Blood Vessels in Zebrafish. Arterioscler Thromb Vasc Biol 2025; 45:386-397. [PMID: 39846165 PMCID: PMC11855996 DOI: 10.1161/atvbaha.124.321588] [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: 10/24/2024] [Revised: 12/23/2024] [Accepted: 01/10/2025] [Indexed: 01/24/2025]
Abstract
BACKGROUND Sprouting blood vessels, reaching the aimed location, and establishing the proper connections are vital for building vascular networks. Such biological processes are subject to precise molecular regulation. So far, the mechanistic insights into understanding how blood vessels grow to the correct position are limited. In particular, the guide cues and the signaling-originating cells remain elusive. METHODS Live imaging analysis was used to observe the vascular developmental process of zebrafish. Whole-mount in situ hybridization and fluorescent in situ hybridization were used to detect the expression profiles of the genes. Single-cell sequencing analysis was conducted to identify the guiding protein and its originating cells. RESULTS Taking advantage of live imaging analysis, we described a directional blood vessel migration in the vascularization process of zebrafish pectoral fins. We demonstrated that pectoral fin vessel c migrated over long distances and was anastomosed with the second pair of intersegmental vessels. Furthermore, we found the cxcl12a-cxcr4a axis specifically guided this long-distance extension of pectoral fin vessel c-intersegmental vessel, and either inhibition or overexpression of cxcl12a-cxcr4a signaling both mislead the growth of pectoral fin vessel c to ectopic areas. Finally, based on an analysis of single-cell sequencing data, we revealed that a population of monocytes expresses the Cxcl12a, which guides the migration of the vascular sprout. CONCLUSIONS Our study identified Cxcl12a as the signaling molecule for orchestrating the organotypic-specific long-distance migration and anastomosis of the pectoral fin vessel and the intersegmental vessels in zebrafish. We discovered a specific cluster of gata1 (globin transcription factor 1)-positive monocytes responsible for expressing Cxcl12a. The findings offer novel insights into the mechanisms underlying organotypic vascularization in vertebrates.
Collapse
Affiliation(s)
- Xiaofeng Lu
- School of Life Science, Nantong Laboratory of Development and Diseases and Co-Innovation Center of Neuroregeneration, Nantong University, China
| | - Xiaoning Wang
- School of Life Science, Nantong Laboratory of Development and Diseases and Co-Innovation Center of Neuroregeneration, Nantong University, China
| | - Bowen Li
- School of Life Science, Nantong Laboratory of Development and Diseases and Co-Innovation Center of Neuroregeneration, Nantong University, China
| | - Xin Wang
- School of Life Science, Nantong Laboratory of Development and Diseases and Co-Innovation Center of Neuroregeneration, Nantong University, China
| | - Xuchu Duan
- School of Life Science, Nantong Laboratory of Development and Diseases and Co-Innovation Center of Neuroregeneration, Nantong University, China
| | - Dong Liu
- School of Life Science, Nantong Laboratory of Development and Diseases and Co-Innovation Center of Neuroregeneration, Nantong University, China
| |
Collapse
|
13
|
Kalies K, Knöpp K, Koch S, Pilowski C, Wurmbrand L, Sedding D. Restoration of angiogenic capacity in senescent endothelial cells by a pharmacological reprogramming approach. PLoS One 2025; 20:e0319381. [PMID: 40019880 PMCID: PMC11870368 DOI: 10.1371/journal.pone.0319381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/31/2025] [Indexed: 03/03/2025] Open
Abstract
Senescent endothelial cells (EC) are key players in the pathophysiology of cardiovascular diseases and are characterized by a reduced angiogenic and regenerative potential. Therefore, targeting these cells has been suggested as an effective therapeutic strategy to reduce vascular disease burden and potentially improve health and lifespan of humans. Here, we aimed to establish a pharmacological, partial reprogramming strategy to improve replicative senescent endothelial cell function in the context of angiogenesis. We demonstrate that our treatment improves tube formation and sprouting capacity but also increases proliferation and migration capacity in vitro. Further, inflammation and DNA damage were reduced in the replicative senescent cells. These processes were initiated by a short and timely-restricted overexpression of the Yamanaka-factors induced by our pharmacological strategy. The advantage of these compounds is that they are FDA approved in their respective concentrations which could pave the way for use in a clinical setting.
Collapse
Affiliation(s)
- Katrin Kalies
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Kai Knöpp
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Susanne Koch
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Claudia Pilowski
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Leonie Wurmbrand
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| | - Daniel Sedding
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
14
|
Pirson S, Gautier-Isola M, Baudin L, Rouaud L, Vanwynsberghe A, Deroye J, Bekisz S, Gucciardo F, Lebeau A, Buntinx F, Ivanova E, Staumont B, Blacher S, Gilles C, Noël A. AXL promotes lymphangiogenesis by amplifying VEGF-C-mediated AKT pathway. Cell Mol Life Sci 2025; 82:95. [PMID: 40011241 PMCID: PMC11865408 DOI: 10.1007/s00018-024-05542-3] [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: 05/27/2024] [Revised: 09/23/2024] [Accepted: 12/06/2024] [Indexed: 02/28/2025]
Abstract
Lymphangiogenesis has gained considerable interest due to its established role in cancer progression and dissemination of metastatic cells through lymph nodes. Deciphering the molecular mechanisms that govern lymphangiogenesis within lymph nodes holds promise for revealing novel targetable molecules and pathways to inhibit metastasis. In this study, we revealed a previously unrecognized role of AXL, a tyrosine kinase receptor, in the lymphatic vessel formation. We first validated the expression of AXL in lymphatic endothelial cells (LECs), followed by functional studies using RNA interference and pharmacological inhibition with R428/Bemcentinib. These approaches provided compelling evidence that AXL promotes LEC migration in both 2D and 3D culture systems. Our findings demonstrated that AXL activation was induced by VEGF-C (Vascular Endothelial Growth Factor C) and further amplified downstream signaling via the AKT pathway. In vivo, the role of AXL in lymphatic vessel sprouting was demonstrated using R428 in a model of VEGF-C-induced lymphangiogenesis in lymph nodes. Interestingly, we discovered that AXL was predominantly expressed in MARCO+ LECs. Strikingly, under metastatic conditions, there was a notable increase in the density and penetration extent of these AXL-expressing LECs into the lymph node parenchyma. Collectively, our findings pinpoint AXL as a potent enhancer of lymphangiogenesis operating through the VEGF-C/AKT pathway. Furthermore, the identification of AXL expression within a distinct LEC subpopulation, particularly in the context of metastasis, underscores the intricate interplay between AXL signaling and lymphatic dynamics within the lymph node microenvironment.
Collapse
Affiliation(s)
- Sébastien Pirson
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Marine Gautier-Isola
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Louis Baudin
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Loïc Rouaud
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Aline Vanwynsberghe
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Jonathan Deroye
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Sophie Bekisz
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
- Biomechanics Research Unit, GIGA In Silico Medicine, ULiège, Liège, Belgium
| | - Fabrice Gucciardo
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Alizée Lebeau
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Florence Buntinx
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Elitsa Ivanova
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Bernard Staumont
- Biomechanics Research Unit, GIGA In Silico Medicine, ULiège, Liège, Belgium
| | - Silvia Blacher
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Christine Gilles
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium
| | - Agnès Noël
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Liege University, Liege, Belgium.
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Wavre, Belgium.
- Laboratory of Tumor and Development Biology, GIGA-Cancer, Tour de Pathologie, B23, +4, Avenue Hippocrate, 13, Liège, 4000, Belgium.
| |
Collapse
|
15
|
He Y, Huang C, Chen J, Shen W. Caesalpinia sappan L. ethyl acetate extract regulated angiogenesis in atherosclerosis by modulating the miR-126/VEGF signalling pathway. Heliyon 2025; 11:e42159. [PMID: 39931479 PMCID: PMC11808623 DOI: 10.1016/j.heliyon.2025.e42159] [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: 06/12/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Aims of the study To investigate the regulatory mechanism of Caesalpinia sappan L. ethyl acetate extract (CSEAE) on angiogenesis in atherosclerosis (AS) based on the miR-126/VEGF signalling pathway. Materials and methods Our study first screened for differentially expressed microRNAs (miRNAs) associated with AS using the Gene Expression Omnibus (GEO) public database at the National Center for Biotechnology Information (NCBI) and R language software. Subsequently, our study verified the target-regulatory relationship between miR-126 and vascular endothelial growth factor (VEGF) in human umbilical vein endothelial cells (HUVECs) by using the "TargetScan" website and dual-luciferase reporter assay. In cellular experiments, Our study used cell proliferation assays and flow cytometry to assess the effects of CSEAE-Mediated serum on the proliferation and apoptosis of HUVECs. In animal experiments, our study used HE staining, Oil Red O staining and immunohistochemistry (IHC) staining to detect plaque area/lumen area (%), lipid area/plaque area (%) and microvessel density (MVD) in mouse aortas. In addition, our study performed RT‒PCR, ELISA and Western blot assays in ex vivo and in vivo experiments. Results A total of 39 differentially expressed miRNAs of AS were identified, among which the miR-126 expression level was significantly downregulated. Dual luciferase reporter gene assay results showed that miR-126 and VEGF have a targeting relationship, and the miR-126 mimic could inhibit the luciferase activity of the wild-type VEGF reporter gene vector (p value < 0.01). In cellular experiments, cell proliferation assays and flow cytometry results showed that CSEAE-Mediated serum significantly increased the proliferative activity after 24-72 h of treatment (p-value <0.01) and decreased the apoptotic level of HUVECs (p value < 0.01), and RT‒PCR results showed that CSEAE-Mediated serum significantly upregulated the expression of miR-126 (p value < 0.01) and downregulated the expression of VEGF mRNA in HUVECs (p value < 0.01). In vivo experiments, HE staining and IHC staining showed that CSEAE significantly reduced the MVD in the aorta and plaques of mice (p value < 0.01) and significantly reduced the aortic plaque area/lumen area (%) (p value < 0.01). Moreover, RT‒PCR assay and Western blot analysis results showed that CSEAE significantly upregulated the expression of miR-126 (p value < 0.01), downregulated the expression of VEGF mRNA (p value < 0.01), and decreased the protein expression levels of VEGF (p value < 0.01), phosphatidyl-inositol-3-kinase (PI3K) (p value < 0.01), and Ser/Thr-protein kinase (AKT1) (p value < 0.01) in mouse aortas, while ELISA showed that CSEAE significantly reduced the serum levels of vascular endothelial growth factor receptor (VEGFR2) (p value < 0.01) and hypoxia-inducible factor-1 (HIF-1) (p value < 0.01) in mice. Conclusion This study emphasises CSEAE as a natural medicinal extract for the treatment of AS that can improve the migratory viability and reduce the apoptosis of HUVECs to maintain the health of the arterial endothelial microenvironment, while CSEAE also inhibits angiogenesis and delays plaque formation in ApoE-/- mice, suggesting that the therapeutic effect of CSEAE for AS may be related to its inhibition of neovascularisation and that its molecular mechanism may be related to the miR-126/VEGF signalling pathway.
Collapse
Affiliation(s)
- Yue He
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University, People's Hospital of Shenzhen Baoan District, Shenzhen, 518000, Guangdong, China
| | - Chao Huang
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University, People's Hospital of Shenzhen Baoan District, Shenzhen, 518000, Guangdong, China
| | - Jingjing Chen
- Changchun University of Traditional Chinese Medicine, Changchun, 130000, Jilin, China
| | - Weizeng Shen
- Department of Traditional Chinese Medicine, The Second Affiliated Hospital of Shenzhen University, People's Hospital of Shenzhen Baoan District, Shenzhen, 518000, Guangdong, China
| |
Collapse
|
16
|
Wallace EJ, O'Dwyer J, Dolan EB, Burke LP, Wylie R, Bellavia G, Straino S, Cianfarani F, Ciotti G, Serini S, Calviello G, Roche ET, Mitra T, Duffy GP. Actuation-Mediated Compression of a Mechanoresponsive Hydrogel by Soft Robotics to Control Release of Therapeutic Proteins. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2401744. [PMID: 39692747 PMCID: PMC11831469 DOI: 10.1002/advs.202401744] [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: 02/26/2024] [Revised: 07/31/2024] [Indexed: 12/19/2024]
Abstract
Therapeutic proteins, the fastest growing class of pharmaceuticals, are subject to rapid proteolytic degradation in vivo, rendering them inactive. Sophisticated drug delivery systems that maintain protein stability, prolong therapeutic effects, and reduce administration frequency are urgently required. Herein, a mechanoresponsive hydrogel is developed contained within a soft robotic drug delivery (SRDD) device. In a step-change from previously reported systems, pneumatic actuation of this system releases the cationic therapeutic protein Vascular Endothelial Growth Factor (VEGF) in a bioactive form which is required for therapeutic angiogenesis, the growth of new blood vessels, in numerous clinical conditions. The ability of the SRDD device to release bioactive VEGF in a spatiotemporal manner from the hydrogel is tested in diabetic rats - a model in which angiogenesis is difficult to stimulate. Daily actuation of the SRDD device in the diabetic rat model significantly increased cluster of differentiation 31+ (CD31+) blood vessel number (p = 0.0335) and the diameter of alpha-smooth muscle actin+ (α-SMA+) blood vessels (p = 0.0025) compared to passive release of VEGF from non-actuated devices. The SRDD device combined with the mechanoresponsive hydrogel offers the potential to deliver an array of bioactive therapeutics in a spatiotemporal manner to mimic their natural release in vivo.
