1
|
Qian C, Huang Y, Zhang S, Yang C, Zheng W, Tang W, Wan G, Wang A, Lu Y, Zhao Y. Integrated identification and mechanism exploration of bioactive ingredients from Salvia miltiorrhiza to induce vascular normalization. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 138:156427. [PMID: 39892310 DOI: 10.1016/j.phymed.2025.156427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 01/22/2025] [Accepted: 01/24/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND The clinical management of ischemic disease and cancer is complex, with disruptions in local vascular function and tumor angiogenesis contributing to blood stasis, which complicates treatment strategies. Salvia miltiorrhiza, a natural product, is known to restore vascular structure and function. However, its specific roles in concurrently addressing ischemic disease and cancer within the same organism remain poorly understood. PURPOSE This study aimed to explore the material basis, pharmacological effects, and underlying mechanisms of Salvia miltiorrhiza extract (SME) in promoting blood flow recovery in ischemic hindlimbs and inducing tumor vascular normalization. METHODS The pharmacological effects of SME were evaluated in a mouse model combining ischemic hindlimbs and tumors. Mice were administered low (SME-L) or high (SME-H) doses of SME daily, and the gastrocnemius muscle mass and tumor vascular structure were assessed. Laser Doppler perfusion imaging (LDPI) was used to monitor hindlimb blood flow recovery and tumor vascular perfusion. The pharmacokinetics of the key bioactive constituents in SME were characterized by liquid chromatography-mass spectrometry (LC-MS). Interactions between SME's active compounds and predicted targets were investigated using molecular docking, microscale thermophoresis (MST), and luciferase reporter assays. The synergistic effects of the primary components, Tanshinone I (Tan I) and Salvianolic acid A (Sal A), were analyzed through tube formation assays, enzyme-linked immunosorbent assays (ELISA), immunofluorescence staining, and western blot. RESULTS Phytochemical profiling revealed that SME contains several active compounds, including Danshensu, Sal A, Sal B, Tan IIA, and Tan I. SME treatment reduced the frequency of necrotic toes, increased muscle mass, and alleviated hypoxia in the gastrocnemius muscle. SME significantly improved tumor vascular perfusion and notably enhanced pericyte coverage and basement membrane integrity. Pharmacokinetic analysis identified Tan I and Sal A as the key bioactive components that promote vascular normalization. Tan I inhibited FoxO1, preventing endothelial cell activation induced by angiopoietin 2 (Ang2), while Sal A bound to Ang2, facilitating Tie2 activation mediated by Ang1. Both in vitro and in vivo results demonstrated that the combination of Tan I and Sal A exerted a synergistic therapeutic effect on correcting abnormal blood vessels in ischemic hindlimbs and tumors. CONCLUSION Our study innovatively revealed a reliable mouse model wherein the Ang2/Tie2 signaling cascade disrupted the endothelial homeostasis to aggravate the progression of hindlimb ischemia and tumor angiogenesis. This balance can be rescued by the combination therapy of Tan I and Sal A that were both from SME, leading to the occurrence of vascular normalization.
Collapse
Affiliation(s)
- Cheng Qian
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Ying Huang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Shan Zhang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Chunmei Yang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Weiwei Zheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Weiwei Tang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Guiping Wan
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, 210028, PR China
| | - Aiyun Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| | - Yang Zhao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China; Department of Biochemistry and Molecular Biology, School of Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China.
| |
Collapse
|
2
|
Herrera JL, Komatsu M. Akt3 activation by R-Ras in an endothelial cell enforces quiescence and barrier stability of neighboring endothelial cells via Jagged1. Cell Rep 2024; 43:113837. [PMID: 38402584 PMCID: PMC11056028 DOI: 10.1016/j.celrep.2024.113837] [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: 07/06/2023] [Revised: 12/06/2023] [Accepted: 02/06/2024] [Indexed: 02/27/2024] Open
Abstract
Communication between adjacent endothelial cells is important for the homeostasis of blood vessels. We show that quiescent endothelial cells use Jagged1 to instruct neighboring endothelial cells to assume a quiescent phenotype and secure the endothelial barrier. This phenotype enforcement by neighboring cells is operated by R-Ras through activation of Akt3, which results in upregulation of a Notch ligand Jagged1 and consequential upregulation of Notch target genes, such as UNC5B, and VE-cadherin accumulation in the neighboring cells. These signaling events lead to the stable interaction between neighboring endothelial cells to continue to fortify juxtacrine signaling via Jagged1-Notch. This mode of intercellular signaling provides a positive feedback regulation of endothelial cell-cell interactions and cellular quiescence required for the stabilization of the endothelium.
Collapse
Affiliation(s)
- Jose Luis Herrera
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701, USA; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Masanobu Komatsu
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, St. Petersburg, FL 33701, USA; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA.
| |
Collapse
|
3
|
Bello Baez A, Nieto Morales ML, Mora Guanche P, Cavada Laza A, Pérez Méndez LI. Can Achilles tendinosis be treated effectively with lidocaine and glucose infiltrations, and if so, is the effect lasting? A longitudinal, observational on 27 consecutive patients. RADIOLOGIA 2023; 65 Suppl 2:S41-S49. [PMID: 37858352 DOI: 10.1016/j.rxeng.2022.05.002] [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: 01/30/2022] [Accepted: 05/09/2022] [Indexed: 10/21/2023]
Abstract
OBJECTIVE Our aim was to add to the small but growing body of evidence on the effectiveness of ultrasound-guided Achilles intratendinous hyperosmolar dextrose prolotherapy and introduce a novel, preceding step of paratenon hydrodissection with lidocaine in patients with chronic Achilles tendinosis resistant to rehabilitation therapy. METHODS We conducted a longitudinal, observational study on 27 consecutive patients diagnosed with Achilles tendinosis, in whom conservative treatment, ie, physiotherapy or shock wave therapy, had failed. A 2% lidocaine paratenon anesthesia and hydrodissection was followed by ultrasound-guided, intratendinous injections of 25% glucose every 5 weeks. Visual analogue scales (VAS) were used for pain assessment at rest, for activities of daily living, and after moderate exercise at the begining and at the end of the treatment. Moreover, tendon thickness and vascularisation were recorded at baseline and final treatment consultation. Effectiveness was estimated from scoring and relative pain reduction using a 95% CI. The non-parametric Wilcoxon test and a general linear model for repeated measures were applied. Statistical significance was established as p < 0.05. RESULTS A median of 5 (1-11) injection consultations per patient were required. Pain scores decreased significantly in all three conditions (p < 0.001). Relative reductions were 75% in pain at rest (95% CI;61-93%), 69% in pain with daily living activities (95% CI; 55-83%), and 70% in pain after moderate exercise (95% CI; 57-84%). Tendon neo-vascularisation was significantly reduced (p < 0.001). We did not observe significant changes in tendon thickness (p = 0.083). CONCLUSIONS Achilles tendinosis treatment with paratenon lidocaine hydrodissection and subsequent prolotherapy with hyperosmolar glucose solution is safe, effective, inexpensive, and virtually painless with results maintained over time.
Collapse
Affiliation(s)
- A Bello Baez
- Servicio de Radiología Osteomuscular, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain.
| | - M L Nieto Morales
- Servicio de Radiología Osteomuscular, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - P Mora Guanche
- Servicio de Radiología Osteomuscular, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - A Cavada Laza
- Servicio de Radiología Osteomuscular, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| | - Lina Inmaculada Pérez Méndez
- Departamento de Epidemiología Clínica y Bioestadística, Unidad de Investigación, Hospital Universitario Nuestra Señora de Candelaria, Santa Cruz de Tenerife, Spain
| |
Collapse
|
4
|
Borek I, Birnhuber A, Voelkel NF, Marsh LM, Kwapiszewska G. The vascular perspective on acute and chronic lung disease. J Clin Invest 2023; 133:e170502. [PMID: 37581311 PMCID: PMC10425217 DOI: 10.1172/jci170502] [Citation(s) in RCA: 35] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/16/2023] Open
Abstract
The pulmonary vasculature has been frequently overlooked in acute and chronic lung diseases, such as acute respiratory distress syndrome (ARDS), pulmonary fibrosis (PF), and chronic obstructive pulmonary disease (COPD). The primary emphasis in the management of these parenchymal disorders has largely revolved around the injury and aberrant repair of epithelial cells. However, there is increasing evidence that the vascular endothelium plays an active role in the development of acute and chronic lung diseases. The endothelial cell network in the capillary bed and the arterial and venous vessels provides a metabolically highly active barrier that controls the migration of immune cells, regulates vascular tone and permeability, and participates in the remodeling processes. Phenotypically and functionally altered endothelial cells, and remodeled vessels, can be found in acute and chronic lung diseases, although to different degrees, likely because of disease-specific mechanisms. Since vascular remodeling is associated with pulmonary hypertension, which worsens patient outcomes and survival, it is crucial to understand the underlying vascular alterations. In this Review, we describe the current knowledge regarding the role of the pulmonary vasculature in the development and progression of ARDS, PF, and COPD; we also outline future research directions with the hope of facilitating the development of mechanism-based therapies.
