1
|
Guo B, Gu J, Zhuang T, Zhang J, Fan C, Li Y, Zhao M, Chen R, Wang R, Kong Y, Xu S, Gao W, Liang L, Yu H, Han T. MicroRNA-126: From biology to therapeutics. Biomed Pharmacother 2025; 185:117953. [PMID: 40036996 DOI: 10.1016/j.biopha.2025.117953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/22/2025] [Accepted: 02/27/2025] [Indexed: 03/06/2025] Open
Abstract
MicroRNA-126 (miR-126) has emerged as one of the most extensively studied microRNAs in the context of human diseases, particularly in vascular disorders and cancer. Its high degree of conservation across vertebrates underscores its evolutionary significance and essential functional roles. Extensive research has been devoted to elucidating the molecular mechanisms through which miR-126 modulates key physiological and pathological processes, including angiogenesis, immune response, inflammation, tumor growth, and metastasis. Furthermore, miR-126 plays a causal role in the pathogenesis of various diseases, serving as potential biomarkers for disease prediction, diagnosis, prognosis and drug response, as well as a promising therapeutic target. In this review, we synthesize findings from 283 articles, focusing on the roles of miR-126 in critical biological processes such as cell development, survival, cycle regulation, proliferation, migration, invasion, communication, and metabolism. Additionally, miR-126 represents a promising candidate for miRNA-based therapeutic strategies. A comprehensive understanding and evaluation of miR-126 are crucial for advancing its clinical applications and therapeutic potential.
Collapse
Affiliation(s)
- Bei Guo
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Jia Gu
- Department of Otolaryngology, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Tongtian Zhuang
- Department of Dermatology, Air Force Hospital of Northern Theater Command, Shenyang, China
| | - Jingbin Zhang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Chunyang Fan
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yiyao Li
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Mengdi Zhao
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Ruoran Chen
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Rui Wang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Yuan Kong
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Shuang Xu
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Wei Gao
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Linlang Liang
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China
| | - Hao Yu
- Department of Metabolism and Endocrinology, General Hospital of Northern Theater Command, Shenyang, China.
| | - Tao Han
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
2
|
Yadu N, Singh M, Singh D, Keshavkant S. Mechanistic insights of diabetic wound: Healing process, associated pathways and microRNA-based delivery systems. Int J Pharm 2025; 670:125117. [PMID: 39719258 DOI: 10.1016/j.ijpharm.2024.125117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/27/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Wounds that represent one of the most critical complications can occur in individuals suffering from diabetes mellitus, and results in the need for hospitalisation and, in severe cases, require amputation. This condition is primarily characterized by infections, persistent inflammation, and delayed healing processes, which exacerbate the overall health of the patients. As per the standard mechanism, signalling pathways such as PI3K/AKT, HIF-1, TGF-β, Notch, Wnt/β-Cat, NF-κB, JAK/STAT, TLR, and Nrf2 play major roles in inflammatory, proliferative and remodelling phases of wound healing. However, dysregulation of the above pathways has been seen during the healing of diabetic wounds. MicroRNAs (miRNAs) are small, non-coding RNAs that regulate the expression of various genes and signalling pathways which are associated with the process of wound healing. In the past few years, there has been a great deal of interest in the potential of miRNAs as biological agents in the management of a number of disorders. These miRNAs have been shown to modulate expression of genes involved in the healing process of wounds. There have been previous reviews pertaining to clinical trials examining miRNAs in several disorders, but only a few clinical studies have examined involvement of miRNAs in healing of wounds. Considering the therapeutic promise, there are several obstacles concerning their instabilities and inefficient delivery into the target cells. Therefore, this review is an attempt to discuss precise roles of signalling pathways and miRNAs in different phases of wound healing, and their aberrant regulation in diabetic wounds, particularly. It has also compiled a range of delivery mechanisms as well as an overview of the latest findings pertaining to miRNAs and associated delivery systems for improved healing of diabetic wounds.
Collapse
Affiliation(s)
- Nidhi Yadu
- School of Studies in Biotechnology, Pt. Ravishankar Shukla University, Raipur 492 010, India
| | - Manju Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492 010, India
| | - Deependra Singh
- University Institute of Pharmacy, Pt. Ravishankar Shukla University, Raipur 492 010, India
| | - S Keshavkant
- School of Studies in Biotechnology, Pt. Ravishankar Shukla University, Raipur 492 010, India.
| |
Collapse
|
3
|
Singh D, Memari E, He S, Yusefi H, Helfield B. Cardiac gene delivery using ultrasound: State of the field. Mol Ther Methods Clin Dev 2024; 32:101277. [PMID: 38983873 PMCID: PMC11231612 DOI: 10.1016/j.omtm.2024.101277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/11/2024]
Abstract
Over the past two decades, there has been tremendous and exciting progress toward extending the use of medical ultrasound beyond a traditional imaging tool. Ultrasound contrast agents, typically used for improved visualization of blood flow, have been explored as novel non-viral gene delivery vectors for cardiovascular therapy. Given this adaptation to ultrasound contrast-enhancing agents, this presents as an image-guided and site-specific gene delivery technique with potential for multi-gene and repeatable delivery protocols-overcoming some of the limitations of alternative gene therapy approaches. In this review, we provide an overview of the studies to date that employ this technique toward cardiac gene therapy using cardiovascular disease animal models and summarize their key findings.
Collapse
Affiliation(s)
- Davindra Singh
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Elahe Memari
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Stephanie He
- Department of Biology, Concordia University, Montreal, QC, Canada
| | - Hossein Yusefi
- Department of Physics, Concordia University, Montreal, QC, Canada
| | - Brandon Helfield
- Department of Biology, Concordia University, Montreal, QC, Canada
- Department of Physics, Concordia University, Montreal, QC, Canada
| |
Collapse
|
4
|
Theofilis P, Oikonomou E, Vogiatzi G, Sagris M, Antonopoulos AS, Siasos G, Iliopoulos DC, Perrea D, Vavouranakis M, Tsioufis K, Tousoulis D. The Role of MicroRNA-126 in Atherosclerotic Cardiovascular Diseases. Curr Med Chem 2023; 30:1902-1921. [PMID: 36043750 DOI: 10.2174/0929867329666220830100530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2022] [Revised: 05/02/2022] [Accepted: 05/31/2022] [Indexed: 11/22/2022]
Abstract
Atherosclerotic cardiovascular diseases remain the leading cause of morbidity and mortality worldwide despite all efforts made towards their management. Other than targeting the traditional risk factors for their development, scientific interest has been shifted towards epigenetic regulation, with microRNAs (miRs) being at the forefront. MiR-126, in particular, has been extensively studied in the context of cardiovascular diseases. Downregulated expression of this miR has been associated with highly prevalent cardiovascular risk factors such as arterial hypertension and diabetes mellitus. At the same time, its diagnostic and prognostic capability concerning coronary artery disease is still under investigation, with up-to-date data pointing towards a dysregulated expression in a stable disease state and acute myocardial infarction. Moreover, a lower expression of miR-126 may indicate a higher disease complexity, as well as an increased risk for future major adverse cardiac and cerebrovascular events. Ultimately, overexpression of miR-126 may emerge as a novel therapeutic target in atherosclerotic cardiovascular diseases due to its potential in promoting therapeutic angiogenesis and anti-inflammatory effects. However, the existing challenges in miR therapeutics need to be resolved before translation to clinical practice.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
| | - Evangelos Oikonomou
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
- 3rd Cardiology Department, "Sotiria" Chest Diseases Hospital, University of Athens Medical School, Athens, Greece
| | - Georgia Vogiatzi
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
- 3rd Cardiology Department, "Sotiria" Chest Diseases Hospital, University of Athens Medical School, Athens, Greece
| | - Marios Sagris
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
| | - Alexios S Antonopoulos
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
| | - Gerasimos Siasos
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
- 3rd Cardiology Department, "Sotiria" Chest Diseases Hospital, University of Athens Medical School, Athens, Greece
| | - Dimitrios C Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research "N.S. Christeas", University of Athens Medical School, Athens, Greece
| | - Despoina Perrea
- Laboratory of Experimental Surgery and Surgical Research "N.S. Christeas", University of Athens Medical School, Athens, Greece
| | - Manolis Vavouranakis
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
- 3rd Cardiology Department, "Sotiria" Chest Diseases Hospital, University of Athens Medical School, Athens, Greece
| | - Konstantinos Tsioufis
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
| | - Dimitris Tousoulis
- 1st Cardiology Department, "Hippokration" General Hospital, University of Athens Medical School, Athens, Greece
| |
Collapse
|
5
|
Liu GW, Guzman EB, Menon N, Langer RS. Lipid Nanoparticles for Nucleic Acid Delivery to Endothelial Cells. Pharm Res 2023; 40:3-25. [PMID: 36735106 PMCID: PMC9897626 DOI: 10.1007/s11095-023-03471-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 01/12/2023] [Indexed: 02/04/2023]
Abstract
Endothelial cells play critical roles in circulatory homeostasis and are also the gateway to the major organs of the body. Dysfunction, injury, and gene expression profiles of these cells can cause, or are caused by, prevalent chronic diseases such as diabetes, cardiovascular disease, and cancer. Modulation of gene expression within endothelial cells could therefore be therapeutically strategic in treating longstanding disease challenges. Lipid nanoparticles (LNP) have emerged as potent, scalable, and tunable carrier systems for delivering nucleic acids, making them attractive vehicles for gene delivery to endothelial cells. Here, we discuss the functions of endothelial cells and highlight some receptors that are upregulated during health and disease. Examples and applications of DNA, mRNA, circRNA, saRNA, siRNA, shRNA, miRNA, and ASO delivery to endothelial cells and their targets are reviewed, as well as LNP composition and morphology, formulation strategies, target proteins, and biomechanical factors that modulate endothelial cell targeting. Finally, we discuss FDA-approved LNPs as well as LNPs that have been tested in clinical trials and their challenges, and provide some perspectives as to how to surmount those challenges.