Collapse
Affiliation(s)
- Eimear J. Wallace
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- Explora‐Bioscience SrlG. Peroni 386Rome00131Italy
| | - Joanne O'Dwyer
- Pharmacology and TherapeuticsSchool of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
| | - Eimear B. Dolan
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- CÚRAMSFI Research Centre for Medical DevicesUniversity of GalwayGalwayH91 W2TYIreland
- Biomedical EngineeringSchool of EngineeringUniversity of GalwayGalwayH91 HX31Ireland
| | - Liam P. Burke
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- Antimicrobial Resistance and Microbial Ecology GroupSchool of Medicine, College of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 DK59Ireland
- Centre for One HealthRyan InstituteUniversity of GalwayGalwayH91 DK59Ireland
| | - Robert Wylie
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
| | | | | | | | | | - Simona Serini
- Department of Translational Medicine and SurgerySection of General Pathology, Faculty of Medicine and SurgeryUniversità Cattolica del Sacro CuoreLargo F. VitoRome1‐00168Italy
| | - Gabriella Calviello
- Department of Translational Medicine and SurgerySection of General Pathology, Faculty of Medicine and SurgeryUniversità Cattolica del Sacro CuoreLargo F. VitoRome1‐00168Italy
| | - Ellen T. Roche
- Institute for Medical Engineering and ScienceMassachusetts Institute of TechnologyCambridgeMA 01239USA
- Harvard‐MIT Program in Health Sciences and TechnologyCambridgeMA02139USA
- Department of Mechanical EngineeringMassachusetts Institute of TechnologyCambridgeMA02139USA
| | - Tapas Mitra
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- CÚRAMSFI Research Centre for Medical DevicesUniversity of GalwayGalwayH91 W2TYIreland
| | - Garry P. Duffy
- Anatomy and Regenerative Medicine Institute (REMEDI)School of MedicineCollege of Medicine Nursing and Health SciencesUniversity of GalwayGalwayH91 W2TYIreland
- CÚRAMSFI Research Centre for Medical DevicesUniversity of GalwayGalwayH91 W2TYIreland
- SFI Centre for Advanced Materials and BioEngineering Research Centre (AMBER)Trinity College DublinDublinD02 W9K7Ireland
| |
Collapse
|
17
|
Butta S. Vessel Co-option: A Promising Therapeutic Strategy in Oral Squamous Cell Carcinoma. Cureus 2025; 17:e79572. [PMID: 40144412 PMCID: PMC11940552 DOI: 10.7759/cureus.79572] [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] [Accepted: 02/24/2025] [Indexed: 03/28/2025] Open
Abstract
Background Vessel co-option is the mechanism by which cancer cells take over existing blood vessels for their own growth and metabolism. It is often associated with treatment failure and metastasis in tumors of the brain, lung, liver, and skin. Limited studies have highlighted the role of vessel co-option in oral squamous cell carcinoma (OSCC). This study aims to determine the association between vessel co-option in OSCC and the Ki67 labeling index (LI) to ascertain its prognostic role. Methodology In this retrospective study, small biopsy specimens were sent and histopathologically processed to prepare formalin-fixed paraffin-embedded blocks. Only cases diagnosed as OSCC were included in the study. Immunohistochemistry for CD34, p40, and Ki67 was performed. Results The study included 39 OSCC patients. The majority (n = 27; 69.23%) of the patients were in the age group of 31-50 years. The male-to-female ratio was 3:1. Tongue (n = 15; 38.46%) was the most common site, followed by the floor of the mouth (n = 11; 28.21%). The majority (n = 25; 64.10%) of the cases were classified as moderately differentiated squamous cell carcinoma. Microscopically, vessel co-option was noted in 71.79% (n = 28) of the cases. Immunohistochemically, Ki67 LI was >10% in 41.03% (n = 16) of the cases. A statistically significant association was found between Ki67 LI and vessel co-option (p = 0.0013). Conclusions Vessel co-option is a potential determinant of the aggressive potential of OSCC. Histopathological assessment of vessel co-option in conjunction with the Ki67 proliferative index has promising potential for assessing the prognosis of OSCC.
Collapse
Affiliation(s)
- Shristi Butta
- Pathology, Institute of Post Graduate Medical Education and Research (IPGMER) and Seth Sukhlal Karnani Memorial (SSKM) Hospital, Kolkata, IND
| |
Collapse
|
18
|
Zhang Y, Qi S, Shen W, Guo Y, Liang Y, Zhuo Q, Kong H, Zhang S, Zhao C. Metabolomic and transcriptomic analysis reveals metabolic-immune interactions in choroid neovascularization. Exp Eye Res 2025; 251:110227. [PMID: 39732424 DOI: 10.1016/j.exer.2024.110227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/27/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024]
Abstract
Choroid neovascularization (CNV) is a distinct type of age-related macular degeneration (AMD) with a poor prognosis and responsible for the majority of vision loss in the elderly population. The laser-induced CNV model is a well-established animal model frequently used to study CNV. In this study, we performed an integrated analysis of metabolomic and transcriptomic data from CNV samples, utilizing multiple approaches including single-sample gene set enrichment analysis (ssGSEA), correlation analysis, and weighted gene co-expression network analysis (WGCNA), alongside various bioinformatics platforms, to identify key metabolic and immune signatures and to investigate their interplay during angiogenesis. Dominant infiltration of macrophages and monocytes was detected and a positive correlation between dysregulated riboflavin metabolism and angiogenesis pathways was characterized. Hub genes such as ectonucleotide pyrophosphatase/phosphodiesterase 1 (Enpp1) and acid phosphatase 5, tartrate resistant (ACP5) emerged as potential central regulators of immune-metabolic crosstalk in CNV. The classification of the immune and metabolic landscape and their critical interactions in CNV models will enhance the understanding of the pathogenesis of neovascular AMD and other neovascular eye diseases, contributing to the development of multi-targeted therapeutic strategies with better efficacy.
Collapse
Affiliation(s)
- Yihan Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Siyi Qi
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Weiai Shen
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Ying Guo
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Yu Liang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Qiao Zhuo
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Hongyu Kong
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China
| | - Shujie Zhang
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China.
| | - Chen Zhao
- Eye Institute and Department of Ophthalmology, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200031, China; Key Laboratory of Myopia and Related Eye Diseases, NHC, Chinese Academy of Medical Sciences, 83 Fenyang Road, Shanghai, 200031, China; Shanghai Key Laboratory of Visual Impairment and Restoration, 83 Fenyang Road, Shanghai, 200031, China.
| |
Collapse
|
19
|
Li Z, Sun X, Zhao Z, Yang Q, Ren Y, Teng X, Tai DCS, Wanless IR, Schattenberg JM, Liu C. A machine learning based algorithm accurately stages liver disease by quantification of arteries. Sci Rep 2025; 15:3143. [PMID: 39856155 PMCID: PMC11759706 DOI: 10.1038/s41598-025-87427-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025] Open
Abstract
A major histologic feature of cirrhosis is the loss of liver architecture with collapse of tissue and vascular changes per unit. We developed qVessel to quantify the arterial density (AD) in liver biopsies with chronic disease of varied etiology and stage. 46 needle liver biopsy samples with chronic hepatitis B (CHB), 48 with primary biliary cholangitis (PBC) and 43 with metabolic dysfunction-associated steatotic liver disease (MASLD) were collected at the Shuguang Hospital. The METAVIR system was used to assess stage. The second harmonic generation (SHG)/two-photon images were generated from unstained slides. Collagen proportionate area (CPA) using SHG. AD was counted using qVessel (previously trained on manually labeled vessels by stained slides (CD34/a-SMA/CK19) and developed by a decision tree algorithm). As liver fibrosis progressed from F1 to F4, we observed that both AD and CPA gradually increases among the three etiologies (P < 0.05). However, at each stage of liver fibrosis, there was no significant difference in AD or CPA between CHB and PBC compared to MASLD (P > 0.05). AD and CPA performed similar diagnostic efficacy in liver cirrhosis (P > 0.05). Using the qVessel algorithm, we discovered a significant correlation between AD, CPA and METAVIR stages in all three etiologies. This suggests that AD could underpin a novel staging system.
Collapse
Affiliation(s)
- Zhengxin Li
- Gongli Hospital of Shanghai Pudong New Area, Shanghai, China
| | - Xin Sun
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203, China
| | - Zhimin Zhao
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203, China
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China
| | - Qiang Yang
- Hangzhou Choutu Tech. Co., Ltd., Hangzhou, China
| | - Yayun Ren
- Hangzhou Choutu Tech. Co., Ltd., Hangzhou, China
| | - Xiao Teng
- Histoindex Pte. Ltd, Singapore, Singapore
| | | | - Ian R Wanless
- Department of Pathology, Queen Elizabeth II Health Sciences Centre, Dalhousie University, Halifax, Canada
| | - Jörn M Schattenberg
- Department of Internal Medicine II, Saarland University Medical Center, Homburg, Germany
- Saarland University, Saarbrücken, Germany
| | - Chenghai Liu
- Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 528 Zhangheng Road, Pudong New Area, Shanghai, 201203, China.
- Shanghai Key Laboratory of Traditional Chinese Clinical Medicine, Shanghai, China.
| |
Collapse
|
20
|
Wagner BJ, Ettner-Sitter A, Ihlo NA, Behr M, Koelbl S, Brunner SM, Weber F, Rau BM, Schlitt HJ, Brochhausen C, Schoenmehl R, Artinger A, Schott D, Pizon M, Pachmann K, Aung T, Haerteis S, Hackl C. Patient-derived xenografts from circulating cancer stem cells as a preclinical model for personalized pancreatic cancer research. Sci Rep 2025; 15:2896. [PMID: 39843495 PMCID: PMC11754431 DOI: 10.1038/s41598-025-87054-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 01/15/2025] [Indexed: 01/24/2025] Open
Abstract
Patient-derived xenografts (PDXs) provide biologically relevant models and potential platforms for the development of treatment strategies for precision medicine in pancreatic cancer. Furthermore, circulating epithelial tumor cells (CETCs/CTCs) are released into the bloodstream by solid tumors and a rare subpopulation-circulating cancer stem cells (cCSCs) - is considered to be responsible for recurrence and plays a key role in metastasis. For the identification of cCSCs, an innovative in vitro assay to generate tumorspheres was established in this study. The number of tumorspheres and CETCs/CTCs was analyzed perioperatively in 25 pancreatic cancer patients. Additionally, an individual in vivo chorioallantoic membrane (CAM) culture system was used to generate PDXs from these tumorspheres. While overall correlations of CETCs/CTCs with clinicopathological parameters did not reach statistical significance, a significant difference in the number of tumorspheres was observed between patient subgroups with lower and higher UICC stages. This finding underscores their potential as biomarkers, providing valuable insights into clinical decision-making and tumor progression. The application of tumorspheres on the CAM successfully established PDXs within 7 days. These xenografts closely resembled the histological features of the primary tumor. Hence, this model represents a novel and fast option for individualized testing of new therapies for PDAC.