Collapse
Affiliation(s)
- Izabela Borek
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
| | - Anna Birnhuber
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Norbert F. Voelkel
- Pulmonary Medicine Department, University of Amsterdam Medical Centers, Amsterdam, Netherlands
- Department of Pulmonary Medicine, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centers, Amsterdam, Netherlands
| | - Leigh M. Marsh
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
| | - Grazyna Kwapiszewska
- Ludwig Boltzmann Institute for Lung Vascular Research, Graz, Austria
- Otto Loewi Research Center, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Institute for Lung Health, German Lung Center (DZL), Cardiopulmonary Institute, Giessen, Germany
| |
Collapse
|
5
|
Bian F, Lan YW, Zhao S, Deng Z, Shukla S, Acharya A, Donovan J, Le T, Milewski D, Bacchetta M, Hozain AE, Tipograf Y, Chen YW, Xu Y, Shi D, Kalinichenko VV, Kalin TV. Lung endothelial cells regulate pulmonary fibrosis through FOXF1/R-Ras signaling. Nat Commun 2023; 14:2560. [PMID: 37137915 PMCID: PMC10156846 DOI: 10.1038/s41467-023-38177-2] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 04/18/2023] [Indexed: 05/05/2023] Open
Abstract
Pulmonary fibrosis results from dysregulated lung repair and involves multiple cell types. The role of endothelial cells (EC) in lung fibrosis is poorly understood. Using single cell RNA-sequencing we identified endothelial transcription factors involved in lung fibrogenesis, including FOXF1, SMAD6, ETV6 and LEF1. Focusing on FOXF1, we found that FOXF1 is decreased in EC within human idiopathic pulmonary fibrosis (IPF) and mouse bleomycin-injured lungs. Endothelial-specific Foxf1 inhibition in mice increased collagen depositions, promoted lung inflammation, and impaired R-Ras signaling. In vitro, FOXF1-deficient EC increased proliferation, invasion and activation of human lung fibroblasts, and stimulated macrophage migration by secreting IL-6, TNFα, CCL2 and CXCL1. FOXF1 inhibited TNFα and CCL2 through direct transcriptional activation of Rras gene promoter. Transgenic overexpression or endothelial-specific nanoparticle delivery of Foxf1 cDNA decreased pulmonary fibrosis in bleomycin-injured mice. Nanoparticle delivery of FOXF1 cDNA can be considered for future therapies in IPF.
Collapse
Affiliation(s)
- Fenghua Bian
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Ying-Wei Lan
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Shuyang Zhao
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Zicheng Deng
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, USA
| | - Samriddhi Shukla
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Anusha Acharya
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Johnny Donovan
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Tien Le
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - David Milewski
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
| | - Matthew Bacchetta
- Departments of Thoracic and Cardiac Surgery, Department of Biomedical Engineering, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ahmed Emad Hozain
- Department of Surgery, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Yuliya Tipograf
- Department of Surgery, State University of New York Downstate Medical Center, Brooklyn, NY, USA
| | - Ya-Wen Chen
- Department of Cell, Developmental, and Regenerative Biology, Department of Otolaryngology, Institute for Airway Sciences, Black Family Stem Cell Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Yan Xu
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Donglu Shi
- The Materials Science and Engineering Program, College of Engineering and Applied Science, University of Cincinnati, Cincinnati, OH, USA
| | - Vladimir V Kalinichenko
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA
- Center for Lung Regenerative Medicine, Perinatal Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, the Perinatal Institute of Cincinnati Children's Research Foundation, Cincinnati, OH, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA.
| |
Collapse
|
6
|
Vähätupa M, Nättinen J, Aapola U, Uusitalo-Järvinen H, Uusitalo H, Järvinen TAH. Proteomics Analysis of R-Ras Deficiency in Oxygen Induced Retinopathy. Int J Mol Sci 2023; 24:ijms24097914. [PMID: 37175621 PMCID: PMC10178533 DOI: 10.3390/ijms24097914] [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: 03/12/2023] [Revised: 04/21/2023] [Accepted: 04/24/2023] [Indexed: 05/15/2023] Open
Abstract
Small GTPase R-Ras regulates vascular permeability in angiogenesis. In the eye, abnormal angiogenesis and hyperpermeability are the leading causes of vision loss in several ischemic retinal diseases such as proliferative diabetic retinopathy (PDR), retinal vein occlusion (RVO), and retinopathy of prematurity (ROP). Oxygen-induced retinopathy (OIR) is the most widely used experimental model for these ischemic retinopathies. To shed more light on how the R-Ras regulates vascular permeability in pathological angiogenesis, we performed a comprehensive (>2900 proteins) characterization of OIR in R-Ras knockout (KO) and wild-type (WT) mice by sequential window acquisition of all theoretical mass spectra (SWATH-MS) proteomics. OIR and age-matched normoxic control retinas were collected at P13, P17, and P42 from R-Ras KO and WT mice and were subjected to SWATH-MS and data analysis. The most significant difference between the R-Ras KO and WT retinas was an accumulation of plasma proteins. The pathological vascular hyperpermeability during OIR in the R-Ras KO retina took place very early, P13. This led to simultaneous hypoxic cell injury/death (ferroptosis), glycolytic metabolism as well compensatory mechanisms to counter the pathological leakage from angiogenic blood vessels in the OIR retina of R-Ras deficient mice.
Collapse
Affiliation(s)
- Maria Vähätupa
- Faculty of Medicine and Health Technology, Tampere University, 33521 Tampere, Finland
| | - Janika Nättinen
- Faculty of Medicine and Health Technology, Tampere University, 33521 Tampere, Finland
- Tampere University Hospital, 33520 Tampere, Finland
| | - Ulla Aapola
- Faculty of Medicine and Health Technology, Tampere University, 33521 Tampere, Finland
- Tampere University Hospital, 33520 Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine and Health Technology, Tampere University, 33521 Tampere, Finland
- Tampere University Hospital, 33520 Tampere, Finland
| | - Hannu Uusitalo
- Faculty of Medicine and Health Technology, Tampere University, 33521 Tampere, Finland
- Tampere University Hospital, 33520 Tampere, Finland
| | - Tero A H Järvinen
- Faculty of Medicine and Health Technology, Tampere University, 33521 Tampere, Finland
- Tampere University Hospital, 33520 Tampere, Finland
| |
Collapse
|
7
|
Barbieri F, Hall IF, Elia L, Civilini E. Vascular malformation rupture in a patient affected by Costello syndrome. BMJ Case Rep 2022; 15:e250948. [PMID: 36526283 PMCID: PMC9764614 DOI: 10.1136/bcr-2022-250948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2022] [Indexed: 12/23/2022] Open
Abstract
Costello syndrome (CS) is a rare genetic syndrome affecting multiple organs, generally caused by mutations of the HRAS gene, belonging to the RAS/MAPK genes family.A male patient with CS developed a painful pulsatile mass on the lateral side of the wrist. An initial ultrasonographic investigation confirmed the presence of a radial artery lesion, possibly an arterial aneurysm. On surgical resection, histological evaluation showed a tangle of vascular structures with variable calibre and abnormal wall histology. Immunohistochemical stainings revealed a very poor endothelial contribution to the central vascular wall structure. These histological observations led us to conclude we had managed an acute vascular malformation (VM) rupture, rather than a common arterial aneurysmal condition. Considering the molecular mechanisms regulated by RAS/MAPK genes, CS patients might have a higher risk of developing VMs and, in the presence of a pulsatile mass with acute onset, VM rupture should be considered.
Collapse
Affiliation(s)
| | | | - Leonardo Elia
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
- DMMT, Università degli Studi di Brescia, Brescia, Italy
| | - Efrem Civilini
- Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
- Humanitas University, Rozzano, Milan, Italy
| |
Collapse
|
8
|
Bello Baez A, Nieto Morales M, Mora Guanche P, Cavada Laza A, Pérez Méndez LI. ¿Puede tratarse la tendinosis aquílea de forma eficaz con lidocaína e infiltraciones de glucosa y, de ser así, es duradero su efecto? Estudio observacional longitudinal en 27 pacientes consecutivos. RADIOLOGIA 2022. [DOI: 10.1016/j.rx.2022.05.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
9
|
Liu J, Dong N, Li N, Zhao H, Li Y, Mao H, Ren H, Feng Y, Liu J, Du L, Mao H. IL-35 enhances angiogenic effects of small extracellular vesicles in breast cancer. FEBS J 2022; 289:3489-3504. [PMID: 35037402 DOI: 10.1111/febs.16359] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 12/15/2021] [Accepted: 01/14/2022] [Indexed: 12/24/2022]
Abstract
As an indispensable process for breast cancer metastasis, tumour angiogenesis requires a tight interaction between cancer cells and endothelial cells in tumour microenvironment. Here, we explored the participation of small extracellular vesicles (sEVs) derived from breast cancer cells in modulating angiogenesis and investigated the effect of IL-35 in facilitating this process. Firstly, we characterized breast cancer cells-derived sEVs untreated or treated with IL-35 and visualized the internalization of these sEVs by human umbilical vein endothelial cells (HUVECs). Breast cancer cells-derived sEVs promoted endothelial cell proliferation through facilitating cell cycle progression and enhanced capillary-like structures formation and microvessel formation. Subsequent results proved that IL-35 further reinforced the angiogenic effect induced by breast cancer cells-derived sEVs. Moreover, sEVs from breast cancer cells significantly enhanced tumour growth and microvessel density in breast tumour-bearing mice model. Microarray analysis showed that IL-35 might alter the mRNA profiles of sEVs and activate the Ras/Raf/MEK/ERK signalling pathway. These findings demonstrated that IL-35 indirectly promoted angiogenesis in breast cancer through regulating the content of breast cancer cells-derived sEVs, which could be internalized by HUVECs.