Collapse
Affiliation(s)
- Gary W Liu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
| | - Edward B Guzman
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| | - Nandita Menon
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- Strand Therapeutics, MA, 02215, Boston, USA
| | - Robert S Langer
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
| |
Collapse
|
6
|
Stable Cavitation-Mediated Delivery of miR-126 to Endothelial Cells. Pharmaceutics 2022; 14:pharmaceutics14122656. [PMID: 36559150 PMCID: PMC9784098 DOI: 10.3390/pharmaceutics14122656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 11/21/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022] Open
Abstract
In endothelial cells, microRNA-126 (miR-126) promotes angiogenesis, and modulating the intracellular levels of this gene could suggest a method to treat cardiovascular diseases such as ischemia. Novel ultrasound-stimulated microbubbles offer a means to deliver therapeutic payloads to target cells and sites of disease. The purpose of this study was to investigate the feasibility of gene delivery by stimulating miR-126-decorated microbubbles using gentle acoustic conditions (stable cavitation). A cationic DSTAP microbubble was formulated and characterized to carry 6 µg of a miR-126 payload per 109 microbubbles. Human umbilical vein endothelial cells (HUVECs) were treated at 20−40% duty cycle with miR-126-conjugated microbubbles in a custom ultrasound setup coupled with a passive cavitation detection system. Transfection efficiency was assessed by RT-qPCR, Western blotting, and endothelial tube formation assay, while HUVEC viability was monitored by MTT assay. With increasing duty cycle, the trend observed was an increase in intracellular miR-126 levels, up to a 2.3-fold increase, as well as a decrease in SPRED1 (by 33%) and PIK3R2 (by 46%) expression, two salient miR-126 targets. Under these ultrasound parameters, HUVECs maintained >95% viability after 96 h. The present work describes the delivery of a proangiogenic miR-126 using an ultrasound-responsive cationic microbubble with potential to stimulate therapeutic angiogenesis while minimizing endothelial damage.
Collapse
|
7
|
Xue W, Zhang Q, Chen Y, Zhu Y. Hydrogen Sulfide Improves Angiogenesis by Regulating the Transcription of pri-miR-126 in Diabetic Endothelial Cells. Cells 2022; 11:cells11172651. [PMID: 36078059 PMCID: PMC9455028 DOI: 10.3390/cells11172651] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 08/16/2022] [Accepted: 08/23/2022] [Indexed: 01/19/2023] Open
Abstract
Introduction: Diabetes mellitus results in high rates of cardiovascular disease, such as microcirculation disorder of the lower limbs, with angiogenesis impairment being the main factor. The endothelium functions as a barrier between blood and the vessel wall. Vascular endothelial cell dysfunction caused by hyperglycemia is the main factor leading to angiogenesis impairment. Hydrogen sulfide (H2S) and miR-126-3p are known for their pro-angiogenesis effects; however, little is known about how H2S regulates miR-126-3p to promote angiogenesis under high-glucose conditions. Objectives: The main objective of this research was to explore how H2S regulates the miR-126-3p levels under high-glucose conditions. Methods: We evaluated the pro-angiogenesis effects of H2S in the diabetic hindlimb of an ischemia mice model and in vivo Matrigel plugs. Two microRNA datasets were used to screen microRNAs regulated by both diabetes and H2S. The mRNA and protein levels were detected through real-time PCR and Western blot, respectively. Immunofluorescent staining was also used to assess the capillary density and to evaluate the protein levels in vascular endothelial cells. Human umbilical vein endothelial cells (HUVECs) were used in in vitro experiments. A scratch wound-healing assay was applied to detect the migration ability of endothelial cells. Methylated DNA immunoprecipitation combined with real-time PCR was chosen to identify the DNA methylation level in the HUVECs. Results: Exogenous H2S improved angiogenesis in diabetic mice. miR-126-3p was regulated by both diabetes and H2S. Exogenous H2S up-regulated the miR-126-3p level and recovered the migration rate of endothelial cells via down-regulating the DNMT1 protein level, which was increased by high glucose. Furthermore, DNMT1 upregulation in the HUVECs increased the methylation levels of the gene sequences upstream of miR-126-3p and then inhibited the transcription of primary-miR-126, thus decreasing the miR-126-3p level. CSE overexpression in the HUVECs rescued the miR-126-3p level, by decreasing the methylation level to improve migration. Conclusion: H2S increases the miR-126-3p level through down-regulating the methylation level, by decreasing the DNMT1 protein level induced by high glucose, thus improving the angiogenesis originally impaired by high glucose.
Collapse
Affiliation(s)
- Wenlong Xue
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai 200032, China
| | - Qingqing Zhang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai 200032, China
| | - Ying Chen
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai 200032, China
| | - Yichun Zhu
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Fudan University, Shanghai 200032, China
- Shanghai Key Laboratory of Clinical Geriatric Medicine, Fudan University, Shanghai 200032, China
- Correspondence: ; Tel./Fax: +86-21-5423-7098
| |
Collapse
|
8
|
Moradi SZ, Jalili F, Hoseinkhani Z, Mansouri K. Regenerative Medicine and Angiogenesis; Focused on Cardiovascular Disease. Adv Pharm Bull 2022; 12:686-699. [PMID: 36415645 PMCID: PMC9675929 DOI: 10.34172/apb.2022.072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2020] [Revised: 03/26/2021] [Accepted: 09/27/2021] [Indexed: 10/11/2023] Open
Abstract
Cardiovascular disease (CVD) is a major concern for health with high mortality rates around the world. CVD is often associated with partial or full occlusion of the blood vessel network. Changes in lifestyle can be useful for management early-stage disease but in the advanced stage, surgical interventions or pharmacological are needed to increase the blood flow through the affected tissue or to reduce the energy requirements. Regeneration medicine is a new science that has provided many different options for treating various diseases, especially in CVD over the years. Stem cell therapy, gene therapy, and tissue engineering are some of the powerful branches of the field that have given patients great hope in improving their condition. In this review, we attempted to examine the beneficial effects, challenges, and contradictory effects of angiogenesis in vivo, and in vitro models' studies of CVD. We hope that this information will be able to help other researchers to design new effective structures and open new avenues for the treatment of CVD with the help of angiogenesis and regeneration medicine in the future.
Collapse
Affiliation(s)
- Seyed Zachariah Moradi
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Faramarz Jalili
- Gradute Studies Student, Sobey School of Business, Saint Mary‚S University, Halifax, NS,Canada
| | - Zohreh Hoseinkhani
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Molecular Medicine Department, Faculty of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
9
|
Soltani S, Mansouri K, Parvaneh S, Thakor AS, Pociot F, Yarani R. Diabetes complications and extracellular vesicle therapy. Rev Endocr Metab Disord 2022; 23:357-385. [PMID: 34647239 DOI: 10.1007/s11154-021-09680-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/30/2021] [Indexed: 02/06/2023]
Abstract
Diabetes is a chronic disorder characterized by dysregulated glycemic conditions. Diabetic complications include microvascular and macrovascular abnormalities and account for high morbidity and mortality rates in patients. Current clinical approaches for diabetic complications are limited to symptomatic treatments and tight control of blood sugar levels. Extracellular vesicles (EVs) released by somatic and stem cells have recently emerged as a new class of potent cell-free therapeutic delivery packets with a great potential to treat diabetic complications. EVs contain a mixture of bioactive molecules and can affect underlying pathological processes in favor of tissue healing. In addition, EVs have low immunogenicity and high storage capacity while maintaining nearly the same regenerative and immunomodulatory effects compared to current cell-based therapies. Therefore, EVs have received increasing attention for diabetes-related complications in recent years. In this review, we provide an outlook on diabetic complications and summarizes new knowledge and advances in EV applications. Moreover, we highlight recommendations for future EV-related research.