Collapse
Affiliation(s)
- Benedikt J Wagner
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Andreas Ettner-Sitter
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany
| | - Nicolas A Ihlo
- Faculty of Informatics and Data Science, University of Regensburg, Bajuwarenstrasse 4, 93053, Regensburg, Germany
| | - Merle Behr
- Faculty of Informatics and Data Science, University of Regensburg, Bajuwarenstrasse 4, 93053, Regensburg, Germany
| | - Sebastian Koelbl
- Technology Campus Hutthurm, Deggendorf Institute of Technology, Hochleiten 1, 94116, Hutthurm, Germany
| | - Stefan M Brunner
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Florian Weber
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Bettina M Rau
- Department of General, Visceral and Thoracic Surgery, Academic Teaching Hospital Neumarkt, Nuernberger Strasse 12, 92318, Neumarkt in der Oberpfalz, Germany
| | - Hans J Schlitt
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| | - Christoph Brochhausen
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Rebecca Schoenmehl
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | - Annalena Artinger
- Institute of Pathology, Medical Faculty Mannheim, Heidelberg University, Theodor-Kutzer-Ufer 1-3, 68167, Mannheim, Germany
| | | | - Monika Pizon
- Simfo GmbH, Kurpromenade 2, 95448, Bayreuth, Germany
- Labor Dr. Pachmann Bayreuth, Kurpromenade 2, 95448, Bayreuth, Germany
| | - Katharina Pachmann
- Simfo GmbH, Kurpromenade 2, 95448, Bayreuth, Germany
- Labor Dr. Pachmann Bayreuth, Kurpromenade 2, 95448, Bayreuth, Germany
| | - Thiha Aung
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany
- Faculty of Applied Healthcare Science, Deggendorf Institute of Technology, Dieter-Goerlitz-Platz 1, 94469, Deggendorf, Germany
| | - Silke Haerteis
- Institute for Molecular and Cellular Anatomy, University of Regensburg, Universitaetsstrasse 31, 93053, Regensburg, Germany.
| | - Christina Hackl
- Department of Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
- Bavarian Cancer Research Center (BZKF), University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany
| |
Collapse
|
21
|
Khodadust F, Philippon EML, Steinz MM, van Hamburg JP, van Meerloo J, van Beijnum JR, Jansen G, Tas SW, van der Laken CJ. Unveiling the Anti-Angiogenic Potential of Small-Molecule (Kinase) Inhibitors for Application in Rheumatoid Arthritis. Cells 2025; 14:102. [PMID: 39851530 PMCID: PMC11764070 DOI: 10.3390/cells14020102] [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: 12/13/2024] [Revised: 12/30/2024] [Accepted: 01/09/2025] [Indexed: 01/26/2025] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease characterized by inflammation leading to joint damage and systemic complications. Angiogenesis promotes inflammation and contributes to RA progression. This study evaluated potential anti-angiogenic effects of several compounds including small-molecule kinase inhibitors, such as sunitinib (pan-kinase inhibitor), tofacitinib (JAK-inhibitor), NIKi (NF-κB-inducing kinase inhibitor), and the integrin-targeting peptide fluciclatide, using a scratch assay and 3D spheroid-based models of angiogenesis. For all drugs tested in the low micromolar range (1-25 μM), sunitinib (as positive anti-angiogenetic control) showed marked inhibition of endothelial cell (EC) migration and sprouting, effectively reducing both scratch closure and sprout formation. Tofacitinib exhibited marginal effectiveness in the scratch assay, but performed better in the 3D models (55% inhibition), whereas NIKi showed around 50% anti-angiogenic effects in both models. Fluciclatide changed EC morphology rather than migration, and only when stimulated with synovial fluid in spheroid model did it show inhibitory effects (at ≥2.5 µM), with none below this dosage. These results highlight the potential of NIKi and tofacitinib for angiogenesis inhibition and of fluciclatide for safe diagnostic targeting of microdose in RA, as well as the need for advanced screening models that mimic RA's complex inflammatory pro-angiogenic environment.
Collapse
Affiliation(s)
- Fatemeh Khodadust
- Department of Rheumatology & Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (F.K.); (S.W.T.)
| | - Eva M. L. Philippon
- Department of Rheumatology & Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (F.K.); (S.W.T.)
- Department of Experimental Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Maarten M. Steinz
- Department of Rheumatology & Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (F.K.); (S.W.T.)
| | - Jan Piet van Hamburg
- Department of Rheumatology & Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (F.K.); (S.W.T.)
- Department of Experimental Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Johan van Meerloo
- Department of Hematology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
- Cancer Center Amsterdam, Amsterdam UMC, 1081 HV Amsterdam, The Netherlands
| | - Judy R. van Beijnum
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Gerrit Jansen
- Department of Rheumatology & Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (F.K.); (S.W.T.)
| | - Sander W. Tas
- Department of Rheumatology & Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (F.K.); (S.W.T.)
- Department of Experimental Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Conny J. van der Laken
- Department of Rheumatology & Clinical Immunology, Amsterdam UMC, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands; (F.K.); (S.W.T.)
| |
Collapse
|
22
|
Huang X, Ye H, Hu Y, Lei Y, Tian Y, Huang X, Zhang J, Zhang Y, Gui B, Liu Q, Zhang G, Deng Q. Ultrasound super-resolution imaging for non-invasive assessment of microvessel in prostate lesion. Cancer Imaging 2025; 25:1. [PMID: 39773358 PMCID: PMC11706184 DOI: 10.1186/s40644-024-00819-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND Prostate cancer (PCa) is the leading cause of cancer-related morbidity and mortality in men worldwide. An early and accurate diagnosis is crucial for effective treatment and prognosis. Traditional invasive procedures such as image-guided prostate biopsy often cause discomfort and complications, deterring some patients from undergoing these necessary tests. This study aimed to explore the feasibility and clinical value of using ultrasound super-resolution imaging (US SRI) for non-invasively assessing the microvessel characteristics of prostate lesion. METHODS This study included 127 patients with prostate lesion who presented at Renmin Hospital of Wuhan University between November 2023 and June 2024 were included in this study. All the patients underwent transrectal US (TRUS), contrast-enhanced US (CEUS), and US SRI. CEUS parameters of time-intensity curve (TIC): arrival time (AT), rising time (RT), time to peak (TTP), peak intensity (PKI), falling time (FT), mean transit time (MTT), ascending slope (AS), descending slope (DS), D/A slope ratio (SR), and area under the TIC (AUC). US SRI parameters: microvessel density (MVD), microvessel diameter (D), microvessel velocity (V), microvessel tortuosity (T), and fractal number (FN), were analyzed and compared between prostate benign and malignant lesion. RESULTS The tumor markers of prostate in the malignant group were all higher than those in the benign group, and the differences were statistically significant (P < 0.001). The TIC parameters of CEUS revealed that the PKI, AS, DS, and AUC were significantly higher in the malignant group than in the benign group (P < 0.001), whereas the RT, TTP and FT in the malignant group were significantly lower (P < 0.001). Malignant lesion exhibited significantly higher MVD, larger D, faster V, greater T, and more complex FN than benign lesion (P < 0.001). CONCLUSIONS US SRI is a promising non-invasive imaging modality that can provide detailed microvessel characteristics of prostate lesion, offering an advancement in the differential diagnosis for prostate lesion. And, US SRI may be a valuable tool in clinical practice with its ability to display and quantify microvessel with high precision.
Collapse
Affiliation(s)
- Xin Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Huarong Ye
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yugang Hu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yumeng Lei
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Yi Tian
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China
| | - Xingyue Huang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jun Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Yao Zhang
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Bin Gui
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qianhui Liu
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Ge Zhang
- Department of Medical Ultrasound, China Resources & Wisco General Hospital, Wuhan University of Science and Technology, Wuhan, 430080, China.
- Physics for Medicine Paris, Inserm U1273, ESPCI Paris, PSL University, Paris, 75015, France.
| | - Qing Deng
- Department of Ultrasound Imaging, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
23
|
Zhong C, Shi Z, Liu CY, Binzel DW, Jin K, Li X, Guo P, Li SK. Inhibition of Endothelial Cell Tube Formation by Anti-Vascular Endothelial Growth Factor/Anti-Angiopoietin-2 RNA Nanoparticles. Pharmaceutics 2025; 17:55. [PMID: 39861703 PMCID: PMC11769471 DOI: 10.3390/pharmaceutics17010055] [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: 11/16/2024] [Revised: 12/13/2024] [Accepted: 12/27/2024] [Indexed: 01/27/2025] Open
Abstract
RNA nanoparticles, derived from the packaging RNA three-way junction motif (pRNA-3WJ) of the bacteriophage phi29 DNA packaging motor, have been demonstrated to be thermodynamically and chemically stable, with promise as a nanodelivery system. Background/Objectives: A previous study showed that RNA nanoparticles with antiangiogenic aptamers (anti-vascular endothelial growth factor (VEGF) and anti-angiopoietin-2 (Ang2) aptamers) inhibited cell proliferation via WST-1 assay. To further investigate the antiangiogenic potential of these RNA nanoparticles, a modified three-dimensional (3D) spheroid sprouting assay model of human umbilical vein endothelial cells was utilized in the present study. Methods: Three groups of RNA nanoparticles were evaluated, namely, pRNA-3WJ series, RNA square series (polygon-type RNA nanoparticles), and 8WJ series (multiple-way junction RNA nanoparticles), which were conjugated with a single anti-VEGF, the combination of one anti-VEGF and one anti-Ang2, or multiple anti-VEGF aptamers. The core scaffold RNA nanoparticles (without aptamers) were used as the references, and bevacizumab was used as the positive control. Results: The results demonstrated the inhibition effects of the RNA nanoparticles on endothelial cell tube formation at 67 nM in a 3D spheroid sprouting model. The results in the 3D spheroid sprouting assay are consistent with those of the WST-1 proliferation assays. Conclusions: Among the RNA nanoparticles evaluated, 3WJ-3VEGF and SQR-VEGF-Ang2 had inhibition effects equivalent to bevacizumab and were promising for anti-angiogenesis treatment.
Collapse
Affiliation(s)
- Cheng Zhong
- Division of Pharmaceutical Sciences, James L Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Zhanquan Shi
- Division of Pharmaceutical Sciences, James L Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Chia-Yang Liu
- Department of Ophthalmology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Daniel W. Binzel
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Kai Jin
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Xin Li
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Peixuan Guo
- Center for RNA Nanobiotechnology and Nanomedicine, College of Pharmacy, James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
- College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - S. Kevin Li
- Division of Pharmaceutical Sciences, James L Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH 45267, USA
| |
Collapse
|
24
|
Zhao Y, Sun M, Pan Z, Kong W, Hong Z, Zhang W, Sun B, Zhang J, Wang X, Wang K. A novel quantitative angiogenesis assay based on visualized vascular organoid. Angiogenesis 2025; 28:10. [PMID: 39751990 DOI: 10.1007/s10456-024-09967-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Angiogenesis describes the sprouting of blood vessels from existing vasculatures and it plays a pivotal role in disease progress such as diabetes, age-related macular degeneration and cancer. However, the most widely used anti-angiogenic agents targeting vascular endothelial growth factor (VEGF) pathway still lacked of specificity and therapeutic efficacy. To establish a method suitable for high-throughput drug screening and faithfully recapitulate the feature of in vivo angiogenesis, we generated a PECAM1-mRuby3-secNluc; ACTA2-EGFP dual reporter human pluripotent stem cell (hPSC) line and utilizing the cell line to establish a visualized and quantifiable in vitro angiogenesis model with stem cell-derived vascular organoid. Using this method, we evaluated the anti-angiogenic effect of VEGFR inhibitor and efficiently identified several potential candidates of pro- and anti-angiogenic therapy via bioluminescence-based quantification. Overall, our study provides a valuable platform for in vitro drug screenings.
Collapse
Affiliation(s)
- Yun Zhao
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Mengze Sun
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Zihang Pan
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Weijing Kong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Zixuan Hong
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China
| | - Wei Zhang
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing, 102200, China
| | - Bingbing Sun
- TianXinFu (Beijing) Medical Appliance Co., Ltd., Beijing, 102200, China
| | - Jingjing Zhang
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Xi Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China.
- State Key Laboratory of Female Fertility Promotion, Department of Obstetrics and Gynecology, Peking University Third Hospital, Institute of Advanced Clinical Medicine, Peking University, Beijing, 100191, China.
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Beijing Advanced Center of Cellular Homeostasis and Aging-Related Diseases, Clinical Stem Cell Research Center, Peking University Third Hospital, Peking University, Beijing, 100191, China.
| |
Collapse
|
25
|
Wolint P, Hofmann S, von Atzigen J, Böni R, Miescher I, Giovanoli P, Calcagni M, Emmert MY, Buschmann J. Standardization to Characterize the Complexity of Vessel Network Using the Aortic Ring Model. Int J Mol Sci 2024; 26:291. [PMID: 39796147 PMCID: PMC11719671 DOI: 10.3390/ijms26010291] [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: 11/13/2024] [Revised: 12/05/2024] [Accepted: 12/30/2024] [Indexed: 01/13/2025] Open
Abstract
Regeneration after ischemia requires to be promoted by (re)perfusion of the affected tissue, and, to date, there is no therapy that covers all needs. In treatment with mesenchymal stem cells (MSC), the secretome acts via paracrine mechanisms and has a positive influence on vascular regeneration via proangiogenic factors. A lack of standardization and the high complexity of vascular structures make it difficult to compare angiogenic readouts from different studies. This emphasizes the need for improved approaches and the introduction of an index in the preclinical setting. A characterization of human MSC secretomes obtained from one of the three formats-single cells, small, and large spheroids-was performed using the chicken aortic ring assay in combination with a modified angiogenic activity index (AAI) and an angiogenic profile. While the secretome of the small spheroid group showed an inhibitory effect on angiogenesis, the large spheroid group impressed with a fully pro-angiogenic response, and a higher AAI compared to the single cell group, underlying the suitability of these three-stem cell-derived secretomes with their distinct angiogenic properties to validate the AAI and the novel angiogenic profile established here.