Collapse
Affiliation(s)
- Jia Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Nana Dong
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ning Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hui Zhao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yali Li
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Huihui Mao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hanxiao Ren
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yimin Feng
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jie Liu
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lutao Du
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Haiting Mao
- Department of Clinical Laboratory, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
10
|
Feng ZY, Huang TL, Li XR, Chen L, Deng S, Xu SR, Ma KT, Li L, Si JQ. 17β-Estradiol promotes angiogenesis of stria vascular in cochlea of C57BL/6J mice. Eur J Pharmacol 2021; 913:174642. [PMID: 34822791 DOI: 10.1016/j.ejphar.2021.174642] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/12/2021] [Accepted: 11/12/2021] [Indexed: 02/08/2023]
Abstract
It is widely accepted that the stria vascularis (SV) in cochlea plays a critical role in the generation of endocochlear potential (EP) and the secretion of the endolymph. 17β-estradiol (E2) is the most potent and abundant endogenous estrogen during the premenopausal period, thus, considered as the reference estrogen. This study aimd to investigate the protective effect of E2 by promoting the expression of vascular endothelial growth factor (VEGF) and thus promoting the vascular regeneration of the SV in elderly mice. After being treated with E2 either in vivo or in vitro, the hearing threshold changes of C57BL/6J elder mice continuously reduced, endothelial cell morphology improved, the number of endothelial cells (ECs) tubular nodes increased significantly, the ability of tubular formation enhanced significantly and the expression of VEGF increased. In vitro, cell model in conjunction with in vivo ovariectomized model was established to demonstrate for the first time that E2 promotes angiogenesis by promoting the secretion of VEGF through the phosphatidylinositol 3-kinase (PI3K)/AKT pathway (PI3K/AKT). In conclusion, E2 demonstrated potent angiogenesis properties with significant protection against Age-Related Hearing Loss (ARHL), which provides a new idea for the improvement of ARHL.
Collapse
Affiliation(s)
- Zi-Yi Feng
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, China; Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China; Maternal and Child Health Care Hospital, Urumqi, Xinjiang, 830000, China
| | - Tian-Lan Huang
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, China; Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Xue-Rui Li
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Long Chen
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Shuang Deng
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Shao-Ran Xu
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Ke-Tao Ma
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China
| | - Li Li
- Department of Physiology, Medical College of Jiaxing University, Jiaxing, Zhejiang, 314000, China.
| | - Jun-Qiang Si
- Department of Physiology, Medical College of Shihezi University, Shihezi, Xinjiang, 832000, China; Department of Neurobiology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430000, China.
| |
Collapse
|
11
|
Targeting the Ang2/Tie2 Axis with Tanshinone IIA Elicits Vascular Normalization in Ischemic Injury and Colon Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:7037786. [PMID: 34804370 PMCID: PMC8598375 DOI: 10.1155/2021/7037786] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/04/2021] [Accepted: 10/15/2021] [Indexed: 02/06/2023]
Abstract
Pathological angiogenesis, as exhibited by aberrant vascular structure and function, has been well deemed to be a hallmark of cancer and various ischemic diseases. Therefore, strategies to normalize vasculature are of potential therapeutic interest in these diseases. Recently, identifying bioactive compounds from medicinal plant extracts to reverse abnormal vasculature has been gaining increasing attention. Tanshinone IIA (Tan IIA), an active component of Salvia miltiorrhiza, has been shown to play significant roles in improving blood circulation and delaying tumor progression. However, the underlying mechanisms responsible for the therapeutic effects of Tan IIA are not fully understood. Herein, we established animal models of HT-29 human colon cancer xenograft and hind limb ischemia to investigate the role of Tan IIA in regulating abnormal vasculature. Interestingly, our results demonstrated that Tan IIA could significantly promote the blood flow, alleviate the hypoxia, improve the muscle quality, and ameliorate the pathological damage after ischemic insult. Meanwhile, we also revealed that Tan IIA promoted the integrity of vascular structure, reduced vascular leakage, and attenuated the hypoxia in HT-29 tumors. Moreover, the circulating angiopoietin 2 (Ang2), which is extremely high in these two pathological states, was substantially depleted in the presence of Tan IIA. Also, the activation of Tie2 was potentiated by Tan IIA, resulting in decreased vascular permeability and elevated vascular integrity. Mechanistically, we uncovered that Tan IIA maintained vascular stability by targeting the Ang2-Tie2-AKT-MLCK cascade. Collectively, our data suggest that Tan IIA normalizes vessels in tumors and ischemic injury via regulating the Ang2/Tie2 signaling pathway.
Collapse
|
12
|
Vähätupa M, Salonen N, Uusitalo-Järvinen H, Järvinen TAH. Selective Targeting and Tissue Penetration to the Retina by a Systemically Administered Vascular Homing Peptide in Oxygen Induced Retinopathy (OIR). Pharmaceutics 2021; 13:pharmaceutics13111932. [PMID: 34834347 PMCID: PMC8618640 DOI: 10.3390/pharmaceutics13111932] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/08/2021] [Accepted: 11/10/2021] [Indexed: 12/17/2022] Open
Abstract
Pathological angiogenesis is the hallmark of ischemic retinal diseases among them retinopathy of prematurity (ROP) and proliferative diabetic retinopathy (PDR). Oxygen-induced retinopathy (OIR) is a pure hypoxia-driven angiogenesis model and a widely used model for ischemic retinopathies. We explored whether the vascular homing peptide CAR (CARSKNKDC) which recognizes angiogenic blood vessels can be used to target the retina in OIR. We were able to demonstrate that the systemically administered CAR vascular homing peptide homed selectively to the preretinal neovessels in OIR. As a cell and tissue-penetrating peptide, CAR also penetrated into the retina. Hyperoxia used to induce OIR in the retina also causes bronchopulmonary dysplasia in the lungs. We showed that the CAR peptide is not targeted to the lungs in normal mice but is targeted to the lungs after hyperoxia-/hypoxia-treatment of the animals. The site-specific delivery of the CAR peptide to the pathologic retinal vasculature and the penetration of the retinal tissue may offer new opportunities for treating retinopathies more selectively and with less side effects.
Collapse
Affiliation(s)
- Maria Vähätupa
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (M.V.); (N.S.); (H.U.-J.)
| | - Niklas Salonen
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (M.V.); (N.S.); (H.U.-J.)
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (M.V.); (N.S.); (H.U.-J.)
- Eye Centre & Department of Orthopedics & Traumatology, Tampere University Hospital, 33520 Tampere, Finland
| | - Tero A. H. Järvinen
- Faculty of Medicine and Health Technology, Tampere University, 33520 Tampere, Finland; (M.V.); (N.S.); (H.U.-J.)
- Eye Centre & Department of Orthopedics & Traumatology, Tampere University Hospital, 33520 Tampere, Finland
- Correspondence:
| |
Collapse
|
13
|
Takino JI, Miyazaki S, Nagamine K, Hori T. The Role of RASGRP2 in Vascular Endothelial Cells-A Mini Review. Int J Mol Sci 2021; 22:ijms222011129. [PMID: 34681791 PMCID: PMC8537898 DOI: 10.3390/ijms222011129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/05/2021] [Accepted: 10/13/2021] [Indexed: 11/16/2022] Open
Abstract
RAS guanyl nucleotide-releasing proteins (RASGRPs) are important proteins that act as guanine nucleotide exchange factors, which activate small GTPases and function as molecular switches for intracellular signals. The RASGRP family is composed of RASGRP1-4 proteins and activates the small GTPases, RAS and RAP. Among them, RASGRP2 has different characteristics from other RASGRPs in that it targets small GTPases and its localizations are different. Many studies related to RASGRP2 have been reported in cells of the blood cell lineage. Furthermore, RASGRP2 has also been reported to be associated with Huntington's disease, tumors, and rheumatoid arthritis. In addition, we also recently reported RASGRP2 expression in vascular endothelial cells, and clarified the involvement of xenopus Rasgrp2 in the vasculogenesis process and multiple signaling pathways of RASGRP2 in human vascular endothelial cells with stable expression of RASGRP2. Therefore, this article outlines the existing knowledge of RASGRP2 and focuses on its expression and role in vascular endothelial cells, and suggests that RASGRP2 functions as a protective factor for maintaining healthy blood vessels.
Collapse
Affiliation(s)
- Jun-ichi Takino
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
- Correspondence: ; Tel.: +81-823-73-8584
| | - Shouhei Miyazaki
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
| | - Kentaro Nagamine
- Faculty of Health Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan;
| | - Takamitsu Hori
- Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure, Hiroshima 737-0112, Japan; (S.M.); (T.H.)
| |
Collapse
|
14
|
Weber SM, Carroll SL. The Role of R-Ras Proteins in Normal and Pathologic Migration and Morphologic Change. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1499-1510. [PMID: 34111428 PMCID: PMC8420862 DOI: 10.1016/j.ajpath.2021.05.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 05/11/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022]
Abstract
The contributions that the R-Ras subfamily [R-Ras, R-Ras2/teratocarcinoma 21 (TC21), and M-Ras] of small GTP-binding proteins make to normal and aberrant cellular functions have historically been poorly understood. However, this has begun to change with the realization that all three R-Ras subfamily members are occasionally mutated in Noonan syndrome (NS), a RASopathy characterized by the development of hematopoietic neoplasms and abnormalities affecting the immune, cardiovascular, and nervous systems. Consistent with the abnormalities seen in NS, a host of new studies have implicated R-Ras proteins in physiological and pathologic changes in cellular morphology, adhesion, and migration in the cardiovascular, immune, and nervous systems. These changes include regulating the migration and homing of mature and immature immune cells, vascular stabilization, clotting, and axonal and dendritic outgrowth during nervous system development. Dysregulated R-Ras signaling has also been linked to the pathogenesis of cardiovascular disease, intellectual disabilities, and human cancers. This review discusses the structure and regulation of R-Ras proteins and our current understanding of the signaling pathways that they regulate. It explores the phenotype of NS patients and their implications for the R-Ras subfamily functions. Next, it covers recent discoveries regarding physiological and pathologic R-Ras functions in key organ systems. Finally, it discusses how R-Ras signaling is dysregulated in cancers and mechanisms by which this may promote neoplasia.