Collapse
Affiliation(s)
- Setareh Soltani
- Clinical Research Development Center, Taleghani and Imam Ali Hospital, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Kamran Mansouri
- Medical Biology Research Center, Health Technology Institute, Kermanshah, University of Medical Sciences, Kermanshah, Iran
| | - Shahram Parvaneh
- Regenerative Medicine and Cellular Pharmacology Laboratory (HECRIN), Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
- Research Institute of Translational Biomedicine, Department of Dermatology and Allergology, University of Szeged, Szeged, Hungary
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA
| | - Flemming Pociot
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark
| | - Reza Yarani
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University School of Medicine, Palo Alto, CA, 94304, USA.
- Translational Type 1 Diabetes Research, Department of Clinical Research, Steno Diabetes Center Copenhagen, Gentofte, Denmark.
| |
Collapse
|
10
|
Becker AB, Chen L, Ning B, Hu S, Hossack JA, Klibanov AL, Annex BH, French BA. Contrast-Enhanced Ultrasound Reveals Partial Perfusion Recovery After Hindlimb Ischemia as Opposed to Full Recovery by Laser Doppler Perfusion Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1058-1069. [PMID: 35287996 PMCID: PMC9872654 DOI: 10.1016/j.ultrasmedbio.2022.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 02/01/2022] [Accepted: 02/03/2022] [Indexed: 06/03/2023]
Abstract
Mouse models are critical in developing new therapeutic approaches to treat peripheral arterial disease (PAD). Despite decades of research and numerous clinical trials, the efficacy of available therapies is limited. This may suggest shortcomings in our current animal models and/or methods of assessment. We evaluated perfusion measurement methods in a mouse model of PAD by comparing laser Doppler perfusion imaging (LDPI, the most common technique), contrast-enhanced ultrasound (CEUS, an emerging technique) and fluorescent microspheres (conventional standard). Mice undergoing a femoral artery ligation were assessed by LDPI and CEUS at baseline and 1, 4, 7, 14, 28, 60, 90 and 150 d post-surgery to evaluate perfusion recovery in the ischemic hindlimb. Fourteen days after surgery, additional mice were measured with fluorescent microspheres, LDPI, and CEUS. LDPI and CEUS resulted in broadly similar trends of perfusion recovery until 7 d post-surgery. However, by day 14, LDPI indicated full recovery of perfusion, whereas CEUS indicated ∼50% recovery, which failed to improve even after 5 mo. In agreement with the CEUS results, fluorescent microspheres at day 14 post-surgery confirmed that perfusion recovery was incomplete. Histopathology and photoacoustic microscopy provided further evidence of sustained vascular abnormalities.
Collapse
Affiliation(s)
- Alyssa B Becker
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Lanlin Chen
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Bo Ning
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Song Hu
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - John A Hossack
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Alexander L Klibanov
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; Department of Medicine, Cardiovascular Division, University of Virginia, Charlottesville, Virginia, USA
| | - Brian H Annex
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; Department of Medicine, Cardiovascular Division, University of Virginia, Charlottesville, Virginia, USA
| | - Brent A French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA; Department of Medicine, Cardiovascular Division, University of Virginia, Charlottesville, Virginia, USA.
| |
Collapse
|
11
|
Applications of Ultrasound-Mediated Gene Delivery in Regenerative Medicine. Bioengineering (Basel) 2022; 9:bioengineering9050190. [PMID: 35621468 PMCID: PMC9137703 DOI: 10.3390/bioengineering9050190] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/22/2022] [Accepted: 04/23/2022] [Indexed: 11/21/2022] Open
Abstract
Research on the capability of non-viral gene delivery systems to induce tissue regeneration is a continued effort as the current use of viral vectors can present with significant limitations. Despite initially showing lower gene transfection and gene expression efficiencies, non-viral delivery methods continue to be optimized to match that of their viral counterparts. Ultrasound-mediated gene transfer, referred to as sonoporation, occurs by the induction of transient membrane permeabilization and has been found to significantly increase the uptake and expression of DNA in cells across many organ systems. In addition, it offers a more favorable safety profile compared to other non-viral delivery methods. Studies have shown that microbubble-enhanced sonoporation can elicit significant tissue regeneration in both ectopic and disease models, including bone and vascular tissue regeneration. Despite this, no clinical trials on the use of sonoporation for tissue regeneration have been conducted, although current clinical trials using sonoporation for other indications suggest that the method is safe for use in the clinical setting. In this review, we describe the pre-clinical studies conducted thus far on the use of sonoporation for tissue regeneration. Further, the various techniques used to increase the effectiveness and duration of sonoporation-induced gene transfer, as well as the obstacles that may be currently hindering clinical translation, are explored.
Collapse
|
12
|
Floriano JF, Emanueli C, Vega S, Barbosa AMP, Oliveira RGD, Floriano EAF, Graeff CFDO, Abbade JF, Herculano RD, Sobrevia L, Rudge MVC. Pro-angiogenic approach for skeletal muscle regeneration. Biochim Biophys Acta Gen Subj 2022; 1866:130059. [PMID: 34793875 DOI: 10.1016/j.bbagen.2021.130059] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/01/2021] [Indexed: 12/19/2022]
Abstract
The angiogenesis process is a phenomenon in which numerous molecules participate in the stimulation of the new vessels' formation from pre-existing vessels. Angiogenesis is a crucial step in tissue regeneration and recovery of organ and tissue function. Muscle diseases affect millions of people worldwide overcome the ability of skeletal muscle to self-repair. Pro-angiogenic therapies are key in skeletal muscle regeneration where both myogenesis and angiogenesis occur. These therapies have been based on mesenchymal stem cells (MSCs), exosomes, microRNAs (miRs) and delivery of biological factors. The use of different calls of biomaterials is another approach, including ceramics, composites, and polymers. Natural polymers are use due its bioactivity and biocompatibility in addition to its use as scaffolds and in drug delivery systems. One of these polymers is the natural rubber latex (NRL) which is biocompatible, bioactive, versatile, low-costing, and capable of promoting tissue regeneration and angiogenesis. In this review, the advances in the field of pro-angiogenic therapies are discussed.
Collapse
Affiliation(s)
- Juliana Ferreira Floriano
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; National Heart and Lung Institute, Imperial College London, London, UK.
| | - Costanza Emanueli
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Sofia Vega
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile
| | | | | | | | | | - Joelcio Francisco Abbade
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil
| | | | - Luis Sobrevia
- São Paulo State University (UNESP), Botucatu Medical School, Botucatu, São Paulo 18.618-687, Brazil; Cellular and Molecular Physiology Laboratory (CMPL), Department of Obstetrics, Division of Obstetrics and Gynaecology, School of Medicine, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago 8330024, Chile; Department of Physiology, Faculty of Pharmacy, Universidad de Sevilla, Seville E-41012, Spain; University of Queensland, Centre for Clinical Research (UQCCR), Faculty of Medicine and Biomedical Sciences, University of Queensland, Herston, QLD, 4029, Queensland, Australia; Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, 9713GZ Groningen, the Netherlands.
| | | |
Collapse
|
13
|
Contrast Ultrasound, Sonothrombolysis and Sonoperfusion in Cardiovascular Disease: Shifting to Theragnostic Clinical Trials. JACC Cardiovasc Imaging 2022; 15:345-360. [PMID: 34656483 PMCID: PMC8837667 DOI: 10.1016/j.jcmg.2021.07.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 07/23/2021] [Indexed: 02/03/2023]
Abstract
Contrast ultrasound has a variety of applications in cardiovascular medicine, both in diagnosing cardiovascular disease as well as providing prognostic information. Visualization of intravascular contrast microbubbles is based on acoustic cavitation, the characteristic oscillation that results in changes in the reflected ultrasound waves. At high power, this acoustic response generates sufficient shear that is capable of enhancing endothelium-dependent perfusion in atherothrombotic cardiovascular disease (sonoperfusion). The oscillation and collapse of microbubbles in response to ultrasound also induces microstreaming and jetting that can fragment thrombus (sonothrombolysis). Several preclinical studies have focused on identifying optimal diagnostic ultrasound settings and treatment regimens. Clinical trials have been performed in acute myocardial infarction, stroke, and peripheral arterial disease often with improved outcome. In the coming years, results of ongoing clinical trials along with innovation and improvements in sonothrombolysis and sonoperfusion will determine whether this theragnostic technique will become a valuable addition to reperfusion therapy.