Collapse
Affiliation(s)
- Petra Wolint
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Silvan Hofmann
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Julia von Atzigen
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Roland Böni
- White House Center for Liposuction, 8044 Zurich, Switzerland;
| | - Iris Miescher
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Pietro Giovanoli
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maurizio Calcagni
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| | - Maximilian Y. Emmert
- Institute for Regenerative Medicine (IREM), University of Zurich, 8952 Zurich, Switzerland;
- Deutsches Herzzentrum der Charité (DHZC), Department of Cardiothoracic and Vascular Surgery, 13353 Berlin, Germany
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 13353 Berlin, Germany
- BIH Center for Regenerative Therapies (BCRT), Berlin Institute of Health at Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Johanna Buschmann
- Division of Surgical Research, University Hospital of Zurich, 8091 Zurich, Switzerland
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (S.H.); (J.v.A.); (I.M.); (P.G.); (M.C.)
| |
Collapse
|
26
|
Zhang S, Xu X, Zhang K, Lei C, Xu Y, Zhang P, Zhang Y, Gu H, Huang C, Qiu Z. Targeting OAS3 for reversing M2d infiltration and restoring anti-tumor immunity in pancreatic cancer. Cancer Immunol Immunother 2024; 74:37. [PMID: 39738657 PMCID: PMC11685377 DOI: 10.1007/s00262-024-03898-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/16/2024] [Indexed: 01/02/2025]
Abstract
Abundant infiltration of tumor-associated macrophages (TAMs) within the tumor stroma plays a pivotal role in inducing immune escape in pancreatic cancer (PC). Lactate serves as a direct regulator of macrophage polarization and functions, although the precise regulation mechanism remains inadequately understood. Our study revealed that PC cells (PCs) promote macrophage polarization toward M2d through high lactate secretion. M2d is characterized by elevated secretion of IL-10 and VEGF-A, which diminish CD8+T cells cytotoxicity and promote tumor neoangiogenesis simultaneously. Additionally, we identify 2,5'-oligoadenylate synthase 3 (OAS3) as an essential regulator of M2d polarization, upregulated by PCs via lactate/METTL3/OAS3 axis. Increased OAS3 expression in TAMs correlates with m6A modification mediated by METTL3 on OAS3 mRNA and is associated with poorer prognosis in PC patients. OAS3 deficiency in macrophages substantially impairs IL-10highVEGF-AhighM2d polarization and their pro-tumor functions while enhancing the therapeutic efficacy of gemcitabine and anti-PD-L1 mAb in humanized mouse models. In conclusion, OAS3 presents as a promising immune therapeutic target for reversing IL-10highVEGF-AhighM2d infiltration and restoring CD8+T cell-mediated anti-tumor immunity in pancreatic cancer.
Collapse
Affiliation(s)
- Shaopeng Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ximo Xu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kundong Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of General Surgery, Jiuquan Branch of Shanghai General Hospital, Jiuquan, Gansu, China
| | - Changzheng Lei
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yitian Xu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pengshan Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuan Zhang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haitao Gu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chen Huang
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Zhengjun Qiu
- Department of Gastrointestinal Surgery, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
27
|
Ding JY, Meng TT, Du RL, Song XB, Li YX, Gao J, Ji R, He QY. Bibliometrics of trends in global research on the roles of stem cells in myocardial fibrosis therapy. World J Stem Cells 2024; 16:1086-1105. [PMID: 39734477 PMCID: PMC11669986 DOI: 10.4252/wjsc.v16.i12.1086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Revised: 10/05/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND Myocardial fibrosis, a condition linked to several cardiovascular diseases, is associated with a poor prognosis. Stem cell therapy has emerged as a potential treatment option and the application of stem cell therapy has been studied extensively. However, a comprehensive bibliometric analysis of these studies has yet to be conducted. AIM To map thematic trends, analyze research hotspots, and project future directions of stem cell-based myocardial fibrosis therapy. METHODS We conducted a bibliometric and visual analysis of studies in the Web of Science Core Collection using VOSviewer and Microsoft Excel. The dataset included 1510 articles published between 2001 and 2024. Countries, organizations, authors, references, keywords, and co-citation networks were examined to identify evolving research trends. RESULTS Our findings revealed a steady increase in the number of publications, with a projected increase to over 200 publications annually by 2030. Initial research focused on stem cell-based therapy, particularly for myocardial infarction and heart failure. More recently, there has been a shift toward cell-free therapy, involving extracellular vesicles, exosomes, and microRNAs. Key research topics include angiogenesis, inflammation, apoptosis, autophagy, and oxidative stress. CONCLUSION This analysis highlights the evolution of stem cell therapies for myocardial fibrosis, with emerging interest in cell-free approaches. These results are expected to guide future scientific exploration and decision-making.
Collapse
Affiliation(s)
- Jing-Yi Ding
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Tian-Tian Meng
- Department of Rehabilitation, Dongfang Hospital, Beijing University of Chinese Medicine, Beijing 100071, China
| | - Ruo-Lin Du
- Department of Emergency Medicine, South Branch of Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Xin-Bin Song
- Department of Intensive Care Unit, Zhumadian Hospital of Traditional Chinese Medicine, Zhumadian 463000, Henan Province, China
| | - Yi-Xiang Li
- Department of Chinese Medicine, The Third People's Hospital of Henan Province, Zhengzhou 450000 Henan Province, China
| | - Jing Gao
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ran Ji
- Department of Intensive Care Unit, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Qing-Yong He
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
28
|
Teodoro L, Carreira ACO, Sogayar MC. Exploring the Complexity of Pan-Cancer: Gene Convergences and in silico Analyses. BREAST CANCER (DOVE MEDICAL PRESS) 2024; 16:913-934. [PMID: 39691553 PMCID: PMC11651076 DOI: 10.2147/bctt.s489246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/06/2024] [Indexed: 12/19/2024]
Abstract
Cancer is a complex and multifaceted group of diseases characterized by highly intricate mechanisms of tumorigenesis and tumor progression, which complicates diagnosis, prognosis, and treatment. In recent years, targeted therapies have gained prominence by focusing on specific mutations and molecular features unique to each tumor type, offering more effective and personalized treatment options. However, it is equally critical to explore the genetic commonalities across different types of cancer, which has led to the rise of pan-cancer studies. These approaches help identify shared therapeutic targets across various tumor types, enabling the development of broader and potentially more widely applicable treatment strategies. This review aims to provide a comprehensive overview of key concepts related to tumors, including tumorigenesis processes, the tumor microenvironment, and the role of extracellular vesicles in tumor biology. Additionally, we explore the molecular interactions and mechanisms driving tumor progression, with a particular focus on the pan-cancer perspective. To achieve this, we conducted an in silico analysis using publicly available datasets, which facilitated the identification of both common and divergent genetic and molecular patterns across different tumor types. By integrating these diverse areas, this review offers a clearer and deeper understanding of the factors influencing tumorigenesis and highlights potential therapeutic targets.
Collapse
Affiliation(s)
- Leandro Teodoro
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| | - Ana Claudia O Carreira
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Center of Human and Natural Sciences, Federal University of ABC, Santo André, São Paulo, 09280-560, Brazil
| | - Mari C Sogayar
- Cell and Molecular Therapy NUCEL Group, School of Medicine, University of São Paulo, São Paulo, São Paulo, 01246-903, Brazil
- Biochemistry Department, Chemistry Institute, University of São Paulo, São Paulo, São Paulo, 05508-900, Brazil
| |
Collapse
|
29
|
Xu J, Zou Z, Liu W, Zhang Q, Shen J, Hao F, Chen G, Yu D, Li Y, Zhang Z, Qin Y, Yang R, Wang Y, Duan L. HAPLN3 p.T34A contributes to incomplete penetrance of moyamoya disease in Chinese carrying RNF213 p.R4810K. Eur J Neurol 2024; 31:e16473. [PMID: 39315749 PMCID: PMC11555006 DOI: 10.1111/ene.16473] [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: 08/10/2024] [Accepted: 08/26/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND AND PURPOSE The penetrance of the RNF213 p.R4810K, a founder mutation of moyamoya disease (MMD), is estimated to be only 1/150-1/300. However, the factors affecting its penetrance remain unclear. Therefore, our study aims to identify modifier genes associated with the incomplete penetrance of this founder mutation. METHODS Whole-exome sequencing (WES) was performed on 36 participants, including 22 MMD patients and 14 non-MMD controls with RNF213 p.R4810K mutation. Fisher's exact test was used to assess the presence of genetic variants that differed significantly between MMD patients and non-MMD controls. In order to exclude false-positive results, another 55 carriers were included to perform Fisher's exact test for the selected sites in the WES discovery stage. Subsequently, human brain microvascular endothelial cells were transfected with wild-type and mutant HAPLN3 for tube formation assays and western blotting to explore the impact of candidate genes on angiogenesis. RESULTS Analysis of variants from WES data revealed a total of 12 non-synonymous variants. Through bioinformatics analysis, the focus was on the HAPLN3 p.T34A variant with a significant p value of 0.00731 in Fisher's exact test. Validation through Sanger sequencing confirmed the presence of this variant in the WES data. In vitro experiments revealed that silencing HAPLN3 and transfecting HAPLN3 p.T34A significantly enhanced tube formation and increased the relative protein expression of vascular endothelial growth factor in endothelial cells. CONCLUSIONS These results suggest that HAPLN3 may function as a modifier gene of RNF213 p.R4810K, promoting the development of MMD and contributing to the incomplete penetrance of MMD founder mutations.
Collapse
Affiliation(s)
- Jun Xu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Zhengxing Zou
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Wanyang Liu
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Qian Zhang
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Juan Shen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Fangbin Hao
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Gan Chen
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Dan Yu
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Yunzhu Li
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Zhengshan Zhang
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Yuchen Qin
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Rimiao Yang
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| | - Yue Wang
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention, Ministry of Education (China Medical University), Department of Nutrition and Food Hygiene, School of Public HealthChina Medical UniversityShenyangChina
| | - Lian Duan
- Department of Neurosurgery, the Fifth Medical CentreChinese PLA General HospitalBeijingChina
| |
Collapse
|
30
|
Yan H, Cai X, Zhang J, Zhao H, Wu H, Zhang J, Xu L, Liu S, Zang Y, Fu S. Gastric cancer cell-derived exosomal miRNA-128-3p promotes angiogenesis by targeting SASH1. Front Oncol 2024; 14:1440996. [PMID: 39664191 PMCID: PMC11631727 DOI: 10.3389/fonc.2024.1440996] [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: 05/30/2024] [Accepted: 11/11/2024] [Indexed: 12/13/2024] Open
Abstract
Exosomes, key components of the tumour microenvironment, can mediate intercellular communication through the delivery of various signalling molecules, including microribonucleic acids (miRNAs), and ultimately participate in regulating the process of tumour development. In this study, we aimed to investigate the reason and mechanism by which exosomal miRNAs derived from gastric cancer cells affect carcinogenesis and metastasis. Among these miRNAs, microRNA-128-3p (miR-128-3p) was highly expressed in serum exosomes isolated from gastric cancer patients, as confirmed by high-throughput sequencing and subsequent experiments. Coculture of gastric cancer-derived exosomes overexpressing miR-128-3p with human umbilical vein endothelial cells (HUVECs) significantly enhanced HUVEC proliferation, migratio n and angiogenesis. Bioinformatics analysis suggested SASH1 as the target gene of miR-128-3p. The dual luciferase assay and Western blot analysis results confirmed that miR-128-3p directly targeted SASH1 to inhibit its expression in HUVECs. Therefore, this study provides preliminary evidence that gastric cancer-derived exosomal miR-128-3p promotes tumour angiogenesis by targeting SASH1, reveals the potential diagnostic and therapeutic value of cancer-derived exosomal miR-128-3p, and provides new insights into the novel molecular mechanisms regulating metastasis. This study provides further information for understanding the role of gastric cancer-derived exosomal miR-128-3p in cancer progression and to discover new therapeutic targets.