Collapse
Affiliation(s)
- Shannon M Weber
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina
| | - Steven L Carroll
- Department of Pathology and Laboratory Medicine, Medical University of South Carolina, Charleston, South Carolina.
| |
Collapse
|
15
|
Cuesta C, Arévalo-Alameda C, Castellano E. The Importance of Being PI3K in the RAS Signaling Network. Genes (Basel) 2021; 12:1094. [PMID: 34356110 PMCID: PMC8303222 DOI: 10.3390/genes12071094] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 07/06/2021] [Accepted: 07/16/2021] [Indexed: 12/12/2022] Open
Abstract
Ras proteins are essential mediators of a multitude of cellular processes, and its deregulation is frequently associated with cancer appearance, progression, and metastasis. Ras-driven cancers are usually aggressive and difficult to treat. Although the recent Food and Drug Administration (FDA) approval of the first Ras G12C inhibitor is an important milestone, only a small percentage of patients will benefit from it. A better understanding of the context in which Ras operates in different tumor types and the outcomes mediated by each effector pathway may help to identify additional strategies and targets to treat Ras-driven tumors. Evidence emerging in recent years suggests that both oncogenic Ras signaling in tumor cells and non-oncogenic Ras signaling in stromal cells play an essential role in cancer. PI3K is one of the main Ras effectors, regulating important cellular processes such as cell viability or resistance to therapy or angiogenesis upon oncogenic Ras activation. In this review, we will summarize recent advances in the understanding of Ras-dependent activation of PI3K both in physiological conditions and cancer, with a focus on how this signaling pathway contributes to the formation of a tumor stroma that promotes tumor cell proliferation, migration, and spread.
Collapse
Affiliation(s)
| | | | - Esther Castellano
- Tumour-Stroma Signalling Laboratory, Centro de Investigación del Cáncer, Instituto de Biología Molecular y Celular del Cáncer, Consejo Superior de Investigaciones Científicas (CSIC)-Universidad de Salamanca, Campus Miguel de Unamuno, 37007 Salamanca, Spain; (C.C.); (C.A.-A.)
| |
Collapse
|
16
|
Zheng R, Li F, Li F, Gong A. Targeting tumor vascularization: promising strategies for vascular normalization. J Cancer Res Clin Oncol 2021; 147:2489-2505. [PMID: 34148156 DOI: 10.1007/s00432-021-03701-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 06/14/2021] [Indexed: 12/17/2022]
Abstract
Tumor recurrence after the clinical cure of tumor often results from the presence of an abnormal microenvironment, including an aberrant vasculature. The tumor microenvironment is rich in pro-angiogenic factors but lacks pro-maturation factors. Pro-angiogenic conditions in the tumor microenvironment, such as hypoxia, are double-edged swords, promoting both the repair of normal tissues and the development of an abnormal blood vessel network. The coexistence of perfusion and hypoxic zones and uneven blood vessel distribution in tumor tissues profoundly influence tumor deterioration, recurrence, and metastasis. Traditional anti-angiogenic therapies have shown limited efficacy, and promote drug resistance, and even metastasis. In contrast, vascular normalization therapy induces a more physiological-like state, leading to better outcomes and fewer side effects. Vascular normalization entails modifying the tumor vascular system to improve tumor oxygenation and substance transport, thereby contributing to improving the efficacy of radiotherapy, chemotherapy, and immunotherapy. This review mainly focuses on the process of tumor vascularization; potential therapeutic targets, including cells, metabolism, signaling pathways, and angiogenesis-related genes; and possible strategies to normalize blood vessels through regulating tumor vessel generation, the development of tumor vessels, and blood vessel fusion and pruning.
Collapse
Affiliation(s)
- Ruiqi Zheng
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Feifan Li
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Fengcen Li
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China
| | - Aihua Gong
- Department of Cell Biology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212003, Jiangsu, China.
| |
Collapse
|
17
|
Ñaupas LVS, Brito DCC, de Souza SS, Brandão FAS, da Silva RF, da Silva Raposo R, de Oliveira Monteiro Moreira AC, Araújo AA, Alves BG, Guedes MIF, Silva JYG, Cordova A, Figueiredo JR, Rodrigues APR. Alpha Lipoic Acid Supplementation Improves Ovarian Tissue Vitrification Outcome: An Alternative to Preserve the Ovarian Function of Morada Nova Ewe. Reprod Sci 2021; 28:3109-3122. [PMID: 34008154 DOI: 10.1007/s43032-021-00593-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 04/20/2021] [Indexed: 10/21/2022]
Abstract
This study evaluated the effect of adding alpha lipoic acid (ALA) to the vitrification solution of sheep ovarian tissue on 7 days of in vitro culture or 15 days of xenotransplantion. ALA was used at two different concentrations (100 μM: ALA100 and 150 μM: ALA150). Ovarian tissue was evaluated by classical histology (follicular morphology, development, and stromal cell density); immunohistochemistry for forkhead box O3a (FOXO3a); Ki67 (cell proliferation); cluster of differentiation 31 (CD31); and alpha smooth muscle actin (α-SMA). Reactive oxygen species (ROS) levels in ovarian tissue, as well as malondialdehyde (MDA) and nitrite levels in the culture medium, were assessed. Similar percentage of morphologically normal follicles was found in the vitrified ovarian tissue in the presence of ALA100 or ALA150 after in vitro culture or xenotransplantation. Follicular development from all treatments was higher (P < 0.05) than the control group. Moreover, an activation of primordial follicles was observed by FOXO3a. Stromal cell density and immunostaining for Ki67 and CD31 were significantly higher (P < 0.05) in ALA150 vitrified tissue. No difference (P > 0.05) was found in α-SMA between ALA concentrations after in vitro culture or xenograft. ROS levels in the ovarian tissue were similar (P > 0.05) in all treatments, as well as MDA and nitrite levels after 7 days of culture. We concluded that the addition of ALA 150 is able to better preserve the stromal cell density favoring granulosa cell proliferation and neovascularization.
Collapse
Affiliation(s)
- Lucy Vanessa Sulca Ñaupas
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará (UECE), Fortaleza, CE, Brazil
| | - Danielle Cristina Calado Brito
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará (UECE), Fortaleza, CE, Brazil
| | - Samara Silva de Souza
- Laboratory of Image Diagnosis Applied to Animal Reproduction, Faculty of Veterinary Medicine, UECE, Fortaleza, CE, Brazil
| | | | - Renato Félix da Silva
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará (UECE), Fortaleza, CE, Brazil
| | | | | | | | - Benner Geraldo Alves
- Animal Bioscience Postgraduate Program, Federal University of Goiás, Jataí, GO, Brazil
| | | | | | - Amanda Cordova
- Department of Biomedical Sciences, University of Guelph, Guelph, ON, Canada
| | - José Ricardo Figueiredo
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará (UECE), Fortaleza, CE, Brazil
| | - Ana Paula Ribeiro Rodrigues
- Laboratory of Manipulation of Oocytes and Ovarian Pre-Antral Follicles, Faculty of Veterinary Medicine, State University of Ceará (UECE), Fortaleza, CE, Brazil.
| |
Collapse
|
18
|
Herrera JL, Komatsu M. R-Ras Deficiency in Pericytes Causes Frequent Microphthalmia and Perturbs Retinal Vascular Development. J Vasc Res 2021; 58:252-266. [PMID: 33873190 PMCID: PMC8263468 DOI: 10.1159/000514555] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
PURPOSE The retinal vasculature is heavily invested by pericytes. Small GTPase R-Ras is highly expressed in endothelial cells and pericytes, suggesting importance of this Ras homolog for the regulation of the blood vessel wall. We investigated the specific contribution of pericyte-expressed R-Ras to the development of the retinal vasculature. METHODS The effect of R-Ras deficiency in pericytes was analyzed in pericyte-targeted conditional Rras knockout mice at birth and during the capillary plexus formation in the neonatal retina. RESULTS The offspring of these mice frequently exhibited unilateral microphthalmia. Analyses of the developing retinal vasculature in the eyes without microphthalmia revealed excessive endothelial cell proliferation, sprouting, and branching of the capillary plexus in these animals. These vessels were structurally defective with diminished pericyte coverage and basement membrane formation. Furthermore, these vessels showed reduced VE-cadherin staining and significantly elevated plasma leakage indicating the breakdown of the blood-retinal barrier. This defect was associated with considerable macrophage infiltration in the retina. CONCLUSIONS The normal retinal vascular development is dependent on R-Ras expression in pericytes, and the absence of it leads to unattenuated angiogenesis and significantly weakens the blood-retinal barrier. Our findings underscore the importance of R-Ras for pericyte function during the normal eye development.
Collapse
Affiliation(s)
- Jose Luis Herrera
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, and Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| | - Masanobu Komatsu
- Cancer and Blood Disorders Institute, Institute for Fundamental Biomedical Research, and Department of Surgery, Johns Hopkins All Children's Hospital, and Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida, USA
| |
Collapse
|
19
|
Sawada J, Perrot CY, Chen L, Fournier-Goss AE, Oyer J, Copik A, Komatsu M. High Endothelial Venules Accelerate Naive T Cell Recruitment by Tumor Necrosis Factor-Mediated R-Ras Upregulation. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:396-414. [PMID: 33159887 DOI: 10.1016/j.ajpath.2020.10.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 10/08/2020] [Accepted: 10/14/2020] [Indexed: 01/01/2023]
Abstract
Recruitment of naive T cells to lymph nodes is essential for the development of adaptive immunity. Upon pathogen infection, lymph nodes promptly increase the influx of naive T cells from the circulation in order to screen and prime the T cells. The precise contribution of the lymph node vasculature to the regulation of this process remains unclear. Here we show a role for the Ras GTPase, R-Ras, in the functional adaptation of high endothelial venules to increase naive T cell trafficking to the lymph nodes. R-Ras is transiently up-regulated in the endothelium of high endothelial venules by the inflammatory cytokine tumor necrosis factor (TNF) within 24 hours of pathogen inoculation. TNF induces R-Ras upregulation in endothelial cells via JNK and p38 mitogen-activated protein kinase but not NF-κB. Studies of T cell trafficking found that the loss of function of endothelial R-Ras impairs the rapid acceleration of naive T cell recruitment to the lymph nodes upon inflammation. This defect diminished the ability of naive OT-1 T cells to develop antitumor activity against ovalbumin-expressing melanoma. Proteomic analyses suggest that endothelial R-Ras facilitates TNF-dependent transendothelial migration (diapedesis) of naive T cells by modulating molecular assembly the at T cell-endothelial cell interface. These findings give new mechanistic insights into the functional adaptation of high endothelial venules to accelerate naive T cell recruitment to the lymph nodes.