Collapse
|
14
|
Walsh AP, Gordon HN, Peter K, Wang X. Ultrasonic particles: An approach for targeted gene delivery. Adv Drug Deliv Rev 2021; 179:113998. [PMID: 34662671 PMCID: PMC8518240 DOI: 10.1016/j.addr.2021.113998] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/24/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
Gene therapy has been widely investigated for the treatment of genetic, acquired, and infectious diseases. Pioneering work utilized viral vectors; however, these are suspected of causing serious adverse events, resulting in the termination of several clinical trials. Non-viral vectors, such as lipid nanoparticles, have attracted significant interest, mainly due to their successful use in vaccines in the current COVID-19 pandemic. Although they allow safe delivery, they come with the disadvantage of off-target delivery. The application of ultrasound to ultrasound-sensitive particles allows for a direct, site-specific transfer of genetic materials into the organ/site of interest. This process, termed ultrasound-targeted gene delivery (UTGD), also increases cell membrane permeability and enhances gene uptake. This review focuses on the advances in ultrasound and the development of ultrasonic particles for UTGD across a range of diseases. Furthermore, we discuss the limitations and future perspectives of UTGD.
Collapse
Affiliation(s)
- Aidan P.G. Walsh
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Henry N. Gordon
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Biochemistry and Pharmacology, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia,Corresponding author at: Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| |
Collapse
|
15
|
Ma J, Zhang Z, Wang Y, Shen H. Investigation of miR-126-3p loaded on adipose stem cell-derived exosomes for wound healing of full-thickness skin defects. Exp Dermatol 2021; 31:362-374. [PMID: 34694648 DOI: 10.1111/exd.14480] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 09/01/2021] [Accepted: 10/16/2021] [Indexed: 11/29/2022]
Abstract
OBJECTIVE To investigate the function of miR-126-3p loaded on adipose stem cell (ADSC)-derived exosomes (ADSC-Exos) in wound healing of full-thickness skin defects. METHODS ADSCs transfected with miR-126-3p mimic, miR-126-3p inhibitor or pcDNA3.1-PIK3R2, or PKH26-marked ADSC-Exos were cultured with fibroblasts or human umbilical vein endothelial cells (HUVECs). The proliferation and migration rates of fibroblasts and angiogenesis of HUVECs were measured. Rats with full-thickness skin defects were injected with ADSC-Exos or exosomes extracted from ADSCs transfected with miR-126-3p inhibitor and the wound healing rates were measured. The wound bed, collagen deposition and angiogenesis in injured rats were assessed. RESULTS ADSC-Exos could be ingested by fibroblasts and HUVECs. Co-incubation with ADSCs or ADSC-Exos promoted the proliferation and migration of fibroblasts and angiogenesis of HUVECs, which was further enhanced by miR-126-3p overexpression. Inhibition of ADSC-Exos or miR-126-3p or PIK3R2 overexpression suppressed the proliferation and migration of fibroblasts and angiogenesis of HUVECs. ADSC-derived exosomal miR-126-3p increased wound healing rate, collagen deposition and newly formed vessels in injured rats. CONCLUSION ADSC-derived exosomal miR-126-3p promotes wound healing of full-thickness skin defects by targeting PIK3R2.
Collapse
Affiliation(s)
- Jie Ma
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Zhaofeng Zhang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Yinmin Wang
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| | - Hua Shen
- Department of Plastic Surgery, Shanghai General Hospital, Shanghai, 201620, P.R. China
| |
Collapse
|
16
|
Wakabayashi I, Sotoda Y, Eguchi R. Contribution of platelet-derived microRNAs to serum microRNAs in healthy men. Platelets 2021; 32:984-987. [PMID: 32865089 DOI: 10.1080/09537104.2020.1810223] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Platelets are a major source of microRNAs (miRNAs) in blood. Relationships between circulating platelet-derived miRNAs were investigated to elucidate their significance as biomarkers. Total miRNAs in serum were analyzed using the 3D-Gene miRNA Oligo chip. Among 22 miRNAs that are included in platelets and play functional roles, sufficient miRNA levels for comparison were detected for 11 miRNAs (let-7b-5p, miR-16-5p, miR-17-5p, miR-24-3p, miR-107, miR-126-3p, miR-150-3p, miR-191-5p, miR-197-3p, miR-223-3p, and miR-326). Among 55 pairs prepared by these miRNAs, relatively strong correlations (Spearman's correlation coefficient >0.8) were shown between miRNAs of 7 pairs including let-7b-5p and miR-16-5p, let-7b-5p and miR-17-5p, let-7b-5p and miR-107, miR-16-5p and miR-17-5p, miR-16-5p and miR-107, miR-17-5p and miR-107, and miR-107 and miR-126-3p. In principal component analysis, the first principal component consisted of let-7b-5p, miR-16-5p, miR-17-5p, miR-107, miR-126-3p, and miR-191-5p. These six miRNAs may be useful biomarkers that reflect platelet condition and function.
Collapse
Affiliation(s)
- Ichiro Wakabayashi
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo Japan
| | - Yoko Sotoda
- Department of Cardiovascular Surgery, Yamagata Saisei Hospital, Yamagata, Japan
| | - Ryoji Eguchi
- Department of Environmental and Preventive Medicine, Hyogo College of Medicine, Nishinomiya, Hyogo Japan
| |
Collapse
|
17
|
Theofilis P, Vogiatzi G, Oikonomou E, Gazouli M, Siasos G, Katifelis H, Perrea D, Vavuranakis M, Iliopoulos DC, Tsioufis C, Tousoulis D. The Effect of MicroRNA-126 Mimic Administration on Vascular Perfusion Recovery in an Animal Model of Hind Limb Ischemia. Front Mol Biosci 2021; 8:724465. [PMID: 34513927 PMCID: PMC8423909 DOI: 10.3389/fmolb.2021.724465] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 08/16/2021] [Indexed: 01/08/2023] Open
Abstract
Background: MicroRNAs have been linked to angiogenesis and could prove to be valuable future therapeutic targets in ischemic cardiovascular diseases. Methods: Ten-week-old male C57Bl/6 mice were subjected to left femoral artery ligation and were treated with microRNA-126 mimic at a dose of 5 mg/kg (Group A, n = 10) or 5 mg/kg microRNA mimic negative control (Group B, n = 10) on days 1, 3, and 7. Laser Doppler imaging was performed to verify successful ligation on day 0 and to evaluate differences in the ischemic-to-normal (I/N) hind limb perfusion ratio on day 28. Muscle tissue expression of microRNA-126 and vascular endothelial growth factor (VEGF) was determined via PCR. Results: Following microRNA-126 mimic administration in Group A subjects, we noted a stepwise increase in I/N hind limb perfusion ratio (Day 0: 0.364 ± 0.032 vs. Day 8: 0.788 ± 0.049 vs. Day 28: 0.750 ± 0.039, p = 0.001). In Group B a stepwise increase in I/N hind limb perfusion ratio was observed (Day 0: 0.272 ± 0.057 vs. Day 8: 0.382 ± 0.020 vs. Day 28: 0.542 ± 0.028, p = 0.074). Muscle tissue expression of microRNA-126 in the ischemic hind limb of Group A was 350-fold lower compared to the ischemic hind limb of Group B (p < 0.001). A higher expression (14.2-fold) of VEGF in the ischemic hind limb of microRNA-126-treated mice compared to that of control group was detected (p < 0.001). A statistically significant negative correlation was noted between microRNA-126 and VEGF tissue expression levels in the ischemic limbs of the entire study population. Conclusion: MicroRNA-126 delivery in the ischemic hind limb of mice improved vascular perfusion with VEGF upregulation.