Collapse
Affiliation(s)
- Hao Yan
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Xinyu Cai
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jianna Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Hongpeng Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Hongwen Wu
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Jingbo Zhang
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Lanzhi Xu
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Shizheng Liu
- Department of Gastroenterology, Shandong Public Health Clinical Center, Shandong University, Jinan, China
| | - Yuanwei Zang
- Department of Urology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Shanshan Fu
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
31
|
Kowolik E, Szczygieł D, Szczygieł M, Drzał A, Vemuri K, Olsson AK, Griffioen AW, Nowak-Sliwinska P, Wolnicka-Glubisz A, Elas M. Preclinical Photodynamic Therapy Targeting Blood Vessels with AGuIX ® Theranostic Nanoparticles. Cancers (Basel) 2024; 16:3924. [PMID: 39682113 DOI: 10.3390/cancers16233924] [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: 10/30/2024] [Revised: 11/13/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Background: Glioblastoma multiforme (GBM) is the most common highly aggressive, primary malignant brain tumor in adults. Current experimental strategies include photodynamic therapy (PDT) and new drug delivery technologies such as nanoparticles, which could play a key role in the treatment, diagnosis, and imaging of brain tumors. Objectives: The purpose of this study was to test the efficacy of PDT using AGuIX-TPP, a polysiloxane-based nanoparticle (AGuIX) that contains TPP (5,10,15,20-tetraphenyl-21H,23H-porphine), in biological models of glioblastoma multiforme and to investigate the vascular mechanisms of action at multiple complexity levels. Methods: PDT effects were studied in monolayer and spheroid cell culture, as well as tumors in chicken chorioallantoic membranes (CAMs) and in mice were studied. Results: Treatment was effective in both endothelial ECRF and glioma U87 cells, as well as in the inhibition of growth of the glioma spheroids. PDT using AGuIX-TPP inhibited U87 tumors growing in CAM and destroyed their vascularization. The U87 tumors were also grown in nude mice. Their vascular network, as well as oxygen partial pressure, were assessed using ultrasound and EPR oximetry. The treatment damaged tumor vessels and slightly decreased oxygen levels. Conclusions: PDT with AGuIX-TPP was effective against glioma cells, spheroids, and tumors; however, in mice, its efficacy appeared to be strongly related to the presence of blood vessels in the tumor before the treatment.
Collapse
Affiliation(s)
- Ewa Kowolik
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Dariusz Szczygieł
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Małgorzata Szczygieł
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Agnieszka Drzał
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Kalyani Vemuri
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Husargatan 3, SE-75123 Uppsala, Sweden
| | - Anna-Karin Olsson
- Department of Medical Biochemistry and Microbiology, Biomedical Center, Uppsala University, Husargatan 3, SE-75123 Uppsala, Sweden
| | - Arjan W Griffioen
- Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam UMC, Cancer Center Amsterdam, 1081 HV Amsterdam, The Netherlands
| | - Patrycja Nowak-Sliwinska
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland
- Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
- Translational Research Center in Oncohaematology, 1211 Geneva, Switzerland
| | - Agnieszka Wolnicka-Glubisz
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| | - Martyna Elas
- Department of Biophysics and Cancer Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 7 Gronostajowa Street, 31-387 Krakow, Poland
| |
Collapse
|
32
|
Yao Z, Xue K, Chen J, Zhang Y, Zhang G, Zheng Z, Li Z, Li Z, Wang F, Sun X, Shen L, Mao C, Lin C. Biliverdin improved angiogenesis and suppressed apoptosis via PI3K/Akt-mediated Nrf2 antioxidant system to promote ischemic flap survival. Free Radic Biol Med 2024; 225:35-52. [PMID: 39332540 DOI: 10.1016/j.freeradbiomed.2024.09.042] [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: 08/15/2024] [Revised: 09/21/2024] [Accepted: 09/24/2024] [Indexed: 09/29/2024]
Abstract
Plastic and reconstructive surgeons frequently utilize random skin flap transplantation to repair skin defects. However, the procedure carries a substantial risk of necrosis. Previous research has suggested that Biliverdin (Bv), the main component of Calculus Bovis, possessed potent anti-ischemic properties, making it a potential therapeutic agent for skin flap survival. Hence, in this study, the potential of Bv in promoting flap survival has been comprehensively investigated. Network pharmacology analysis revealed that the pharmacological effects of Bv on ischemic diseases may be attributed to its modulation of various signaling molecules, including the PI3K-Akt pathway. In vitro results demonstrated that Bv treatment significantly promoted angiogenesis in human umbilical vein endothelial cells (HUVEC), even in the presence of H2O2. This was evident by the increased cell proliferation, enhanced migration, and improved tube formation. Bv also effectively attenuated the intracellular generation of reactive oxygen species (ROS) induced by H2O2, which was achieved by suppressing mitochondrial ROS production through the PI3K/Akt-mediated activation of Nrf2/HO-1 signaling pathway. Consequently, Bv treatment led to a significant reduction in apoptosis and an increase in cell viability of HUVEC. Furthermore, in vivo experiment demonstrated that Bv treatment vastly elevated flap survival through enhancing angiogenesis while decreasing oxidative stress and apoptosis, which was comparable to the results of positive control of N-acetylcysteine (Nac). In conclusion, this study not only established a solid foundation for future study on therapeutic potential of Bv, but also proposed a promising treatment approach for enhancing the success rate of flap transplants and other ischemic-related tissue repair.
Collapse
Affiliation(s)
- Zhe Yao
- Department of Burn, Wound Repair and Regenerative Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Kaikai Xue
- Department of Burn, Wound Repair and Regenerative Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jinghao Chen
- Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Yu Zhang
- Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Guojian Zhang
- Department of Burn, Wound Repair and Regenerative Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zimin Zheng
- Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou, China
| | - Zihao Li
- Department of Burn, Wound Repair and Regenerative Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China; Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zi Li
- Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Fulin Wang
- Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoqi Sun
- Department of Psychiatry, Ruian Fifth People's Hospital, China
| | - Liyan Shen
- Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Cong Mao
- Zhejiang Provincial Key Laboratory of Orthopedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Cai Lin
- Department of Burn, Wound Repair and Regenerative Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| |
Collapse
|
33
|
Xu J, Tang Z. Progress on angiogenic and antiangiogenic agents in the tumor microenvironment. Front Oncol 2024; 14:1491099. [PMID: 39629004 PMCID: PMC11611712 DOI: 10.3389/fonc.2024.1491099] [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: 09/04/2024] [Accepted: 10/31/2024] [Indexed: 12/06/2024] Open
Abstract
The development of tumors and their metastasis relies heavily on the process of angiogenesis. When the volume of a tumor expands, the resulting internal hypoxic conditions trigger the body to enhance the production of various angiogenic factors. These include vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), platelet-derived growth factor (PDGF), and transforming growth factor-α (TGF-α), all of which work together to stimulate the activation of endothelial cells and catalyze angiogenesis. Antiangiogenic therapy (AAT) aims to normalize tumor blood vessels by inhibiting these angiogenic signals. In this review, we will explore the molecular mechanisms of angiogenesis within the tumor microenvironment, discuss traditional antiangiogenic drugs along with their limitations, examine new antiangiogenic drugs and the advantages of combination therapy, and consider future research directions in the field of antiangiogenic drugs. This comprehensive overview aims to provide insights that may aid in the development of more effective anti-tumor treatments.
Collapse
Affiliation(s)
| | - Zhihua Tang
- Department of Pharmacy, Shaoxing People’s Hospital, Shaoxing, China
| |
Collapse
|
34
|
Bazan NG, Obenaus A, Khoutorova L, Mukherjee PK, Jun B, Semikov R, Belayev L. Elovanoids, a Novel Class of Lipid Mediators, Are Neuroprotective in a Traumatic Brain Injury Model in Rats. Biomedicines 2024; 12:2555. [PMID: 39595120 PMCID: PMC11591722 DOI: 10.3390/biomedicines12112555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/22/2024] [Accepted: 11/01/2024] [Indexed: 11/28/2024] Open
Abstract
BACKGROUND In the United States, traumatic brain injury (TBI) contributes significantly to mortality and morbidity. Elovanoids (ELVs), a novel class of homeostatic lipid mediators we recently discovered and characterized, have demonstrated neuroprotection in experimental stroke models but have never been tested after TBI. METHODS A moderate fluid-percussion injury (FPI) model was used on male rats that were treated with ELVs by intravenous (IV) or intranasal (IN) delivery. In addition, using liquid chromatography-mass spectrometry (LC-MS/MS), we examined whether ELVs could be detected in brain tissue after IN delivery. RESULTS ELVs administered intravenously 1 h after FPI improved behavior on days 2, 3, 7, and 14 by 20, 23, 31, and 34%, respectively, and preserved hippocampal CA3 and dentate gyrus (DG) volume loss compared to the vehicle. Whole-brain tractography revealed that ELV-IV treatment increased corpus callosum white matter fibers at the injury site. In comparison to treatment with saline on days 2, 3, 7, and 14, ELVs administered intranasally at 1 h and 24 h after FPI showed improved neurological scores by 37, 45, 41, and 41%. T2-weighted imaging (T2WI) abnormalities, such as enlarged ventricles and cortical thinning, were reduced in rats treated by ELV-IN delivery compared to the vehicle. On day 3, ELVs were detected in the striatum and ipsilateral cortex of ELV-IN-treated rats. CONCLUSION We have demonstrated that both ELV-IN and ELV-IV administration offer high-grade neuroprotection that can be selectively supplied to the brain. This discovery may lead to innovative therapeutic targets for secondary injury cascade prevention following TBI.
Collapse
Affiliation(s)
- Nicolas G. Bazan
- Neuroscience Center of Excellence, School of Medicine, LSU Health New Orleans, New Orleans, LA 70112, USA; (L.K.); (P.K.M.); (R.S.)
| | - Andre Obenaus
- Division of Biomedical Sciences, University of California Riverside, Riverside, CA 92507, USA;
| | - Larissa Khoutorova
- Neuroscience Center of Excellence, School of Medicine, LSU Health New Orleans, New Orleans, LA 70112, USA; (L.K.); (P.K.M.); (R.S.)
| | - Pranab K. Mukherjee
- Neuroscience Center of Excellence, School of Medicine, LSU Health New Orleans, New Orleans, LA 70112, USA; (L.K.); (P.K.M.); (R.S.)
| | - Bokkyoo Jun
- Neuroscience Center of Excellence, School of Medicine, LSU Health New Orleans, New Orleans, LA 70112, USA; (L.K.); (P.K.M.); (R.S.)
| | - Rostyslav Semikov
- Neuroscience Center of Excellence, School of Medicine, LSU Health New Orleans, New Orleans, LA 70112, USA; (L.K.); (P.K.M.); (R.S.)
- Audubon Bioscience, Houston, TX 77021, USA
| | - Ludmila Belayev
- Neuroscience Center of Excellence, School of Medicine, LSU Health New Orleans, New Orleans, LA 70112, USA; (L.K.); (P.K.M.); (R.S.)
| |
Collapse
|
35
|
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
|
36
|
Luo AC, Wang J, Wang K, Zhu Y, Gong L, Lee U, Li X, Tremmel DM, Lin RZ, Ingber DE, Gorman J, Melero-Martin JM. A streamlined method to generate endothelial cells from human pluripotent stem cells via transient doxycycline-inducible ETV2 activation. Angiogenesis 2024; 27:779-795. [PMID: 38969874 PMCID: PMC11577265 DOI: 10.1007/s10456-024-09937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/28/2024] [Indexed: 07/07/2024]
Abstract
The development of reliable methods for producing functional endothelial cells (ECs) is crucial for progress in vascular biology and regenerative medicine. In this study, we present a streamlined and efficient methodology for the differentiation of human induced pluripotent stem cells (iPSCs) into induced ECs (iECs) that maintain the ability to undergo vasculogenesis in vitro and in vivo using a doxycycline-inducible system for the transient expression of the ETV2 transcription factor. This approach mitigates the limitations of direct transfection methods, such as mRNA-mediated differentiation, by simplifying the protocol and enhancing reproducibility across different stem cell lines. We detail the generation of iPSCs engineered for doxycycline-induced ETV2 expression and their subsequent differentiation into iECs, achieving over 90% efficiency within four days. Through both in vitro and in vivo assays, the functionality and phenotypic stability of the derived iECs were rigorously validated. Notably, these cells exhibit key endothelial markers and capabilities, including the formation of vascular networks in a microphysiological platform in vitro and in a subcutaneous mouse model. Furthermore, our results reveal a close transcriptional and proteomic alignment between the iECs generated via our method and primary ECs, confirming the biological relevance of the differentiated cells. The high efficiency and effectiveness of our induction methodology pave the way for broader application and accessibility of iPSC-derived ECs in scientific research, offering a valuable tool for investigating endothelial biology and for the development of EC-based therapies.