Collapse
Affiliation(s)
- Junko Sawada
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Carole Y Perrot
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Linyuan Chen
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Ashley E Fournier-Goss
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida
| | - Jeremiah Oyer
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, St. Petersburg, Florida; Burnett School of Biomedical Sciences, University of Central Florida, Orlando Florida
| | - Alicja Copik
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando Florida
| | - Masanobu Komatsu
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, Florida; Tumor Microenvironment and Cancer Immunology Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla California.
| |
Collapse
|
20
|
Microtubule Stabilization Promotes Microcirculation Reconstruction After Spinal Cord Injury. J Mol Neurosci 2020; 71:583-595. [PMID: 32901373 PMCID: PMC7851021 DOI: 10.1007/s12031-020-01679-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Accepted: 08/05/2020] [Indexed: 12/23/2022]
Abstract
Spinal cord microcirculation plays an important role in maintaining the function of spinal cord neurons and other cells. Previous studies have largely focused on the ability of microtubule stabilization to inhibit the fibroblast migration and promote axon regeneration after spinal cord injury (SCI). However, the effect of microtubule stabilization treatment on microcirculation reconstruction after SCI remains unclear. By using immunofluorescence, we found that microtubule stabilization treatment improved microcirculation reconstruction via increasing the number of microvessels, pericytes, and the perfused microvessels after SCI. To clarify the underlying mechanisms, rat brain microvascular endothelial cells and pericytes were subjected to glucose oxygen deprivation. By using flow cytometry and western blotting, we found that microtubule stabilization treatment inhibited apoptosis and migration of endothelial cells and pericytes but promoted proliferation and survival of endothelial cells and pericytes through upregulated expression of vascular endothelial growth factor A (VEGFA), VEGF receptor 2, platelet-derived growth factor-B (PDGFB), PDGF receptor β, and angiopoietin-1 after SCI. Taken together, this study provides evidence for the mechanisms underlying the promotion of microcirculation reconstruction after SCI by microtubule stabilization treatment. Importantly, this study suggests the potential of microtubule stabilization as a therapeutic target to reduce microcirculation dysfunction after SCI in the clinic.
Collapse
|
21
|
Perrot CY, Herrera JL, Fournier-Goss AE, Komatsu M. Prostaglandin E2 breaks down pericyte-endothelial cell interaction via EP1 and EP4-dependent downregulation of pericyte N-cadherin, connexin-43, and R-Ras. Sci Rep 2020; 10:11186. [PMID: 32636414 PMCID: PMC7341885 DOI: 10.1038/s41598-020-68019-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 06/05/2020] [Indexed: 12/13/2022] Open
Abstract
A close association between pericytes and endothelial cells (ECs) is crucial to the stability and function of capillary blood vessels and microvessels. The loss or dysfunction of pericytes results in significant disruption of these blood vessels as observed in pathological conditions, including cancer, diabetes, stroke, and Alzheimer’s disease. Prostaglandin E2 (PGE2) is a lipid mediator of inflammation, and its tissue concentration is elevated in cancer and neurological disorders. Here, we show that the exposure to PGE2 switches pericytes to a fast-migrating, loosely adhered phenotype that fails to intimately interact with ECs. N-cadherin and connexin-43 in adherens junction and gap junction between pericytes and ECs are downregulated by EP-4 and EP-1-dependent mechanisms, leading to breakdown of the pericyte–EC interaction. Furthermore, R-Ras, a small GTPase important for vascular normalization and vessel stability, is transcriptionally repressed by PGE2 in an EP4-dependent manner. Mouse dermal capillary vessels lose pericyte coverage substantially upon PGE2 injection into the skin. Our results suggest that EP-mediated direct disruption of pericytes by PGE2 is a key process for vascular destabilization. Restoring pericyte–EC interaction using inhibitors of PGE2 signaling may offer a therapeutic strategy in cancer and neurological disorders, in which pericyte dysfunction contributes to the disease progression.
Collapse
Affiliation(s)
- Carole Y Perrot
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, 33701, USA.,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Jose L Herrera
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, 33701, USA.,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Ashley E Fournier-Goss
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, 33701, USA.,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA
| | - Masanobu Komatsu
- Cancer and Blood Disorders Institute and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St. Petersburg, FL, 33701, USA. .,Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, 21287, USA.
| |
Collapse
|
22
|
Vähätupa M, Järvinen TAH, Uusitalo-Järvinen H. Exploration of Oxygen-Induced Retinopathy Model to Discover New Therapeutic Drug Targets in Retinopathies. Front Pharmacol 2020; 11:873. [PMID: 32595503 PMCID: PMC7300227 DOI: 10.3389/fphar.2020.00873] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 05/27/2020] [Indexed: 12/11/2022] Open
Abstract
Oxygen-induced retinopathy (OIR) is a pure hypoxia-driven angiogenesis model and the most widely used model for ischemic retinopathies, such as retinopathy of prematurity (ROP), proliferative diabetic retinopathy (PDR), and retinal vein occlusion (RVO). OIR model has been used to test new potential anti-angiogenic factors for human diseases. We have recently performed the most comprehensive characterization of OIR by a relatively novel mass spectrometry (MS) technique, sequential window acquisition of all theoretical fragment ion mass spectra (SWATH-MS) proteomics and used genetically modified mice strains to identify novel molecular drug targets in angiogenic retinal diseases. We have confirmed the relevance of the identified molecular targets to human diseases by determining their expression pattern in neovascular membranes obtained from PDR and RVO patients. Based on our results, crystallins were the most prominent proteins induced by early hypoxic environment during the OIR, while actomyosin complex and Filamin A-R-Ras axis, that regulates vascular permeability of the angiogenic blood vessels, stood out at the peak of angiogenesis. Our results have revealed potential new therapeutic targets to address hypoxia-induced pathological angiogenesis and the associated vascular permeability in number of retinal diseases.
Collapse
Affiliation(s)
- Maria Vähätupa
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Tero A. H. Järvinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Department of Orthopedics and Traumatology, Tampere University Hospital, Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
- Eye Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
23
|
Shi X, Luo X, Xu X. Dimethylarginine dimethylaminohydrolase-1 contributes to exercise-induced cardiac angiogenesis in mice. Biosci Trends 2020; 14:115-122. [PMID: 32238672 DOI: 10.5582/bst.2019.01351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Affiliation(s)
- Xiaowei Shi
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xueting Luo
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
| | - Xin Xu
- School of Kinesiology, Shanghai University of Sport, Shanghai, China
- Department of Exercise Rehabilitation, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
24
|
Wei X, Adak S, Zayed M, Yin L, Feng C, Speck SL, Kathayat RS, Zhang Q, Dickinson BC, Semenkovich CF. Endothelial Palmitoylation Cycling Coordinates Vessel Remodeling in Peripheral Artery Disease. Circ Res 2020; 127:249-265. [PMID: 32233916 DOI: 10.1161/circresaha.120.316752] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
RATIONALE Peripheral artery disease, common in metabolic syndrome and diabetes mellitus, responds poorly to medical interventions and is characterized by chronic vessel immaturity leading to lower extremity amputations. OBJECTIVE To define the role of reversible palmitoylation at the endothelium in the maintenance of vascular maturity. METHODS AND RESULTS Endothelial knockout of the depalmitoylation enzyme APT-1 (acyl-protein thioesterase 1) in mice impaired recovery from chronic hindlimb ischemia, a model of peripheral artery disease. Endothelial APT-1 deficiency decreased fibronectin processing, disrupted adherens junctions, and inhibited in vitro lumen formation. In an unbiased palmitoylation proteomic screen of endothelial cells from genetically modified mice, R-Ras, known to promote vessel maturation, was preferentially affected by APT-1 deficiency. R-Ras was validated as an APT-1 substrate, and click chemistry analyses demonstrated increased R-Ras palmitoylation in cells with APT-1 deficiency. APT-1 enzyme activity was decreased in endothelial cells from db/db mice. Hyperglycemia decreased APT-1 activity in human umbilical vein endothelial cells, due, in part, to altered acetylation of the APT-1 protein. Click chemistry analyses demonstrated increased R-Ras palmitoylation in the setting of hyperglycemia. Altered R-Ras trafficking, increased R-Ras palmitoylation, and fibronectin retention were found in diabetes mellitus models. Loss of R-Ras depalmitoylation caused by APT-1 deficiency constrained R-Ras membrane trafficking, as shown by total internal reflection fluorescence imaging. To rescue cellular phenotypes, we generated an R-Ras molecule with an inserted hydrophilic domain to circumvent membrane rigidity caused by defective palmitoylation turnover. This modification corrected R-Ras membrane trafficking, restored fibronectin processing, increased adherens junctions, and rescued defective lumen formation induced by APT-1 deficiency. CONCLUSIONS These results suggest that endothelial depalmitoylation is regulated by the metabolic milieu and controls plasma membrane partitioning to maintain vascular homeostasis.