Collapse
Affiliation(s)
- Panagiotis Theofilis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, Athens, Greece
| | - Georgia Vogiatzi
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, Athens, Greece
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, Athens, Greece
| | - Evangelos Oikonomou
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, Athens, Greece
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, Athens, Greece
| | - Maria Gazouli
- Department of Basic Medical Sciences, Laboratory of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, Athens, Greece
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, Athens, Greece
| | - Hector Katifelis
- Department of Basic Medical Sciences, Laboratory of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Despoina Perrea
- Laboratory of Experimental Surgery and Surgical Research “N.S. Christeas”, University of Athens Medical School, Athens, Greece
| | - Manolis Vavuranakis
- Third Department of Cardiology, Thoracic Diseases General Hospital “Sotiria”, University of Athens Medical School, Athens, Greece
| | - Dimitrios C Iliopoulos
- Laboratory of Experimental Surgery and Surgical Research “N.S. Christeas”, University of Athens Medical School, Athens, Greece
| | - Costas Tsioufis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, Athens, Greece
| | - Dimitris Tousoulis
- First Department of Cardiology, “Hippokration” General Hospital, University of Athens Medical School, Athens, Greece
| |
Collapse
|
18
|
CircDIP2C ameliorates oxidized low-density lipoprotein-induced cell dysfunction by binding to miR-556-5p to induce TET2 in human umbilical vein endothelial cells. Vascul Pharmacol 2021; 139:106887. [PMID: 34147657 DOI: 10.1016/j.vph.2021.106887] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/24/2021] [Accepted: 06/15/2021] [Indexed: 12/20/2022]
Abstract
Circular RNAs (circRNAs) are a group of conserved noncoding RNAs. Recent reports reveal that circRNAs play vital parts in cardiovascular system, including atherosclerosis (AS). The present study is designed to reveal the role of circRNA DIP2C-disco interacting protein 2 homolog C (circDIP2C) in oxidized low-density lipoprotein (ox-LDL)-triggered damage of human umbilical vein endothelial cells (HUVECs). The expression levels of circDIP2C, microRNA-556-5p (miR-556-5p) and tet methylcytosine dioxygenase 2 (TET2) were detected by quantitative real-time polymerase chain reaction (qRT-PCR). Protein expression was determined by western blot analysis. Cell viability and angiogenesis were demonstrated by cell counting kit-8 and tube formation assays, respectively. The levels of reactive oxygen species (ROS) and malondialdehyde (MDA) were checked by ROS and MDA determination assays. Superoxide dismutase (SOD) and lactate dehydrogenase (LDH) activity assays were performed to detect the activity of SOD and LDH. The binding sites of miR-556-5p in circDIP2C or TET2 were predicted by online databases, and identified by dual-luciferase reporter, RNA immunoprecipitation and RNA pull-down assays. CircDIP2C and TET2 expression were obviously decreased, while miR-556-5p expression was increased in ox-LDL-induced HUVECs in comparison with untreated HUVECs. Ox-LDL treatment inhibited cell viability and angiogenesis, promoted oxidative stress, enhanced cytotoxicity and activated NLR family pyrin domain containing 3 (NLRP3) inflammasome pathway. CircDIP2C upregulation protected HUVECs from ox-LDL-induced injury. Additionally, circDIP2C directly bound to miR-556-5p, which was further found to target TET2. MiR-556-5p mimics or TET2 silencing could attenuate the effect of circDIP2C overexpression on ox-LDL-induced cell disorder. Thus, we came a conclusion that circDIP2C protected against ox-LDL-induced HUVEC damage by upregulating TET2 expression through sponging miR-556-5p, which provided a strategy for the therapy of AS.
Collapse
|
19
|
Interfering microRNA-410 attenuates atherosclerosis via the HDAC1/KLF5/IKBα/NF-κB axis. MOLECULAR THERAPY-NUCLEIC ACIDS 2021; 24:646-657. [PMID: 33981482 PMCID: PMC8076652 DOI: 10.1016/j.omtn.2021.03.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 03/10/2021] [Indexed: 01/05/2023]
Abstract
MicroRNA (miR)-410 plays a potential role in the pathogenesis of atherosclerosis. The current study mainly focuses on the underlying mechanism of miR-410/histone deacetylase 1 (HDAC1)/KLF5/nuclear factor κB (NF-κB) inhibitor α (IKBα)/NF-κB axis in atherosclerosis. miR-410 expression was determined using quantitative real-time PCR in both mouse models of atherosclerosis and human umbilical endothelial cells (HUVECs) treated with oxidized low-density lipoprotein (ox-LDL). The study subsequently predicted regulators associated with miR-410 through bioinformatics, and their binding relation was further verified through dual luciferase reporter gene and RNA immunoprecipitation (RIP) assays, and how HDAC1 regulated KLF5 was tested through coimmunoprecipitation (coIP). In HUVECs, miR-410 and HDAC1 mRNA expression; HDAC1, KLF5, IKBα, p65, p-p65, VCAM-1, ICAM-1, and MCP-1 protein expression; and inflammatory cytokine expressions were detected using quantitative real-time PCR, western blot, and ELISA. The present study further tested cell functions by Cell Counting Kit-8 (CCK-8), flow cytometry, and the colony-formation assay. It was revealed that miR-410 could target HDAC1, whereas HDAC1 could target transcription factor KLF5, increasing IKBα expression, thus suppressing NF-κB in atherosclerosis. Furthermore, silencing miR-410 or overexpressing HDAC1 increased cell viability and suppressed apoptosis and an inflammatory reaction in HUVECs in atherosclerosis. Blocking miR-410 promotes HDAC1 expression and increases IKBα levels through KLF5 to suppress NF-κB, thus preventing development of atherosclerosis.
Collapse
|
20
|
Martinez B, Peplow PV. MicroRNAs in laser-induced choroidal neovascularization in mice and rats: their expression and potential therapeutic targets. Neural Regen Res 2021; 16:621-627. [PMID: 33063711 PMCID: PMC8067925 DOI: 10.4103/1673-5374.295271] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Choroidal neovascularization characterizes wet age-related macular degeneration. Choroidal neovascularization formation involves a primarily angiogenic process that is combined with both inflammation and proteolysis. A primary cause of choroidal neovascularization pathogenesis is alterations in pro- and anti-angiogenic factors derived from the retinal pigment epithelium, with vascular endothelium growth factor being mainly responsible for both clinical and experimental choroidal neovascularization. MicroRNAs (miRNAs) which are short, non-coding, endogenous RNA molecules have a major role in regulating various pathological processes, including inflammation and angiogenesis. A review of recent studies with the mouse laser-induced choroidal neovascularization model has shown alterations in miRNA expression in choroidal neovascularization tissues and could be potential therapeutic targets for wet age-related macular degeneration. Upregulation of miR-505 (days 1 and 3 post-laser), miR-155 (day 14) occurred in retina; miR-342-5p (days 3 and 7), miR-126-3p (day 14) in choroid; miR-23a, miR-24, miR-27a (day 7) in retina/choroid; miR-505 (days 1 and 3) in retinal pigment epithelium/choroid; downregulation of miR-155 (days 1 and 3), miR-29a, miR-29b, miR-29c (day 5), miR-93 (day 14), miR-126 (day 14) occurred in retinal pigment epithelium/choroid. Therapies using miRNA mimics or inhibitors were found to decrease choroidal neovascularization lesions. Choroidal neovascularization development was reduced by overexpression of miR-155, miR-188-5p, miR-(5,B,7), miR-126-3p, miR-342-5p, miR-93, miR-126, miR-195a-3p, miR-24, miR-21, miR-31, miR-150, and miR-184, or suppression of miR-505, miR-126-3p, miR-155, and miR-23/27. Further studies are warranted to determine miRNA expression in mouse laser-induced choroidal neovascularization models in order to validate and extend the reported findings. Important experimental variables need to be standardized; these include the strain and age of animals, gender, number and position of laser burns to the eye, laser parameters to induce choroidal neovascularization lesions including wavelength, power, spot size, and duration.
Collapse
Affiliation(s)
- Bridget Martinez
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA; Department of Medicine, St. Georges University School of Medicine, Grenada
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
21
|
Madonna R, Pieragostino D, Rossi C, Guarnieri S, Nagy CT, Giricz Z, Ferdinandy P, Del Boccio P, Mariggiò MA, Geng YJ, De Caterina R. Transplantation of telomerase/myocardin-co-expressing mesenchymal cells in the mouse promotes myocardial revascularization and tissue repair. Vascul Pharmacol 2020; 135:106807. [PMID: 33130246 DOI: 10.1016/j.vph.2020.106807] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 10/26/2020] [Indexed: 12/17/2022]
Abstract
AIM Cell therapies are hampered by poor survival and growth of grafts. We tested whether forced co-expression of telomerase reverse transcriptase (TERT) and myocardin (MYOCD) improves post-infarct revascularization and tissue repair by adipose tissue-derived mesenchymal stromal cells (AT-MSCs). METHODS AND RESULTS We transplanted AT-MSCs overexpressing MYOCD and TERT in a murine model of acute myocardial infarction (AMI). We characterized paracrine effects of AT-MSCs. When transplanted into infarcted hearts of C57BL/6 mice, AT-MSCs overexpressing TERT and MYOCD decreased scar tissue and the intra-scar CD3 and B220 lymphocyte infiltration; and increased arteriolar density as well as ejection fraction compared with saline or mock-transduced AT-MSCs. These effects were accompanied by higher persistence of the injected cells in the heart, increased numbers of Ki-67+ and CD117+ cells, and the expression of cardiac actin and β-myosin heavy chain. Intramyocardial delivery of the secretome and its extracellular vesicle (EV)-enriched fraction also decreased scar tissue formation and increased arteriolar density in the murine AMI model. Proteomic analysis of AT-MSCs-EV-enriched fraction predicted the activation of vascular development and the inhibition of immune cell trafficking. Elevated concentrations of miR-320a, miR-150-5p and miR-126-3p associated with regulation of apoptosis and vasculogenesis were confirmed in the AT-MSCs-EV-enriched fraction. CONCLUSIONS AT-MSCs overexpressing TERT and MYOCD promote persistence of transplanted aged AT-MSCs and enhance arteriolar density in a murine model of AMI. EV-enriched fraction is the component of the paracrine secretion by AT-MSCs with pro-angiogenic and anti-fibrotic activities.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States of America; Chair of Cardiology, Department of Surgical, Medical and Molecular Pathology, University of Pisa, Pisa, Italy.