Collapse
Affiliation(s)
- Allen Chilun Luo
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
| | - Jiuhai Wang
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Yonglin Zhu
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Liyan Gong
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Umji Lee
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Xiang Li
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Daniel M Tremmel
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA
| | - Donald E Ingber
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
- Vascular Biology Program, Boston Children's Hospital, Boston, MA, 02115, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Cambridge, MA, 02138, USA
| | - James Gorman
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, 300 Longwood Ave, Boston, MA, 02115, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, 02115, USA.
- Harvard Stem Cell Institute, Cambridge, MA, 02138, USA.
| |
Collapse
|
37
|
Zhao Y, Zhang P, Zhang J. Microglia-mediated endothelial protection: the role of SHPL-49 in ischemic stroke. Biomed Pharmacother 2024; 180:117530. [PMID: 39388998 DOI: 10.1016/j.biopha.2024.117530] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/23/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
It was previously shown that SHPL-49, a glycoside derivative of salidroside formed through structural modification, exhibited neuroprotective effects in a rat cerebral ischemia model of permanent middle cerebral artery occlusion (pMCAO). Additionally, SHPL-49 enhanced the mRNA expression of vascular endothelial growth factor-a (Vegf-a) in macrophages. Microglia, functioning as resident macrophages within the brain, promptly respond to cerebral ischemia and engage in interactions with the cells of the Glial-Vascular Unit to orchestrate nerve injury responses. We postulated that the neuroprotective effects of SHPL-49 were mediated through microglia-dependent amelioration of endothelial dysfunction following cerebral ischemia. The present study demonstrates that SHPL-49 effectively mitigated microglia-dependent endothelial dysfunction in the pMCAO model by upregulating the expression of VEGF and suppressing the release of MMP-9 from microglia. Further MRI analyses confirmed that SHPL-49 significantly reduced nerve and endothelial function when microglia were depleted in the brains of pMCAO rats. The above phenomenon was also confirmed in the in vitro experiment investigating microglia-mediated brain endothelial cell function. Furthermore, we discovered that SHPL-49 activates the VEGFR2/Akt/eNOS pathways in endothelial cells and suppresses the p38 MAPK/MMP-9 pathways in microglia cells, thereby facilitating brain endothelial cell protection. Altogether, we have demonstrated that SHPL-49 effectively ameliorates endothelial dysfunction induced by cerebral ischemia through a microglia-dependent mechanism, thereby providing more valuable insights and references for the clinical evaluation of SHPL-49 injection for ischemic stroke.
Collapse
Affiliation(s)
- Yu Zhao
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Pei Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jiange Zhang
- The Research Center of Chiral Drugs, Innovation Research Institute of Traditional Chinese Medicine (IRI), Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
38
|
Long Y, Dan Y, Jiang Y, Ma J, Zhou T, Fang L, Wang Z. Colorectal Cancer Cell-Derived Extracellular Vesicles Promote Angiogenesis Through JAK/STAT3/VEGFA Signaling. BIOLOGY 2024; 13:873. [PMID: 39596828 PMCID: PMC11591796 DOI: 10.3390/biology13110873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/15/2024] [Accepted: 10/25/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND Angiogenesis plays a crucial role in the growth of colorectal cancer (CRC). Recent studies have identified extracellular vesicles (EVs) in the tumor microenvironment as important mediators of cell-to-cell communication. However, the specific role and mechanisms of CRC-derived EVs in regulating tumor angiogenesis remain to be further investigated. METHODS EVs were isolated from the conditioned medium of the CRC cells using ultracentrifugation. We investigated the effects of HT-29-derived EVs on tumor growth and angiogenesis in a subcutaneous HT-29 CRC tumor model in mice. Additionally, we evaluated the impact of HT-29-derived EVs on the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs). Subsequently, bioinformatics analysis was performed to identify relevant signaling pathways, and pathway inhibitors were used to block the activation of these pathways, aiming to elucidate their roles in angiogenesis. RESULTS We found that HT-29-derived EVs can promote tumor growth and angiogenesis in vivo, as well as significantly enhance the proliferation, migration, and tube formation of HUVECs. Bioinformatics analysis revealed that HT-29-derived EVs may regulate angiogenesis through the JAK/STAT3 signaling pathway. Specifically, we observed that CRC-derived EVs promoted the phosphorylation of STAT3 (p-STAT3) and the expression of VEGFA in the nucleus of HUVECs. Treatment with the STAT3 inhibitor Stattic reduced the nuclear expression of p-STAT3, which impaired its function as a transcription factor, thereby inhibiting VEGFA expression and the pro-angiogenic effects of CRC-derived EVs. CONCLUSIONS EVs derived from CRC cells promote CRC tumor angiogenesis by regulating VEGFA through the JAK/STAT3 pathway in endothelial cells.
Collapse
Affiliation(s)
- Yuqing Long
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China; (Y.L.); (Y.D.); (Y.J.); (J.M.); (T.Z.)
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yuxi Dan
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China; (Y.L.); (Y.D.); (Y.J.); (J.M.); (T.Z.)
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Yao Jiang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China; (Y.L.); (Y.D.); (Y.J.); (J.M.); (T.Z.)
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Jing Ma
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China; (Y.L.); (Y.D.); (Y.J.); (J.M.); (T.Z.)
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Tao Zhou
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China; (Y.L.); (Y.D.); (Y.J.); (J.M.); (T.Z.)
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Liaoqiong Fang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China; (Y.L.); (Y.D.); (Y.J.); (J.M.); (T.Z.)
- National Engineering Research Center of Ultrasound Medicine, Chongqing 401121, China
| | - Zhibiao Wang
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China; (Y.L.); (Y.D.); (Y.J.); (J.M.); (T.Z.)
- Chongqing Key Laboratory of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
- National Engineering Research Center of Ultrasound Medicine, Chongqing 401121, China
| |
Collapse
|
39
|
Liu X, Fan H, Chen Z, Liu C. Exploring the significance and potential mechanisms of hippo pathway-associated genes in prognosis of glioma patients. Discov Oncol 2024; 15:536. [PMID: 39382606 PMCID: PMC11464986 DOI: 10.1007/s12672-024-01391-5] [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: 05/25/2024] [Accepted: 09/24/2024] [Indexed: 10/10/2024] Open
Abstract
PURPOSE Despite the efforts of countless researchers to develop glioma treatment strategies, the current therapeutic effect of glioma is still not ideal, and it is necessary to further explore the mechanism to guide treatment. Thus, this study aims to introduce a novel approach for predicting patient prognosis and guiding further treatment interventions. METHODS Initially, we conducted a differential gene expression analysis to identify Hippo pathway-associated genes overexpressed in tumors and determined genes correlated with prognosis. Subsequently, employing cluster analysis, we categorized samples into two groups and performed further analyses including prediction, immune cell infiltration abundance, and drug response rates. We utilized weighted gene co-expression analysis to reveal gene sets with high co-variation, delineate inter-sample gene correlation patterns, and conduct enrichment analysis. Prognostic models were built using ten machine learning algorithms combined in 101 different combinations, followed by evaluation and validation. Immune infiltration analysis, differential expression analysis of depleted T cell-related markers, drug sensitivity analysis, and exploration of pathway dysregulation were performed for different risk groups. Quality control and batch integration were performed, and single-cell data were analyzed using dimensionality reduction clustering algorithms and annotation tools to evaluate the activity of the prognostic model in malignant cells. RESULTS We conducted data filtering to identify genes overexpressed in tumors, intersecting these genes with Hippo pathway-related genes, identifying 62 genes correlated with prognosis, and performing cluster analysis to divide tumor tissues into two groups. Cluster 2 exhibited a poorer prognosis and demonstrated differences in immune cell infiltration. Utilizing weighted gene co-expression analysis on Cluster 2, we identified gene modules, conducted functional enrichment analysis, and delineated pathways. Employing a combined model based on ten machine learning algorithm combinations, we selected the optimal prognostic model system and validated the model's predictive ability within the dataset. Through immune-related analysis and drug sensitivity analysis, we uncovered differences in immune infiltration and varying sensitivities to chemotherapy drugs. Additionally, the enrichment analysis of gene set revealed discrepancies in upregulation within relevant pathways between the high and low-risk groups. Finally, annotation and evaluation of malignant cells via single-cell analysis showed increased activity of the prognostic model and variations in distribution across different prognostic levels in malignant cells. CONCLUSION This study introduces a novel approach utilizing the Hippo pathway and associated genes for glioma prognosis research, demonstrating the potential and significance of this method in evaluating the outcome for patients with glioma. These findings hold substantial clinical significance in guiding therapy and predicting outcomes for individuals diagnosed with glioma, offering significant clinical utility.
Collapse
Affiliation(s)
- XuKai Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China
| | - Hongjun Fan
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China
| | - Zebo Chen
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China
| | - Chao Liu
- Department of Neurosurgery, Central Hospital of Zhuzhou, Zhuzhou, Hunan, China.
| |
Collapse
|
40
|
Hough RF, Alvira CM, Bastarache JA, Erzurum SC, Kuebler WM, Schmidt EP, Shimoda LA, Abman SH, Alvarez DF, Belvitch P, Bhattacharya J, Birukov KG, Chan SY, Cornfield DN, Dudek SM, Garcia JGN, Harrington EO, Hsia CCW, Islam MN, Jonigk DD, Kalinichenko VV, Kolb TM, Lee JY, Mammoto A, Mehta D, Rounds S, Schupp JC, Shaver CM, Suresh K, Tambe DT, Ventetuolo CE, Yoder MC, Stevens T, Damarla M. Studying the Pulmonary Endothelium in Health and Disease: An Official American Thoracic Society Workshop Report. Am J Respir Cell Mol Biol 2024; 71:388-406. [PMID: 39189891 PMCID: PMC11450313 DOI: 10.1165/rcmb.2024-0330st] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Indexed: 08/28/2024] Open
Abstract
Lung endothelium resides at the interface between the circulation and the underlying tissue, where it senses biochemical and mechanical properties of both the blood as it flows through the vascular circuit and the vessel wall. The endothelium performs the bidirectional signaling between the blood and tissue compartments that is necessary to maintain homeostasis while physically separating both, facilitating a tightly regulated exchange of water, solutes, cells, and signals. Disruption in endothelial function contributes to vascular disease, which can manifest in discrete vascular locations along the artery-to-capillary-to-vein axis. Although our understanding of mechanisms that contribute to endothelial cell injury and repair in acute and chronic vascular disease have advanced, pathophysiological mechanisms that underlie site-specific vascular disease remain incompletely understood. In an effort to improve the translatability of mechanistic studies of the endothelium, the American Thoracic Society convened a workshop to optimize rigor, reproducibility, and translation of discovery to advance our understanding of endothelial cell function in health and disease.
Collapse
|
41
|
Lessiak U, Melchert M, Walter I, Kummer S, Nell B, Tschulenk W, Pratscher B. Isolation-protocol, characterization, and in-vitro performance of equine umbilical vein endothelial cells. Front Vet Sci 2024; 11:1421946. [PMID: 39411390 PMCID: PMC11473255 DOI: 10.3389/fvets.2024.1421946] [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: 04/23/2024] [Accepted: 09/12/2024] [Indexed: 10/19/2024] Open
Abstract
Angiogenesis plays a crucial role in various physiological and pathological conditions. However, research in equine angiogenesis is relative limited, necessitating the development of suitable in-vitro models. To effectively analyze angiogenesis in-vitro, it is essential to target the specific cells responsible for this process, namely endothelial cells. Human umbilical vein endothelial cells (HUVECs) are one of the most used in vitro models for studying angiogenesis in humans. Serving as an equivalent to HUVECs, we present a comprehensive isolation protocol for equine umbilical vein endothelial cells (EqUVECs) with relatively minimal requirements, thereby enhancing accessibility for researchers. Umbilical cords obtained from five foals were used to isolate endothelial cells, followed by morphological and immunohistochemical identification. Performance of the cells in various assays commonly used in angiogenesis research was studied. Additionally, EqUVEC expression of vascular endothelial growth factor (VEGF) was assessed using ELISA. EqUVECs exhibited endothelial characteristics, forming a homogeneous monolayer with distinctive morphology. Immunohistochemical staining confirmed positive expression of key endothelial markers including von Willebrand factor (vWF), CD31, and vascular endothelial growth factor receptor-2 (VEGFR-2). Furthermore, performance assessments in in-vitro assays demonstrated the viability, proliferation, migration, tube formation and VEGF-expression capabilities of EqUVECs. The findings suggest that EqUVECs are a promising in-vitro model for studying equine angiogenesis, offering a foundation for further investigations into equine-specific vascular processes and therapeutic interventions.