Collapse
Affiliation(s)
- Xiaochao Wei
- From the Division of Endocrinology, Metabolism and Lipid Research (X.W., S.A., L.Y., C.F., S.L.S., Q.Z., C.F.S.), Washington University, St Louis, MO
| | - Sangeeta Adak
- From the Division of Endocrinology, Metabolism and Lipid Research (X.W., S.A., L.Y., C.F., S.L.S., Q.Z., C.F.S.), Washington University, St Louis, MO
| | - Mohamed Zayed
- Section of Vascular Surgery, Department of Surgery (M.Z.), Washington University, St Louis, MO.,Veterans Affairs St Louis Health Care System, MO (M.Z.)
| | - Li Yin
- From the Division of Endocrinology, Metabolism and Lipid Research (X.W., S.A., L.Y., C.F., S.L.S., Q.Z., C.F.S.), Washington University, St Louis, MO
| | - Chu Feng
- From the Division of Endocrinology, Metabolism and Lipid Research (X.W., S.A., L.Y., C.F., S.L.S., Q.Z., C.F.S.), Washington University, St Louis, MO
| | - Sarah L Speck
- From the Division of Endocrinology, Metabolism and Lipid Research (X.W., S.A., L.Y., C.F., S.L.S., Q.Z., C.F.S.), Washington University, St Louis, MO
| | - Rahul S Kathayat
- Department of Chemistry, University of Chicago, IL (R.S.K., B.C.D.)
| | - Qiang Zhang
- From the Division of Endocrinology, Metabolism and Lipid Research (X.W., S.A., L.Y., C.F., S.L.S., Q.Z., C.F.S.), Washington University, St Louis, MO
| | | | - Clay F Semenkovich
- From the Division of Endocrinology, Metabolism and Lipid Research (X.W., S.A., L.Y., C.F., S.L.S., Q.Z., C.F.S.), Washington University, St Louis, MO.,Department of Cell Biology and Physiology (C.F.S.), Washington University, St Louis, MO
| |
Collapse
|
25
|
Takino JI, Sato T, Nagamine K, Hori T. The inhibition of Bax activation-induced apoptosis by RasGRP2 via R-Ras-PI3K-Akt signaling pathway in the endothelial cells. Sci Rep 2019; 9:16717. [PMID: 31723205 PMCID: PMC6854084 DOI: 10.1038/s41598-019-53419-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Accepted: 10/15/2019] [Indexed: 12/31/2022] Open
Abstract
Apoptosis of endothelial cells is a very important event in various diseases and angiogenesis. We recently reported that ras guanyl nucleotide releasing protein 2 (RasGRP2), which is a guanine nucleotide exchange factor, was expressed in the human umbilical vein endothelial cells (HUVECs) and that Rap1 activation by its overexpression inhibited apoptosis by suppressing tumor necrosis factor-α induced-reactive oxygen species (ROS) production. However, other signaling pathways and roles of RasGRP2 not mediated via Rap1 are not well understood. Therefore, we compared the Mock (M) and the RasGRP2-stable overexpression (R) immortalized HUVECs using BAM7 and anisomycin, which are apoptosis inducers. BAM7 and anisomycin induced apoptosis without causing ROS production, and such apoptosis was significantly increased in M cells, but not in R cells. RasGRP2 suppressed BAM7- and anisomycin-induced apoptosis, but not via the Rap1 pathway as observed using Rap1 knockdown. Furthermore, RasGRP2 activated not only Rap1 but also R-Ras, and suppressed apoptosis by activating R-Ras-phosphoinositide 3-kinase (PI3K)-Akt signaling pathway. The phosphorylation of Akt by RasGRP2 inhibited Bax translocation by promoting translocation of hexokinase-2 (HK-2) from cytoplasm to mitochondria. Taken together, it was suggested that RasGRP2 suppresses the Bax activation-induced apoptosis by promoting HK-2 translocation to mitochondria via R-Ras-PI3K-Akt signaling pathway.
Collapse
Affiliation(s)
- Jun-Ichi Takino
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan.
| | - Takuma Sato
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan
| | - Kentaro Nagamine
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan
- Department of Clinical Nutrition, Hiroshima International University, Hiroshima, Japan
| | - Takamitsu Hori
- Laboratory of Biochemistry, Hiroshima International University, Hiroshima, Japan
| |
Collapse
|
26
|
Affiliation(s)
- Tero Ah Järvinen
- Faculty of Medicine and Health Technologies, Tampere University and Department of Orthopedics & Traumatology, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
27
|
Wang K, Peng K. RRAS2 knockdown suppresses osteosarcoma progression by inactivating the MEK/ERK signaling pathway. Anticancer Drugs 2019; 30:933-939. [PMID: 31517733 DOI: 10.1097/cad.0000000000000799] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aberrant function of RRAS2 drives malignant transformation in a various of cancers. However, little information exists on the function of RRAS2 in tumorigenesis of osteosarcoma. In this study, we investigated the effect of RRAS2 on osteosarcoma progression and its underlying mechanism. The gene expression level and prognostic power of RRAS2 in osteosarcoma were first investigated using the data from the Gene Expression Omnibus database. Then RNA interference was performed to silence the expression of RRAS2 in osteosarcoma cells. Quantitative real-time-PCR and western blot were used to examine the gene and protein expressions of RRAS2 in osteosarcoma cells. In-vitro cancer proliferation and migration were determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolum bromide solution and wound-healing assays, respectively. We found that RRAS2 was significantly upregulated in osteosarcoma cells and high expression of RRAS2 was associated with a poor prognosis for patients with osteosarcoma. RNA interference decreased the gene and protein expression of RRAS2, reduced in-vitro the proliferation and migration of osteosarcoma cells, and suppressed the activation of the MEK/ERK signaling pathway. RRAS2 as an adverse prognostic factor promoted cell proliferation and migration by activating the MEK/ERK signaling pathway, and may provide new therapeutic value for osteosarcoma.
Collapse
Affiliation(s)
- Kejun Wang
- Department of Orthopaedics, Jingzhou Central Hospital, Jingzhou
| | - Kan Peng
- Department of Trauma Orthopaedics, West China Hospital of Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
28
|
Stephens CJ, Spector JA, Butcher JT. Biofabrication of thick vascularized neo-pedicle flaps for reconstructive surgery. Transl Res 2019; 211:84-122. [PMID: 31170376 PMCID: PMC6702068 DOI: 10.1016/j.trsl.2019.05.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 05/06/2019] [Accepted: 05/14/2019] [Indexed: 01/01/2023]
Abstract
Wound chronicity due to intrinsic and extrinsic factors perturbs adequate lesion closure and reestablishment of the protective skin barrier. Immediate and proper care of chronic wounds is necessary for a swift recovery and a reduction of patient vulnerability to infection. Advanced therapies supplemented with standard wound care procedures have been clinically implemented to restore aberrant tissue; however, these treatments are ineffective if local vasculature is too compromised to support minimally-invasive strategies. Autologous "flaps", which are tissues equipped with their own hierarchical vascular supply, can be harvested from one region of the patient and transplanted to the wound where it is reperfused upon microsurgical anastomosis to appropriate recipient vessels. Despite the success of autologous flap transfer, these procedures are extremely invasive, incur obligatory donor-site morbidity, and require sufficient donor-tissue availability, microsurgical expertise, and specialized equipment. 3D-bioprinting modalities, such as extrusion-based bioprinting, can be used to address the clinical constraints of autologous flap transfer, primarily addressing donor-site morbidity and tissue availability. This advancement in regenerative medicine allows the biofabrication of heterogeneous tissue structures with high shape fidelity and spatial resolution to generate biomimetic constructs with the anatomically-precise geometries of native tissue to ensure tissue-specific function. Yet, meaningful progress toward this clinical application has been limited by the lack of vascularization required to meet the nutrient and oxygen demands of clinically relevant tissue volumes. Thus, various criteria for the fabrication of functional tissues with hierarchical, patent vasculature must be considered when implementing 3D-bioprinting technologies for deep, chronic wounds.
Collapse
Affiliation(s)
- Chelsea J Stephens
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York
| | - Jason A Spector
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York; Division of Plastic Surgery, Weill Cornell Medical College, New York, New York
| | - Jonathan T Butcher
- Nancy E. and Peter C. Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York.
| |
Collapse
|
29
|
Wen N, Guo B, Zheng H, Xu L, Liang H, Wang Q, Wang D, Chen X, Zhang S, Li Y, Zhang L. Bromodomain inhibitor jq1 induces cell cycle arrest and apoptosis of glioma stem cells through the VEGF/PI3K/AKT signaling pathway. Int J Oncol 2019; 55:879-895. [PMID: 31485609 PMCID: PMC6741838 DOI: 10.3892/ijo.2019.4863] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/13/2019] [Indexed: 12/12/2022] Open
Abstract
Bromodomain and extraterminal domain proteins, especially bromodomain-containing protein 4 (Brd4), have recently emerged as therapeutic targets for several cancers, although the role and mechanism of Brd4 in glioblastoma multiforme (GBM) are unclear. In this study, we aimed to explore the underlying mechanisms of the anti-tumor effects of Brd4 and the bromodomain inhibitor JQ1 on glioma stem cells (GSCs). In vitro, JQ1 and small interfering RNAs targeting Brd4 (siBrd4) inhibited the proliferation and self-renewal of GSCs. In vivo, JQ1 significantly inhibited the growth of xenograft GSCs tumors. The RNA-seq analysis revealed that the PI3K-AKT pathway played an important role in GBM. Vascular endothelial growth factor (VEGF) and VEGF receptor 2 phosphorylation was downregulated by exposure to JQ1 in GSCs, thereby reducing PI3K and AKT activity. In addition, treatment with JQ1 inhibited MMP expression, thereby inhibiting degradation of the extracellular matrix by MMP and angiogenesis in GBM tumors. Suppression of AKT phosphorylation inhibited the expression of the retinoblastoma/E2F1 complex, resulting in cell cycle arrest. In addition, treatment with siBrd4 or JQ1 induced apoptosis by activating AKT downstream target genes involved in apoptosis. In conclusion, these results suggest that Brd4 has great potential as a therapeutic target, and JQ1 has notable anti-tumor effects against GBM which may be mediated via the VEGF/PI3K/AKT signaling pathway.