| | - Damiana Pieragostino
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Claudia Rossi
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Department of Medical, Oral and Biotechnological Sciences, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Simone Guarnieri
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara and StemTeCh Group, Chieti, Italy
| | - Csilla T Nagy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Piero Del Boccio
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Department of Pharmacy, University "G. d'Annunzio" of Chieti-Pescara, Chieti, Italy
| | - Maria Addolorata Mariggiò
- Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, "G. D'Annunzio" University, Chieti-Pescara, Chieti, Italy; Department of Neuroscience, Imaging and Clinical Sciences, "G. d'Annunzio" University, Chieti-Pescara and StemTeCh Group, Chieti, Italy
| | - Yong-Jian Geng
- Department of Internal Medicine, McGovern School of Medicine, The University of Texas Health Science Center at Houston, Houston, TX, United States of America
| | - Raffaele De Caterina
- Chair of Cardiology, Department of Surgical, Medical and Molecular Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
22
|
Gao B, Zhou S, Sun C, Cheng D, Zhang Y, Li X, Zhang L, Zhao J, Xu D, Bai Y. Brain Endothelial Cell-Derived Exosomes Induce Neuroplasticity in Rats with Ischemia/Reperfusion Injury. ACS Chem Neurosci 2020; 11:2201-2213. [PMID: 32574032 DOI: 10.1021/acschemneuro.0c00089] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Exosomes derived from the cerebral endothelial cells play essential roles in protecting neurons from hypoxia injury, but little is known regarding the biological effects and mechanisms of exosomes on brain plasticity. In this study, exosomes were isolated from rodent cerebral endothelial cells (bEnd.3 cells) by ultracentrifugation, either endothelial cell-derived exosomes (EC-Exo) or PBS was injected intraventricularly 2 h after the middle cerebral artery occlusion/reperfusion (MCAO/R) model surgery in the Exo group and control group, respectively. Sham group rats received the same surgical but not ischemic procedure. We evaluated the motor function of rats after MCAO/R, and the foot-fault rate of the Exo group was significantly lower than that of the control group within 23 days (p < 0.05); the Catwalk analysis also showed gait difference between two groups (p < 0.05). On day 28 after MCAO/R, we euthanized the rats, removed the motor cortex from the brain, and then sequenced the genes by using GO and KEGG to find transcriptome analysis of biological terms and functional annotations: The pathway enrichment revealed that the function of synaptic transmission, regulation of synaptic plasticity, and regulation of synaptic vesicle cycle was significantly enriched with the Exo group than control group. Furthermore, the upregulation of synapsin-I expression in the motor cortex (p < 0.05) as well as the increase of the length of the dendrites were found in the Exo group (p < 0.05) than the control group. We determined the content of exosome microRNA levels, and microRNA-126-3p was the highest (TPM) by transcriptome analysis. Moreover, the microRNA-126-3p protected PC12 cells from apoptosis and increased neurite outgrowth, illustrating the mechanism of how exosomes play a role in altering brain plasticity. This study demonstrated that EC-Exo promoted functional motor recovery in the MCAO/R model, exosomes were critical for the reconstruction of synaptic function in ischemic brain injury, and microRNA-126-3p from EC-Exo could serve as a treatment for nerve damage.
Collapse
Affiliation(s)
- Beiyao Gao
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Jing’an District, Shanghai, China 200041
| | - Shaoting Zhou
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, 170 Xinsong Rd, Minhang District, Shanghai, China 201100
| | - Chengcheng Sun
- Rehabilitation Section, Department of Spine Surgery, Tongji Hospital Affiliated to Tongji University, 389 Xincun Road, Putuo District, Shanghai, China 200065
| | - Dandan Cheng
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Jing’an District, Shanghai, China 200041
| | - Ye Zhang
- Rehabilitation Section, Department of Spine Surgery, Tongji Hospital Affiliated to Tongji University, 389 Xincun Road, Putuo District, Shanghai, China 200065
| | - Xutong Li
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, 170 Xinsong Rd, Minhang District, Shanghai, China 201100
| | - Li Zhang
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Jing’an District, Shanghai, China 200041
| | - Jing Zhao
- Department of Neurology, Minhang Hospital Affiliated to Fudan University, 170 Xinsong Rd, Minhang District, Shanghai, China 201100
| | - Dongsheng Xu
- Department of Rehabilitation Medicine, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, 110 Ganhe Road,
Hongkou District, Shanghai, China 201203
- School of Rehabilitation Science, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai, China 201203
- Institute of Rehabilitation Medicine, Shanghai University of Traditional Chinese Medicine, 1200 Cailun Road, Pudong New District, Shanghai, China 201203
| | - Yulong Bai
- Department of Rehabilitation Medicine, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Jing’an District, Shanghai, China 200041
| |
Collapse
|
23
|
Norris EG, Dalecki D, Hocking DC. Using Acoustic Fields to Fabricate ECM-Based Biomaterials for Regenerative Medicine Applications. RECENT PROGRESS IN MATERIALS 2020; 2:1-24. [PMID: 33604591 PMCID: PMC7889011 DOI: 10.21926/rpm.2003018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Ultrasound is emerging as a promising tool for both characterizing and fabricating engineered biomaterials. Ultrasound-based technologies offer a diverse toolbox with outstanding capacity for optimization and customization within a variety of therapeutic contexts, including improved extracellular matrix-based materials for regenerative medicine applications. Non-invasive ultrasound fabrication tools include the use of thermal and mechanical effects of acoustic waves to modify the structure and function of extracellular matrix scaffolds both directly, and indirectly via biochemical and cellular mediators. Materials derived from components of native extracellular matrix are an essential component of engineered biomaterials designed to stimulate cell and tissue functions and repair or replace injured tissues. Thus, continued investigations into biological and acoustic mechanisms by which ultrasound can be used to manipulate extracellular matrix components within three-dimensional hydrogels hold much potential to enable the production of improved biomaterials for clinical and research applications.
Collapse
Affiliation(s)
- Emma G Norris
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
| | - Diane Dalecki
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| | - Denise C Hocking
- Department of Pharmacology and Physiology, University of Rochester, Rochester, New York, USA
- Department of Biomedical Engineering, University of Rochester, Rochester, New York, USA
| |
Collapse
|
24
|
Kong Z, Wang Y, Zhang Y, Shan W, Wu J, Wang Q. MicroRNA-126 promotes endothelial progenitor cell proliferation and migration ability via the Notch pathway. Cardiovasc Diagn Ther 2020; 10:490-499. [PMID: 32695628 DOI: 10.21037/cdt-20-178] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background Effective regulation of the biological function of endothelial progenitor cells (EPCs) is of great importance in its clinical application. This study aimed to explore the effect of microRNA-126 (miR-126) on the proliferation and migration of EPCs and the possible mechanism involved. Methods EPCs was isolated and cultured in vitro, and differences in the expression of miR-126 in endothelial cells (ECs) and EPCs, respectively, were detected by quantitative real-time PCR (RT-PCR). EPCs proliferation was then observed through CCK8 and colony formation experiments. Flow cytometry was also used to observe changes in the cycle and apoptosis of EPCs, and their migration ability was detected by scratch healing and Transwell assays. RT-PCR and Western blotting were carried out to observe the expression of key mRNA molecules and proteins of the Notch pathway. Results The relative expression of miR-126 in the EPCs group were 1.91±0.21, which was significantly higher than that in the EC group (1.25±0.06, P<0.05). When si-miR-126 and si-NC were transfected into the EPCs, it was found that the proliferation ability of cells in the si-miR-126 group decreased significantly (P<0.05), the apoptotic rate of the cells transfected with si-miR-126 was significantly increased, and the cell cycle was blocked at G0/G1 phase. RT-PCR and Western blotting demonstrated that the mRNA and protein expressions of Notch 1 and HES were significantly decreased in the si-miR-126 group. Conclusions miR-126 can effectively promote the proliferation, invasion, and migration of EPCS, while inhibiting apoptosis, through the Notch1 pathway.