Collapse
Affiliation(s)
- Ulrike Lessiak
- Ophthalmology Unit, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Maria Melchert
- Centre for Animal Reproduction, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Ingrid Walter
- Department of Biomedical Science and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- VetCore Facility for Research, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Stefan Kummer
- VetCore Facility for Research, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Nell
- Ophthalmology Unit, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Waltraud Tschulenk
- Department of Biomedical Science and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Barbara Pratscher
- Research Unit Internal Medicine, Department of Companion Animals and Horses, University of Veterinary Medicine Vienna, Vienna, Austria
| |
Collapse
|
42
|
Kroenig J, Görg C, Prosch H, Von Schumann L, Westhoff CC, Alhyari A, Koenig FRM, Findeisen H, Safai Zadeh E. Perfusion Patterns of Peripheral Pulmonary Metastasis Using Contrast-Enhanced Ultrasound (CEUS) and Their Correlation with Immunohistochemically Detected Vascularization Pattern. Cancers (Basel) 2024; 16:3365. [PMID: 39409985 PMCID: PMC11475622 DOI: 10.3390/cancers16193365] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/20/2024] Open
Abstract
PURPOSE Description of the perfusion of pulmonary metastasis by contrast-enhanced ultrasound (CEUS) and their correlation with vascularization patterns represented by immunohistochemical CD34 endothelial staining. PATIENTS AND METHODS The data of 54 patients with histologic proven peripheral pulmonary metastasis, investigated between 2004 and 2023 by CEUS. These CEUS parameters were evaluated: time to enhancement (TE), categorized as early pulmonary-arterial (PA) or delayed bronchial-arterial (BA) patterns; extent of enhancement (EE), either marked or reduced; homogeneity of enhancement (HE), homogeneous or inhomogeneous; and decrease of enhancement (DE), rapid washout (<120 s) or late washout (≥120 s). Additionally, tissue samples in 45 cases (83.3%) were stained with CD34 antibody for immunohistochemical analysis. RESULTS In total, 4 lesions (7.4 %) exhibited PA enhancement, and 50 lesions (92.6%) demonstrated BA enhancement. Furthermore, 37 lesions (68.5%) showed marked enhancement, while 17 lesions (31.5%) exhibited reduced enhancement. The enhancement was homogeneous in 28 lesions (51.86%) and inhomogeneous in 26 lesions (48.14%). Additionally, 53 lesions (98.1%) displayed a rapid washout. A chaotic vascular pattern indicative of a bronchial arterial blood supply was identified in all cases (45/45, 100%), including all 4 lesions with PA enhancement. CONCLUSION Pulmonary metastases in CEUS predominantly reveal bronchial arterial enhancement and a rapid washout. Regarding EE and HE, pulmonary metastases show heterogeneous perfusion patterns. A PA enhancement in CEUS does not exclude BA neoangiogenesis.
Collapse
Affiliation(s)
- Johannes Kroenig
- Lung Center Mainz, Clinic for Pneumology, Center for Thoracic Diseases, University Medical Center Mainz, 55131 Mainz, Germany;
- Interdisciplinary Center of Ultrasound Diagnostics, Gastroenterology, Endocrinology, Metabolism and Clinical Infectiology, University Hospital Giessen and Marburg, Philipp University of Marburg, Baldingerstraße, 35033 Marburg, Germany (A.A.)
| | - Christian Görg
- Interdisciplinary Center of Ultrasound Diagnostics, Gastroenterology, Endocrinology, Metabolism and Clinical Infectiology, University Hospital Giessen and Marburg, Philipp University of Marburg, Baldingerstraße, 35033 Marburg, Germany (A.A.)
| | - Helmut Prosch
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Wien, Austria; (H.P.); (F.R.M.K.)
| | - Lara Von Schumann
- Interdisciplinary Center of Ultrasound Diagnostics, Gastroenterology, Endocrinology, Metabolism and Clinical Infectiology, University Hospital Giessen and Marburg, Philipp University of Marburg, Baldingerstraße, 35033 Marburg, Germany (A.A.)
| | - Christina C. Westhoff
- Institute of Pathology, University Hospital Giessen and Marburg, Philipps University Marburg, Baldingerstraße, 35043 Marburg, Germany
| | - Amjad Alhyari
- Interdisciplinary Center of Ultrasound Diagnostics, Gastroenterology, Endocrinology, Metabolism and Clinical Infectiology, University Hospital Giessen and Marburg, Philipp University of Marburg, Baldingerstraße, 35033 Marburg, Germany (A.A.)
| | - Felix R. M. Koenig
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Wien, Austria; (H.P.); (F.R.M.K.)
| | - Hajo Findeisen
- Department for Internal Medicine, Red Cross Hospital Bremen, 28209 Bremen, Germany
| | - Ehsan Safai Zadeh
- Interdisciplinary Center of Ultrasound Diagnostics, Gastroenterology, Endocrinology, Metabolism and Clinical Infectiology, University Hospital Giessen and Marburg, Philipp University of Marburg, Baldingerstraße, 35033 Marburg, Germany (A.A.)
- Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, 1090 Wien, Austria; (H.P.); (F.R.M.K.)
| |
Collapse
|
43
|
Lee U, Zhang Y, Zhu Y, Luo AC, Gong L, Tremmel DM, Kim Y, Villarreal VS, Wang X, Lin RZ, Cui M, Ma M, Yuan K, Wang K, Chen K, Melero-Martin JM. Robust differentiation of human pluripotent stem cells into mural progenitor cells via transient activation of NKX3.1. Nat Commun 2024; 15:8392. [PMID: 39349465 PMCID: PMC11442894 DOI: 10.1038/s41467-024-52678-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 09/13/2024] [Indexed: 10/02/2024] Open
Abstract
Mural cells are central to vascular integrity and function. In this study, we demonstrate the innovative use of the transcription factor NKX3.1 to guide the differentiation of human induced pluripotent stem cells into mural progenitor cells (iMPCs). By transiently activating NKX3.1 in mesodermal intermediates, we developed a method that diverges from traditional growth factor-based differentiation techniques. This approach efficiently generates a robust iMPC population capable of maturing into diverse functional mural cell subtypes, including smooth muscle cells and pericytes. These iMPCs exhibit key mural cell functionalities such as contractility, deposition of extracellular matrix, and the ability to support endothelial cell-mediated vascular network formation in vivo. Our study not only underscores the fate-determining significance of NKX3.1 in mural cell differentiation but also highlights the therapeutic potential of these iMPCs. We envision these insights could pave the way for a broader use of iMPCs in vascular biology and regenerative medicine.
Collapse
Affiliation(s)
- Umji Lee
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Yadong Zhang
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Yonglin Zhu
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Allen Chilun Luo
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
| | - Liyan Gong
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Daniel M Tremmel
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Yunhye Kim
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA, USA
| | | | - Xi Wang
- Department of Biological and Environmental Engineering, Cornell University, NY, USA
| | - Ruei-Zeng Lin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA
- Department of Surgery, Harvard Medical School, Boston, MA, USA
| | - Miao Cui
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA
| | - Minglin Ma
- Department of Biological and Environmental Engineering, Cornell University, NY, USA
| | - Ke Yuan
- Division of Pulmonary Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kai Wang
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, USA.
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing, China.
| | - Kaifu Chen
- Department of Cardiology, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Juan M Melero-Martin
- Department of Cardiac Surgery, Boston Children's Hospital, Boston, MA, USA.
- Department of Surgery, Harvard Medical School, Boston, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
44
|
Zaytseva AV, Karelina NR, Bedyaev EV, Vavilov PS, Sesorova IS, Mironov AA. During Postnatal Ontogenesis, the Development of a Microvascular Bed in an Intestinal Villus Depends on Intussusceptive Angiogenesis. Int J Mol Sci 2024; 25:10322. [PMID: 39408652 PMCID: PMC11476829 DOI: 10.3390/ijms251910322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 08/02/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
The mechanisms responsible for the growth and development of vascular beds in intestinal villi during postnatal ontogenesis remain enigmatic. For instance, according to the current consensus, in the sprouting type of angiogenesis, there is no blood flow in the rising capillary sprout. However, it is known that biomechanical forces resulting from blood flow play a key role in these processes. Here, we present evidence for the existence of the intussusception type of angiogenesis during the postnatal development of micro-vessel patterns in the intestinal villi of rats. This process is based on the high-level flattening of blood capillaries on the flat surfaces of intestinal villi, contacts among the opposite apical plasma membrane of endothelial cells in the area of inter-endothelial contacts, or the formation of bridges composed of blood leucocytes or local microthrombi. We identified factors that, in our opinion, ensure the splitting of the capillary lumen and the formation of two parallel vessels. These phenomena are in agreement with previously described features of intussusception angiogenesis.
Collapse
Affiliation(s)
- Anna V. Zaytseva
- Department of Anatomy, Saint Petersburg State Pediatric Medical University, Saint Petersburg 194100, Russia;
| | - Natalia R. Karelina
- Department of Anatomy, Saint Petersburg State Pediatric Medical University, Saint Petersburg 194100, Russia;
| | - Eugeny V. Bedyaev
- Department of Anatomy, Ivanovo State Medical University, Ivanovo 153012, Russia; (E.V.B.); (P.S.V.); (I.S.S.)
| | - Pavel S. Vavilov
- Department of Anatomy, Ivanovo State Medical University, Ivanovo 153012, Russia; (E.V.B.); (P.S.V.); (I.S.S.)
| | - Irina S. Sesorova
- Department of Anatomy, Ivanovo State Medical University, Ivanovo 153012, Russia; (E.V.B.); (P.S.V.); (I.S.S.)
| | - Alexander A. Mironov
- Department of Cell Biology, IFOM ETS—The AIRC Institute of Molecular Oncology, Via Adamello, 16, 20139 Milan, Italy
| |
Collapse
|
45
|
Ayoub NM, Sardiah S, Al-Share QY, Alkader MS. Exploring angiogenic pathways in breast cancer: Clinicopathologic correlations and prognostic implications based on gene expression profiles from a large-scale genomic dataset. PLoS One 2024; 19:e0310557. [PMID: 39302921 DOI: 10.1371/journal.pone.0310557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 09/03/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Angiogenesis inhibitors targeting VEGF, or its receptors have consistently produced disappointing clinical outcomes in breast cancer. Therefore, there is an urgent need to explore alternative angiogenic pathways in breast cancer. This study aimed to describe the gene expression of pivotal pro-angiogenic genes in breast cancer and to further analyze the associations with the clinicopathologic tumor features, prognostic factors, and overall survival. Such findings would expand the understanding of the role of different angiogenic pathways in breast cancer pathogenesis and identify patients at risk of more aggressive disease who could be eligible for intense treatment regimens. Additionally, exploring angiogenic pathways helps identify new potential drug targets for breast cancer. METHODS The mRNA expression levels for eight pro-angiogenic genes [VEGFA, HGF, FGF1, FGF2, ANGPT1, ANGPT2, PDGFA, and PDGFB] were obtained from the METABRIC (Molecular Taxonomy of Breast Cancer International Consortium) dataset available at cBioPortal public domain. Pertinent demographic and tumor information were retrieved. RESULTS VEGFA and ANGPT2 genes had the highest expression levels with average mRNA log intensities of 7.18±0.7 and 7.11±0.53, respectively. VEGFA expression was not correlated with the expression of other pro-angiogenic genes, the clinicopathologic tumor features, and the overall survival of patients. FGF1, ANGPT1, and PDGFA mRNA levels were negatively correlated with the age of patients at diagnosis. The expression of FGF1 and FGF2 correlated inversely with tumor size and the Nottingham Prognostic Index (p = 0.03 and p = 0.002, respectively). Expression of HGF was significantly associated with advanced tumor stage (p<0.05). Expression of ANGPT1 and ANGPT2 was associated with hormone receptor-negative status and the non-luminal subtypes. PDGFB expression was significantly higher in patients with high-grade disease and HER2-positive status. Patients with high expression status of ANGPT2 and PDGFB had significantly reduced overall survival compared to those with low expression levels of these genes (p = 0.004 and p = 0.0001, respectively). CONCLUSIONS In this dataset of patients with breast cancer, the expression levels of 8 different pro-angiogenic genes revealed remarkable differences in terms of their association with clinicopathologic tumor characteristics and prognosis. The expression of ANGPTs and PDGFs was associated with adverse tumor features, worse prognosis, and reduced survival in patients. Targeting ANGPTs and PDGF pathways could provide new insights for effective anti-angiogenic drugs in breast cancer.