Collapse
Affiliation(s)
- Naiyan Wen
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Baofeng Guo
- Department of Plastic Surgery, China‑Japan Union Hospital, Jilin University, Changchun, Jilin 130033, P.R. China
| | - Hongwu Zheng
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | - Libo Xu
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Hang Liang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Qian Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ding Wang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Xuyang Chen
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Shengnan Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Yang Li
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| | - Ling Zhang
- Department of Pathophysiology, College of Basic Medical Sciences, Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
30
|
Vähätupa M, Nättinen J, Jylhä A, Aapola U, Kataja M, Kööbi P, Järvinen TAH, Uusitalo H, Uusitalo-Järvinen H. SWATH-MS Proteomic Analysis of Oxygen-Induced Retinopathy Reveals Novel Potential Therapeutic Targets. Invest Ophthalmol Vis Sci 2019; 59:3294-3306. [PMID: 30025079 DOI: 10.1167/iovs.18-23831] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose Oxygen-induced retinopathy (OIR) is the most widely used model for ischemic retinopathies such as retinopathy of prematurity (ROP), proliferative diabetic retinopathy (PDR), and retinal vein occlusion (RVO). The purpose of this study was to perform the most comprehensive characterization of OIR by a recently developed technique, sequential window acquisition of all theoretical mass spectra (SWATH-MS) proteomics. Methods Control and OIR retina samples collected from various time points were subjected to SWATH-MS and detailed data analysis. Immunohistochemistry from mouse retinas as well as neovascular membranes from human PDR and RVO patients were used for the detection of the localization of the proteins showing altered expression in the retina and to address their relevance to human ischemic retinopathies. Results We report the most extensive proteomic profiling of OIR to date by quantifying almost 3000 unique proteins and their expression differences between control and OIR retinas. Crystallins were the most prominent proteins induced by hypoxia in the retina, while angiogenesis related proteins such as Filamin A and nonmuscle myosin IIA stand out at the peak of angiogenesis. Majority of the changes in protein expression return to normal at P42, but there is evidence to suggest that proteins involved in neurotransmission remain at reduced level. Conclusions The results reveal new potential therapeutic targets to address hypoxia-induced pathological angiogenesis taking place in number of retinal diseases. The extensive proteomic profiling combined with pathway analysis also identifies novel molecular networks that could contribute to the pathogenesis of retinal diseases.
Collapse
Affiliation(s)
- Maria Vähätupa
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland
| | - Janika Nättinen
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland.,The Center for Proteomics and Personalized Medicine, Tampere, Finland
| | - Antti Jylhä
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland.,The Center for Proteomics and Personalized Medicine, Tampere, Finland
| | - Ulla Aapola
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland.,The Center for Proteomics and Personalized Medicine, Tampere, Finland
| | - Marko Kataja
- Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Peeter Kööbi
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland.,Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Tero A H Järvinen
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland.,Department of Musculoskeletal Disorders, Tampere University Hospital, Tampere, Finland
| | - Hannu Uusitalo
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland.,The Center for Proteomics and Personalized Medicine, Tampere, Finland.,Eye Centre, Tampere University Hospital, Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine & Life Sciences, University of Tampere, Tampere, Finland.,Eye Centre, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
31
|
Khan AQ, Kuttikrishnan S, Siveen KS, Prabhu KS, Shanmugakonar M, Al-Naemi HA, Haris M, Dermime S, Uddin S. RAS-mediated oncogenic signaling pathways in human malignancies. Semin Cancer Biol 2019; 54:1-13. [PMID: 29524560 DOI: 10.1016/j.semcancer.2018.03.001] [Citation(s) in RCA: 119] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 03/06/2018] [Indexed: 02/07/2023]
Abstract
Abnormally activated RAS proteins are the main oncogenic driver that governs the functioning of major signaling pathways involved in the initiation and development of human malignancies. Mutations in RAS genes and or its regulators, most frequent in human cancers, are the main force for incessant RAS activation and associated pathological conditions including cancer. In general, RAS is the main upstream regulator of the highly conserved signaling mechanisms associated with a plethora of important cellular activities vital for normal homeostasis. Mutated or the oncogenic RAS aberrantly activates a web of interconnected signaling pathways including RAF-MEK (mitogen-activated protein kinase kinase)-ERK (extracellular signal-regulated kinase), phosphoinositide-3 kinase (PI3K)/AKT (protein kinase B), protein kinase C (PKC) and ral guanine nucleotide dissociation stimulator (RALGDS), etc., leading to uncontrolled transcriptional expression and reprogramming in the functioning of a range of nuclear and cytosolic effectors critically associated with the hallmarks of carcinogenesis. This review highlights the recent literature on how oncogenic RAS negatively use its signaling web in deregulating the expression and functioning of various effector molecules in the pathogenesis of human malignancies.
Collapse
Affiliation(s)
- Abdul Q Khan
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Shilpa Kuttikrishnan
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Kodappully S Siveen
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | - Kirti S Prabhu
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar
| | | | - Hamda A Al-Naemi
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Mohammad Haris
- Translational Medicine Research Branch, Sidra Medical and Research Center, Doha, Qatar
| | - Said Dermime
- Translational Cancer Research Facility, National Center for Cancer Care and Research, Hamad Medical Corporation, Doha, Qatar
| | - Shahab Uddin
- Academic Health System, Translational Research Institute, Hamad Medical Corporation, Doha, Qatar.
| |
Collapse
|
32
|
Ketomäki T, Vähätupa M, May U, Pemmari T, Ruikka E, Hietamo J, Kaipiainen P, Barker H, Parkkila S, Uusitalo-Järvinen H, Järvinen TAH. R-Ras regulates vascular permeability, but not overall healing in skin wounds. Exp Dermatol 2018; 28:202-206. [PMID: 30489650 DOI: 10.1111/exd.13851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 11/07/2018] [Accepted: 11/20/2018] [Indexed: 12/12/2022]
Abstract
Wounds close by keratinocytes migrating from the edge of the wound and re-epithelializing the epidermis. It has been proposed that the major stimuli for wound closure are blood-derived growth factors, chemokines and cytokines. The small GTPase R-Ras, a known integrin activator, also regulates vascular permeability during angiogenesis, and blood vessels lacking R-Ras leak plasma proteins constantly. We explored whether the access to blood-derived proteins influences skin wound healing in R-Ras knockout (KO) mice. In skin wounds, R-Ras expression was mostly restricted to the vasculature in the granulation tissue. Angiogenic blood vessels in the R-Ras KO mice were significantly more permeable than in wild-type (WT) controls. Although the distances between epidermal tongues, and the panniculus carnosus muscles, were significantly longer in R-Ras KO than WT controls before the granulation tissue formation took place, there were no differences in the wound closure or re-epithelialization rates or granulation tissue formation. These findings were also corroborated in a special splint excision wound model. Our study shows that although R-Ras does not influence the skin wound healing itself, the blood vessels lacking R-Ras are leaky and thus could facilitate the access of blood-derived proteins to the wound.
Collapse
Affiliation(s)
- Tuomo Ketomäki
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Maria Vähätupa
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Ulrike May
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Toini Pemmari
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Ella Ruikka
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Jussi Hietamo
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Pirkka Kaipiainen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Harlan Barker
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland
| | - Seppo Parkkila
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Fimlab laboratories, Eye Centre & Department of Orthopedics & Traumatology, Tampere University Hospital, Tampere, Finland
| | - Hannele Uusitalo-Järvinen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Fimlab laboratories, Eye Centre & Department of Orthopedics & Traumatology, Tampere University Hospital, Tampere, Finland
| | - Tero A H Järvinen
- Faculty of Medicine and Life Sciences, University of Tampere, Tampere, Finland.,Fimlab laboratories, Eye Centre & Department of Orthopedics & Traumatology, Tampere University Hospital, Tampere, Finland
| |
Collapse
|
33
|
Ohata S, Uga H, Okamoto H, Katada T. Small GTPase R-Ras participates in neural tube formation in zebrafish embryonic spinal cord. Biochem Biophys Res Commun 2018; 501:786-790. [PMID: 29772239 DOI: 10.1016/j.bbrc.2018.05.074] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 05/12/2018] [Indexed: 10/16/2022]
Abstract
Ras related (R-Ras), a small GTPase, is involved in the maintenance of apico-basal polarity in neuroepithelial cells of the zebrafish hindbrain, axonal collapse in cultured murine hippocampal neurons, and maturation of blood vessels in adult mice. However, the role of R-Ras in neural tube formation remains unknown. Using antisense morpholino oligonucleotides (AMOs), we found that in the spinal cord of zebrafish embryos, the lumen was formed bilaterally in rras morphants, whereas it was formed at the midline in control embryos. As AMO can cause off-target effects, we generated rras mutant zebrafish lines using CRISPR/Cas9 technology. Although these rras mutant embryos did not have a bilateral lumen in the spinal cord, the following findings suggest that the phenotype is unlikely due to an off-target effect of rras AMO: 1) The rras morphant phenotype was rescued by an injection of AMO-resistant rras mRNA, and 2) a bilaterally segregated spinal cord was not observed in rras mutant embryos injected with rras AMO. The results suggest that the function of other ras family genes may be redundant in rras mutants. Previous research reported a bilaterally formed lumen in the spinal cord of zebrafish embryos with a mutation in a planar cell polarity (PCP) gene, van gogh-like 2 (vangl2). In the present study, in cultured cells, R-Ras was co-immunoprecipitated with Vangl2 but not with another PCP regulator, Pricke1. Interestingly, the interaction between R-Ras and Vangl2 was stronger in guanine-nucleotide free point mutants of R-Ras than in wild-type or constitutively active (GTP-bound) forms of R-Ras. R-Ras may regulate neural tube formation in cooperation with Vangl2 in the developing zebrafish spinal cord.