Collapse
Affiliation(s)
- Zhaohong Kong
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yunfeng Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Yudi Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China
| | - Wei Shan
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| | - Jianping Wu
- China National Clinical Research Center for Neurological Diseases, Beijing, China.,Advanced Innovation Center for Human Brain Protection, Capital Medical University, Beijing, China
| | - Qun Wang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.,China National Clinical Research Center for Neurological Diseases, Beijing, China.,Beijing Institute for Brain Disorders, Beijing, China
| |
Collapse
|
25
|
Kooiman K, Roovers S, Langeveld SAG, Kleven RT, Dewitte H, O'Reilly MA, Escoffre JM, Bouakaz A, Verweij MD, Hynynen K, Lentacker I, Stride E, Holland CK. Ultrasound-Responsive Cavitation Nuclei for Therapy and Drug Delivery. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:1296-1325. [PMID: 32165014 PMCID: PMC7189181 DOI: 10.1016/j.ultrasmedbio.2020.01.002] [Citation(s) in RCA: 199] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 12/20/2019] [Accepted: 01/07/2020] [Indexed: 05/03/2023]
Abstract
Therapeutic ultrasound strategies that harness the mechanical activity of cavitation nuclei for beneficial tissue bio-effects are actively under development. The mechanical oscillations of circulating microbubbles, the most widely investigated cavitation nuclei, which may also encapsulate or shield a therapeutic agent in the bloodstream, trigger and promote localized uptake. Oscillating microbubbles can create stresses either on nearby tissue or in surrounding fluid to enhance drug penetration and efficacy in the brain, spinal cord, vasculature, immune system, biofilm or tumors. This review summarizes recent investigations that have elucidated interactions of ultrasound and cavitation nuclei with cells, the treatment of tumors, immunotherapy, the blood-brain and blood-spinal cord barriers, sonothrombolysis, cardiovascular drug delivery and sonobactericide. In particular, an overview of salient ultrasound features, drug delivery vehicles, therapeutic transport routes and pre-clinical and clinical studies is provided. Successful implementation of ultrasound and cavitation nuclei-mediated drug delivery has the potential to change the way drugs are administered systemically, resulting in more effective therapeutics and less-invasive treatments.
Collapse
Affiliation(s)
- Klazina Kooiman
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - Silke Roovers
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Simone A G Langeveld
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Robert T Kleven
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA
| | - Heleen Dewitte
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Laboratory for Molecular and Cellular Therapy, Medical School of the Vrije Universiteit Brussel, Jette, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | | | - Ayache Bouakaz
- UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Martin D Verweij
- Department of Biomedical Engineering, Thoraxcenter, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands; Laboratory of Acoustical Wavefield Imaging, Faculty of Applied Sciences, Delft University of Technology, Delft, The Netherlands
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada; Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada; Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Canada
| | - Ine Lentacker
- Ghent Research Group on Nanomedicines, Lab for General Biochemistry and Physical Pharmacy, Department of Pharmaceutical Sciences, Ghent University, Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent University Hospital, Ghent University, Ghent, Belgium
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, Oxford, United Kingdom
| | - Christy K Holland
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, University of Cincinnati, Cincinnati, OH, USA; Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
26
|
Low MicroRNA-126 Levels in Right Ventricular Endomyocardial Biopsies Coincide With Cardiac Allograft Vasculopathy in Heart Transplant Patients. Transplant Direct 2020; 6:e549. [PMID: 32548243 PMCID: PMC7213604 DOI: 10.1097/txd.0000000000000995] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 02/02/2020] [Accepted: 03/02/2020] [Indexed: 11/26/2022] Open
Abstract
Endothelium-enriched microRNAs (miRs) are involved in the development of cardiac allograft vasculopathy (CAV). Recently, serum-derived miR-126-3p and -5p, known endothelial microRNAs with a crucial function in angiogenesis and re-endothelialization, provided additional predictive power for cardiac allograft vasculopathy in addition to clinical predictors. However, their myocardial expression in and relationship with CAV are still unknown. Our study aim was to investigate the expression of endomyocardial microRNA-126-3p and microRNA-126-5p levels in heart transplant recipients and their relationship with allograft vasculopathy. Methods We studied 39 heart transplant recipients, 21 with proven allograft vasculopathy (CAV+) and 18 without allograft vasculopathy (CAV-) with serial coronary angiograms. Additionally, 8 patients with end-stage native coronary artery disease (CAD) were added to the study to investigate disease specificity of the microRNA signature. The mRNA levels of miR-126-3p and miR-126-5p were determined by qRT-PCR in the right ventricular endomyocardial biopsies obtained at baseline and during routine follow-up. Results MiR-126-3p levels were significantly lower in the CAV+ group compared to the CAV- group at follow-up, while miR-126-5p levels were unaltered. This was in stark contrast to native CAD patients in whom miR-126-3p and -5p levels were significantly higher. qPCR levels of miR-126 targets are differentially regulated in CAV versus ischemic cardiomyopathy and are influenced by the administration of immunosuppressive agents in endothelial cells. Conclusions Our data provide evidence for a distinct microRNA signature in heart transplantation patients with allograft vasculopathy. In contrast to CAD patients, lower miR-126-3p levels coincide with the development of cardiac allograft vasculopathy. Further studies in a larger patient population are warranted to determine if the serial measurement of myocardial microRNA-126 products could help in risk assessment and early detection of CAV.
Collapse
|
27
|
Heuslein JL, Gorick CM, Price RJ. Epigenetic regulators of the revascularization response to chronic arterial occlusion. Cardiovasc Res 2020; 115:701-712. [PMID: 30629133 DOI: 10.1093/cvr/cvz001] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/13/2018] [Accepted: 01/03/2019] [Indexed: 12/12/2022] Open
Abstract
Peripheral arterial disease (PAD) is the leading cause of lower limb amputation and estimated to affect over 202 million people worldwide. PAD is caused by atherosclerotic lesions that occlude large arteries in the lower limbs, leading to insufficient blood perfusion of distal tissues. Given the severity of this clinical problem, there has been long-standing interest in both understanding how chronic arterial occlusions affect muscle tissue and vasculature and identifying therapeutic approaches capable of restoring tissue composition and vascular function to a healthy state. To date, the most widely utilized animal model for performing such studies has been the ischaemic mouse hindlimb. Despite not being a model of PAD per se, the ischaemic hindlimb model does recapitulate several key aspects of PAD. Further, it has served as a valuable platform upon which we have built much of our understanding of how chronic arterial occlusions affect muscle tissue composition, muscle regeneration and angiogenesis, and collateral arteriogenesis. Recently, there has been a global surge in research aimed at understanding how gene expression is regulated by epigenetic factors (i.e. non-coding RNAs, histone post-translational modifications, and DNA methylation). Thus, perhaps not unexpectedly, many recent studies have identified essential roles for epigenetic factors in regulating key responses to chronic arterial occlusion(s). In this review, we summarize the mechanisms of action of these epigenetic regulators and highlight several recent studies investigating the role of said regulators in the context of hindlimb ischaemia. In addition, we focus on how these recent advances in our understanding of the role of epigenetics in regulating responses to chronic arterial occlusion(s) can inform future therapeutic applications to promote revascularization and perfusion recovery in the setting of PAD.