Collapse
Affiliation(s)
- Nehad M Ayoub
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Salam Sardiah
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Qusai Y Al-Share
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid, Jordan
| | - Mohammad S Alkader
- Department of Medical Oncology, Military Cancer Center, Jordanian Royal Medical Services, Amman, Jordan
| |
Collapse
|
46
|
Liu X, Cheng R, Cao H, Wu L. 3D-Cultured MC3T3-E1-Derived Exosomes Promote Endothelial Cell Biological Function under the Effect of LIPUS. Biomolecules 2024; 14:1154. [PMID: 39334920 PMCID: PMC11430381 DOI: 10.3390/biom14091154] [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: 08/18/2024] [Revised: 09/04/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Porous Ti-6Al-4V scaffold materials can be used to heal massive bone defects because they can provide space for vascularisation and bone formation. During new bone tissue development, rapid vascular ingrowth into scaffold materials is very important. Osteoblast-derived exosomes are capable of facilitating angiogenesis-osteogenesis coupling. Low-intensity pulsed ultrasound (LIPUS) is a physical therapy modality widely utilised in the field of bone regeneration and has been proven to enhance the production and functionality of exosomes on two-dimensional surfaces. The impact of LIPUS on exosomes derived from osteoblasts cultured in three dimensions remains to be elucidated. In this study, exosomes produced by osteoblasts on porous Ti-6Al-4V scaffold materials under LIPUS and non-ultrasound stimulated conditions were co-cultured with endothelial cells. The findings indicated that the exosomes were consistently and stably taken up by the endothelial cells. Compared to the non-ultrasound group, the LIPUS group facilitated endothelial cell proliferation and angiogenesis. After 24 h of co-culture, the migration ability of endothelial cells in the LIPUS group was 17.30% higher relative to the non-ultrasound group. LIPUS may represent a potentially viable strategy to promote the efficacy of osteoblast-derived exosomes to enhance the angiogenesis of porous Ti-6Al-4V scaffold materials.
Collapse
Affiliation(s)
- Xiaohan Liu
- Department of Prosthodontics, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang 110002, China
| | - Rui Cheng
- Department of Prosthodontics, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang 110002, China
| | - Hongjuan Cao
- Department of Prosthodontics, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang 110002, China
| | - Lin Wu
- Department of Prosthodontics, School and Hospital of Stomatology, China Medical University, Shenyang 110002, China
- Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang 110002, China
| |
Collapse
|
47
|
Li Z, Nguyen Canh H, Takahashi K, Le Thanh D, Nguyen Thi Q, Yang R, Yoshimura K, Sato Y, Nguyen Thi K, Nakata H, Ikeda H, Kozaka K, Kobayashi S, Yagi S, Harada K. Histopathological growth pattern and vessel co-option in intrahepatic cholangiocarcinoma. Med Mol Morphol 2024; 57:200-217. [PMID: 38960952 PMCID: PMC11343874 DOI: 10.1007/s00795-024-00392-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 07/05/2024]
Abstract
Intrahepatic cholangiocarcinoma (iCCA) exhibits different blood imaging features and prognosis depending on histology. To clarity histopathological growth patterns (HGPs) and vascularization processes of iCCA, we collected 145 surgical specimens and histologically classified them into large bile duct (LBD) (20 cases), small bile duct (SBD) (54), cholangiolocarcinoma (CLC) (35), combined SBD-CLC (cSBD-CLC) (26), and ductal plate malformation (DPM) (10) (sub)types. According to the invasive pattern at the interface between tumor and adjacent background liver, HGPs were classified into desmoplastic, pushing, and replacing HGPs. Desmoplastic HGP predominated in LBD type (55.5%), while replacing HGP was common in CLC (82.9%) and cSBD-CLC (84.6%) subtypes. Desmoplastic HGP reflected angiogenesis, while replacing HGP showed vessel co-option in addition to angiogenesis. By evaluating microvessel density (MVD) using vascular markers, ELTD1 identified vessel co-option and angiogenesis, and ELTD1-positive MVD at invasive margin in replacing HGP was significantly higher than those in desmoplastic and pushing HGPs. REDD1, an angiogenesis-related marker, demonstrated preferably higher MVD in the tumor center than in other areas. iCCA (sub)types and HGPs were closely related to vessel co-option and immune-related factors (lymphatic vessels, lymphocytes, and neutrophils). In conclusion, HGPs and vascular mechanisms characterize iCCA (sub)types and vessel co-option linked to the immune microenvironment.
Collapse
Affiliation(s)
- Zihan Li
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Hiep Nguyen Canh
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Kenta Takahashi
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Dong Le Thanh
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Quynh Nguyen Thi
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Rui Yang
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Kaori Yoshimura
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Yasunori Sato
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan
| | - Khuyen Nguyen Thi
- Center of Pathology and Molecular Biology, National Cancer Hospital, Hanoi, Vietnam
| | - Hiroki Nakata
- Department of Clinical Engineering, Faculty of Health Sciences, Komatsu University, Komatsu, Japan
- Department of Integrative Cancer Therapy and Urology, Kanazawa University Graduate School of Medical Science, Kanazawa, Japan
| | - Hiroko Ikeda
- Department of Diagnostic Pathology, Kanazawa University Hospital, Kanazawa, Japan
| | - Kazuto Kozaka
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Satoshi Kobayashi
- Department of Radiology, Kanazawa University Hospital, Kanazawa, Japan
| | - Shintaro Yagi
- Department of Hepato-Biliary-Pancreatic Surgery and Transplantation, Kanazawa University, Kanazawa, Japan
| | - Kenichi Harada
- Department of Human Pathology, Kanazawa University Graduate School of Medicine, Kanazawa, 920-8640, Japan.
| |
Collapse
|
48
|
She X, Xu J, Zhang H, Yu C, Rao Z, Zhang J, Zhan W, Hu F, Song D, Li H, Luo X, Wang G, Hu J, Lai S. ETHE1 dampens colorectal cancer angiogenesis by promoting TC45 Dephosphorylation of STAT3 to inhibit VEGF-A expression. Cell Death Dis 2024; 15:631. [PMID: 39198402 PMCID: PMC11358511 DOI: 10.1038/s41419-024-07021-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/01/2024]
Abstract
Angiogenesis is critical for colorectal cancer (CRC) progression, but its mechanisms remain unclear. Here, we reveal that ethylmalonic encephalopathy protein 1 (ETHE1), an essential enzyme in hydrogen sulfide catabolism, inhibits VEGF-A expression and tumor angiogenesis in vitro and in vivo. Moreover, we find that this biological function of ETHE1 depends on the STAT3/VEGF-A pathway. Further investigation demonstrates that ETHE1 promotes the interaction between T cell protein tyrosine phosphatase (TC45) and STAT3, resulting in decreased STAT3 phosphorylation and inhibition of the STAT3 signaling pathway. In clinical samples, we find that ETHE1 is downregulated in CRC and positively correlates with survival outcomes of CRC patients. Meanwhile, the negative correlation of ETHE1 and VEGF-A expression is verified in CRC specimens, and the patients with low ETHE1 and high VEGF-A expression exhibits poorer prognosis. Collectively, our study identifies ETHE1 as a novel regulator of tumor angiogenesis, implying its potential as a prognostic biomarker and promising antiangiogenic target for CRC patients.
Collapse
Affiliation(s)
- Xiaowei She
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jialu Xu
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haokun Zhang
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Chengxin Yu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Zejun Rao
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Jiakun Zhang
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Wenli Zhan
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Fuqing Hu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Da Song
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Haijie Li
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Xuelai Luo
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Guihua Wang
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
- Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Disease, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Junbo Hu
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Senyan Lai
- GI Cancer Research Institute, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
49
|
Wan YX, Qi XW, Lian YY, Liu ZY, Wang H, Qiu YQ, Zhang CG, Li WN, Jiang HL, Yang DH, Zhao W, Chen ZS, Huang JC. Electroacupuncture facilitates vascular normalization by inhibiting Glyoxalase1 in endothelial cells to attenuate glycolysis and angiogenesis in triple-negative breast cancer. Cancer Lett 2024; 598:217094. [PMID: 38945204 DOI: 10.1016/j.canlet.2024.217094] [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: 01/04/2024] [Revised: 06/13/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Recent therapeutic strategies for the treatment of triple-negative breast cancer (TNBC) have shifted the focus from vascular growth factors to endothelial cell metabolism. This study highlights the underexplored therapeutic potential of peri-tumoral electroacupuncture, a globally accepted non-pharmacological intervention for TNBC, and molecular mechanisms. Our study showed that peri-tumoral electroacupuncture effectively reduced the density of microvasculature and enhanced vascular functionality in 4T1 breast cancer xenografts, with optimal effects on day 3 post-acupuncture. The timely integration of peri-tumoral electroacupuncture amplified the anti-tumor efficacy of paclitaxel. Multi-omics analysis revealed Glyoxalase 1 (Glo1) and the associated methylglyoxal-glycolytic pathway as key mediators of electroacupuncture-induced vascular normalization. Peri-tumoral electroacupuncture notably reduced Glo1 expression in the endothelial cells of 4T1 xenografts. Using an in vivo matrigel plug angiogenesis assay, we demonstrated that either Glo1 knockdown or electroacupuncture inhibited angiogenesis. In contrast, Glo1 overexpression increased blood vessel formation. In vitro pharmacological inhibition and genetic knockdown of Glo1 in human umbilical vein endothelial cells inhibited proliferation and promoted apoptosis via downregulating the methylglyoxal-glycolytic pathway. The study using the Glo1-silenced zebrafish model further supported the role of Glo1 in vascular development. This study underscores the pivotal role of Glo1 in peri-tumoral electroacupuncture, spotlighting a promising avenue for enhancing vascular normalization and improving TNBC treatment outcomes.
Collapse
Affiliation(s)
- Yu-Xiang Wan
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xue-Wei Qi
- Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100029, China
| | - Yan-Yan Lian
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ze-Yu Liu
- Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Hui Wang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu-Qin Qiu
- The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chun-Guang Zhang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Wen-Na Li
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hong-Lin Jiang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Dong-Hua Yang
- New York College of Traditional Chinese Medicine, Mineola, NY, 11501, USA
| | - Wei Zhao
- Guanganmen Hospital, Chinese Academy of Traditional Chinese Medicine, Beijing, 100029, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY, 11439, USA.
| | - Jin-Chang Huang
- The Third Affiliated Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
50
|
Abbasi-Malati Z, Khanicheragh P, Narmi MT, Mardi N, Khosrowshahi ND, Hiradfar A, Rezabakhsh A, Sadeghsoltani F, Rashidi S, Chegeni SA, Roozbahani G, Rahbarghazi R. Tumoroids, a valid preclinical screening platform for monitoring cancer angiogenesis. Stem Cell Res Ther 2024; 15:267. [PMID: 39183337 PMCID: PMC11346257 DOI: 10.1186/s13287-024-03880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024] Open
Abstract
In recent years, biologists and clinicians have witnessed prominent advances in in vitro 3D culture techniques related to biomimetic human/animal tissue analogs. Numerous data have confirmed that unicellular and multicellular (tumoroids) tumor spheroids with dense native cells in certain matrices are sensitive and valid analytical tools for drug screening, cancer cell dynamic growth, behavior, etc. in laboratory settings. Angiogenesis/vascularization is a very critical biological phenomenon to support oxygen and nutrients to tumor cells within the deep layer of solid masses. It has been shown that endothelial cell (EC)-incorporated or -free spheroid/tumoroid systems provide a relatively reliable biological platform for monitoring the formation of nascent blood vessels in micron/micrometer scales. Besides, the paracrine angiogenic activity of cells within the spheroid/tumoroid systems can be monitored after being treated with different therapeutic approaches. Here, we aimed to collect recent advances and findings related to the monitoring of cancer angiogenesis using unicellular and multicellular tumor spheroids. Vascularized spheroids/tumoroids can help us in the elucidation of mechanisms related to cancer formation, development, and metastasis by monitoring the main influencing factors.
Collapse
Affiliation(s)
- Zahra Abbasi-Malati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Khanicheragh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Narges Mardi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Amirataollah Hiradfar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Somayyeh Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Golbarg Roozbahani
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|