Collapse
Affiliation(s)
- Shinya Ohata
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan; RIKEN Center for Brain Science, Saitama, 351-0198, Japan.
| | - Hideko Uga
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| | | | - Toshiaki Katada
- Molecular Cell Biology Laboratory, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, Tokyo, 202-8585, Japan; Department of Physiological Chemistry, Graduate School of Pharmaceutical Sciences, University of Tokyo, Tokyo, 113-0033, Japan
| |
Collapse
|
34
|
Studentsova V, Mora KM, Glasner MF, Buckley MR, Loiselle AE. Obesity/Type II Diabetes Promotes Function-limiting Changes in Murine Tendons that are not reversed by Restoring Normal Metabolic Function. Sci Rep 2018; 8:9218. [PMID: 29907811 PMCID: PMC6003963 DOI: 10.1038/s41598-018-27634-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 06/05/2018] [Indexed: 12/19/2022] Open
Abstract
Type II Diabetes (T2DM) negatively alters baseline tendon function, including decreased range of motion and mechanical properties; however, the biological mechanisms that promote diabetic tendinopathy are unknown. To facilitate identification of therapeutic targets we developed a novel murine model of diabetic tendinopathy. Mice fed a High Fat Diet (HFD) developed diet induced obesity and T2DM and demonstrated progressive impairments in tendon gliding function and mechanical properties, relative to mice fed a Low Fat Diet (LFD). We then determined if restoration of normal metabolic function, by switching mice from HFD to LFD, was sufficient to halt the pathological changes in tendon due to obesity/T2DM. However, switching from a HFD to LFD resulted in greater impairments in tendon gliding function than mice maintained on a HFD. Mechanistically, IRβ signaling is decreased in obese/T2DM murine tendons, suggesting altered IRβ signaling as a driver of diabetic tendinopathy. However, knock-down of IRβ expression in S100a4-lineage cells (IRcKOS100a4) was not sufficient to induce diabetic tendinopathy as no impairments in tendon gliding function or mechanical properties were observed in IRcKOS100a4, relative to WT. Collectively, these data define a murine model of diabetic tendinopathy, and demonstrate that restoring normal metabolism does not slow the progression of diabetic tendinopathy.
Collapse
Affiliation(s)
- Valentina Studentsova
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA
| | - Keshia M Mora
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Melissa F Glasner
- Cell Biology of Disease Graduate Program, University of Rochester Medical Center, Rochester, NY, USA
| | - Mark R Buckley
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA.,Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Alayna E Loiselle
- Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY, USA. .,Department of Orthopaedics & Rehabilitation, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
35
|
Perrot CY, Sawada J, Komatsu M. Prolonged activation of cAMP signaling leads to endothelial barrier disruption via transcriptional repression of RRAS. FASEB J 2018; 32:fj201700818RRR. [PMID: 29775418 PMCID: PMC6181640 DOI: 10.1096/fj.201700818rrr] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 04/30/2018] [Indexed: 01/01/2023]
Abstract
The increase in cAMP levels in endothelial cells triggers cellular signaling to alter vascular permeability. It is generally considered that cAMP signaling stabilizes the endothelial barrier function and reduces permeability. However, previous studies have only examined the permeability shortly after cAMP elevation and thus have only investigated acute responses. Because cAMP is a key regulator of gene expression, elevated cAMP may have a delayed but profound impact on the endothelial permeability by altering the expression of the genes that are vital for the vessel wall stability. The small guanosine triphosphate hydrolase Ras-related protein (R-Ras) stabilizes VE-cadherin clustering and enhances endothelial barrier function, thereby stabilizing the integrity of blood vessel wall. Here we show that cAMP controls endothelial permeability through RRAS gene regulation. The prolonged cAMP elevation transcriptionally repressed RRAS in endothelial cells via a cAMP response element-binding protein (CREB) 3-dependent mechanism and significantly disrupted the adherens junction. These effects resulted in a marked increase of endothelial permeability that was reversed by R-Ras transduction. Furthermore, cAMP elevation in the endothelium by prostaglandin E2 or phosphodiesterase type 4 inhibition caused plasma leakage from intact microvessels in mouse skin. Our study demonstrated that, contrary to the widely accepted notion, cAMP elevation in endothelial cells ultimately increases vascular permeability, and the cAMP-dependent RRAS repression critically contributes to this effect.-Perrot, C. Y., Sawada, J., Komatsu, M. Prolonged activation of cyclic AMP signaling leads to endothelial barrier disruption via transcriptional repression of RRAS.
Collapse
Affiliation(s)
- Carole Y. Perrot
- Cancer Center and Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida, USA
| | - Junko Sawada
- Cancer Center and Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida, USA
| | - Masanobu Komatsu
- Cancer Center and Center for Metabolic Origins of Disease, Sanford Burnham Prebys Medical Discovery Institute at Lake Nona, Orlando, Florida, USA
| |
Collapse
|
36
|
Wei Y, Zhou F, Zhou H, Huang J, Yu D, Wu G. Endothelial progenitor cells contribute to neovascularization of non-small cell lung cancer via histone deacetylase 7-mediated cytoskeleton regulation and angiogenic genes transcription. Int J Cancer 2018; 143:657-667. [PMID: 29490434 DOI: 10.1002/ijc.31349] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2017] [Revised: 01/26/2018] [Accepted: 02/15/2018] [Indexed: 12/19/2022]
Abstract
To supply tumor tissues with nutrients and oxygen, endothelial progenitor cells (EPCs) home to tumor sites and contribute to neovascularization. Although the precise mechanism of EPCs-induced neovascularization remains poorly understood in non-small cell lung cancer (NSCLC), histone deacetylase 7 (HDAC7) is considered as a critical regulator. To explore the function of HDAC7 in neovascularization induced by EPCs, tube formation assay, immunofluorescence, microarray, Western blot analysis and animal models were performed. In vitro, HDAC7 abrogation led to the activation of Rho-associated coiled-coil containing protein kinase/myosin light chain 2 pathway concomitant with ERK dephosphorylation, causing the instability of cytoskeleton and collapse of tube formation. In vivo, absence of HDAC7 impaired the vascular lumen integrity and decreased the functional blood perfusion, inhibiting the growth of tumor. At the level of transcription, HDAC7 silencing upregulated antiangiogenic genes and suppressed proangiogenic genes collectively, turning off the angiogenic switch during vessel formation. Taken together, HDAC7 plays a dual role in maintaining the structural and nonstructural functions of EPCs. Our work demonstrates the molecular mechanism by which HDAC7 contributes to the angiogenic property of EPCs and provides a rational basis for specific targeting of antiangiogenic strategies in lung cancer.
Collapse
Affiliation(s)
- Ye Wei
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fangzheng Zhou
- Department of Oncology, Suizhou Hospital, Hubei University of Medicine, Suizhou, Hubei, China
| | - Haibo Zhou
- The First College of Clinical Medical Science, China Three Gorges University and Department of Oncology, Yichang Central People's Hospital, Yichang, Hubei, People's Republic of China
| | - Jing Huang
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Gang Wu
- Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
37
|
Caicedo D, Díaz O, Devesa P, Devesa J. Growth Hormone (GH) and Cardiovascular System. Int J Mol Sci 2018; 19:ijms19010290. [PMID: 29346331 PMCID: PMC5796235 DOI: 10.3390/ijms19010290] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 01/02/2023] Open
Abstract
This review describes the positive effects of growth hormone (GH) on the cardiovascular system. We analyze why the vascular endothelium is a real internal secretion gland, whose inflammation is the first step for developing atherosclerosis, as well as the mechanisms by which GH acts on vessels improving oxidative stress imbalance and endothelial dysfunction. We also report how GH acts on coronary arterial disease and heart failure, and on peripheral arterial disease, inducing a neovascularization process that finally increases flow in ischemic tissues. We include some preliminary data from a trial in which GH or placebo is given to elderly people suffering from critical limb ischemia, showing some of the benefits of the hormone on plasma markers of inflammation, and the safety of GH administration during short periods of time, even in diabetic patients. We also analyze how Klotho is strongly related to GH, inducing, after being released from the damaged vascular endothelium, the pituitary secretion of GH, most likely to repair the injury in the ischemic tissues. We also show how GH can help during wound healing by increasing the blood flow and some neurotrophic and growth factors. In summary, we postulate that short-term GH administration could be useful to treat cardiovascular diseases.
Collapse
Affiliation(s)
- Diego Caicedo
- Department of Angiology and Vascular Surgery, Complejo Hospitalario Universitario de Pontevedra, 36701 Pontevedra, Spain.
| | - Oscar Díaz
- Department of Cardiology, Complejo Hospitalario Universitario de Pontevedra, 36701 Pontevedra, Spain.
| | - Pablo Devesa
- Research and Development, The Medical Center Foltra, 15886 Teo, Spain.
| | - Jesús Devesa
- Scientific Direction, The Medical Center Foltra, 15886 Teo, Spain.
| |
Collapse
|