Collapse
Affiliation(s)
- Joshua L Heuslein
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd, Box 800759, Health System, Charlottesville, VA, USA
| | - Catherine M Gorick
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd, Box 800759, Health System, Charlottesville, VA, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, 415 Lane Rd, Box 800759, Health System, Charlottesville, VA, USA
| |
Collapse
|
28
|
Peters MM, Sampaio-Pinto V, da Costa Martins PA. Non-coding RNAs in endothelial cell signalling and hypoxia during cardiac regeneration. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118515. [DOI: 10.1016/j.bbamcr.2019.07.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2019] [Revised: 06/19/2019] [Accepted: 07/18/2019] [Indexed: 01/08/2023]
|
29
|
Cui L, Liang J, Liu H, Zhang K, Li J. Nanomaterials for Angiogenesis in Skin Tissue Engineering. TISSUE ENGINEERING PART B-REVIEWS 2020; 26:203-216. [PMID: 31964266 DOI: 10.1089/ten.teb.2019.0337] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Damage to skin tissue, which causes the disorder of the patient's body homeostasis, threatens the patient's life and increases the personal and social treatment burden. Angiogenesis, a key step in the wound healing process, provides sufficient oxygen and nutrients to the wound area. However, traditional clinical interventions are not enough to stabilize the formation of the vascular system to support wound healing. Due to the unique properties and multiple functions of nanomaterials, it has made a major breakthrough in the application of medicine. Nanomaterials provide a more effective treatment to hasten the angiogenesis and wound healing, by stimulating fundamental factors in the vascular regeneration phase. In the present review article, the basic stages and molecular mechanisms of angiogenesis are analyzed, and the types, applications, and prospects of nanomaterials used in angiogenesis are detailed. Impact statement Wound healing (especially chronic wounds) is currently a clinically important issue. The long-term nonhealing of chronic wounds often plagues patients, medical systems, and causes huge losses to the social economy. There is currently no effective method of treating chronic wounds in the clinic. Angiogenesis is an important step in wound healing. Nanomaterials had properties that are not found in conventional materials, and they have been extensively studied in angiogenesis. This review article provides readers with the molecular mechanisms of angiogenesis and the types and applications of angiogenic nanomaterials, hoping to bring inspiration to overcome chronic wounds.
Collapse
Affiliation(s)
- Longlong Cui
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Jiaheng Liang
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Han Liu
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, China
| | - Jingan Li
- Henan Key Laboratory of Advanced Magnesium Alloy, Key Laboratory of Materials Processing and Mold Technology (Ministry of Education), School of Material Science and Engineering, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
30
|
Pérez-Cremades D, Cheng HS, Feinberg MW. Noncoding RNAs in Critical Limb Ischemia. Arterioscler Thromb Vasc Biol 2020; 40:523-533. [PMID: 31893949 DOI: 10.1161/atvbaha.119.312860] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Peripheral artery disease, caused by chronic arterial occlusion of the lower extremities, affects over 200 million people worldwide. Peripheral artery disease can progress into critical limb ischemia (CLI), its more severe manifestation, which is associated with higher risk of limb amputation and cardiovascular death. Aiming to improve tissue perfusion, therapeutic angiogenesis held promise to improve ischemic limbs using delivery of growth factors but has not successfully translated into benefits for patients. Moreover, accumulating studies suggest that impaired downstream signaling of these growth factors (or angiogenic resistance) may significantly contribute to CLI, particularly under harsh environments, such as diabetes mellitus. Noncoding RNAs are essential regulators of gene expression that control a range of pathophysiologies relevant to CLI, including angiogenesis/arteriogenesis, hypoxia, inflammation, stem/progenitor cells, and diabetes mellitus. In this review, we summarize the role of noncoding RNAs, including microRNAs and long noncoding RNAs, as functional mediators or biomarkers in the pathophysiology of CLI. A better understanding of these ncRNAs in CLI may provide opportunities for new targets in the prevention, diagnosis, and therapeutic management of this disabling disease state.
Collapse
Affiliation(s)
- Daniel Pérez-Cremades
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.P.-C., H.S.C., M.W.F.).,Department of Physiology, University of Valencia and INCLIVA Biomedical Research Institute, Spain (D.P.-C.)
| | - Henry S Cheng
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.P.-C., H.S.C., M.W.F.)
| | - Mark W Feinberg
- From the Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA (D.P.-C., H.S.C., M.W.F.)
| |
Collapse
|
31
|
Sugiyama MG, Mintsopoulos V, Raheel H, Goldenberg NM, Batt JE, Brochard L, Kuebler WM, Leong-Poi H, Karshafian R, Lee WL. Lung Ultrasound and Microbubbles Enhance Aminoglycoside Efficacy and Delivery to the Lung in Escherichia coli-induced Pneumonia and Acute Respiratory Distress Syndrome. Am J Respir Crit Care Med 2019; 198:404-408. [PMID: 29638143 DOI: 10.1164/rccm.201711-2259le] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Michael G Sugiyama
- 1 University of Toronto Toronto, Ontario, Canada.,2 St. Michael's Hospital Toronto, Ontario, Canada
| | - Victoria Mintsopoulos
- 1 University of Toronto Toronto, Ontario, Canada.,2 St. Michael's Hospital Toronto, Ontario, Canada
| | - Hira Raheel
- 1 University of Toronto Toronto, Ontario, Canada.,2 St. Michael's Hospital Toronto, Ontario, Canada
| | | | - Jane E Batt
- 1 University of Toronto Toronto, Ontario, Canada.,2 St. Michael's Hospital Toronto, Ontario, Canada
| | - Laurent Brochard
- 1 University of Toronto Toronto, Ontario, Canada.,2 St. Michael's Hospital Toronto, Ontario, Canada
| | - Wolfgang M Kuebler
- 2 St. Michael's Hospital Toronto, Ontario, Canada.,3 Charité - Universitätsmedizin Berlin Berlin, Germany and
| | - Howard Leong-Poi
- 1 University of Toronto Toronto, Ontario, Canada.,2 St. Michael's Hospital Toronto, Ontario, Canada
| | | | - Warren L Lee
- 1 University of Toronto Toronto, Ontario, Canada.,2 St. Michael's Hospital Toronto, Ontario, Canada
| |
Collapse
|
32
|
Cereda M, Xin Y, Goffi A, Herrmann J, Kaczka DW, Kavanagh BP, Perchiazzi G, Yoshida T, Rizi RR. Imaging the Injured Lung: Mechanisms of Action and Clinical Use. Anesthesiology 2019; 131:716-749. [PMID: 30664057 PMCID: PMC6692186 DOI: 10.1097/aln.0000000000002583] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Acute respiratory distress syndrome (ARDS) consists of acute hypoxemic respiratory failure characterized by massive and heterogeneously distributed loss of lung aeration caused by diffuse inflammation and edema present in interstitial and alveolar spaces. It is defined by consensus criteria, which include diffuse infiltrates on chest imaging-either plain radiography or computed tomography. This review will summarize how imaging sciences can inform modern respiratory management of ARDS and continue to increase the understanding of the acutely injured lung. This review also describes newer imaging methodologies that are likely to inform future clinical decision-making and potentially improve outcome. For each imaging modality, this review systematically describes the underlying principles, technology involved, measurements obtained, insights gained by the technique, emerging approaches, limitations, and future developments. Finally, integrated approaches are considered whereby multimodal imaging may impact management of ARDS.
Collapse
Affiliation(s)
- Maurizio Cereda
- Department of Anesthesiology and Critical Care, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Xin
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Alberto Goffi
- Interdepartmental Division of Critical Care Medicine and Department of Medicine, University of Toronto, ON, Canada
| | - Jacob Herrmann
- Departments of Anesthesia and Biomedical Engineering, University of Iowa, IA
| | - David W. Kaczka
- Departments of Anesthesia, Radiology, and Biomedical Engineering, University of Iowa, IA
| | | | - Gaetano Perchiazzi
- Hedenstierna Laboratory and Uppsala University Hospital, Uppsala University, Sweden
| | - Takeshi Yoshida
- Hospital for Sick Children, University of Toronto, ON, Canada
| | - Rahim R. Rizi
- Department of Radiology, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
33
|
Lu Y, Thavarajah T, Gu W, Cai J, Xu Q. Impact of miRNA in Atherosclerosis. Arterioscler Thromb Vasc Biol 2019; 38:e159-e170. [PMID: 30354259 DOI: 10.1161/atvbaha.118.310227] [Citation(s) in RCA: 155] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Yao Lu
- From the Center of Clinical Pharmacology (Y.L.)
| | - Tanuja Thavarajah
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Wenduo Gu
- School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| | - Jingjing Cai
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China
| | - Qingbo Xu
- Department of Cardiology (J.C., Q.X.), Third Xiangya Hospital, Central South University, Changsha, China.,School of Cardiovascular Medicine and Sciences, King's College London British Heart Foundation Centre, United Kingdom (T.T., W.G., Q.X.)
| |
Collapse
|
34
|
Qu M, Pan J, Wang L, Zhou P, Song Y, Wang S, Jiang L, Geng J, Zhang Z, Wang Y, Tang Y, Yang GY. MicroRNA-126 Regulates Angiogenesis and Neurogenesis in a Mouse Model of Focal Cerebral Ischemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:15-25. [PMID: 30825669 PMCID: PMC6393705 DOI: 10.1016/j.omtn.2019.02.002] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 01/30/2019] [Accepted: 02/05/2019] [Indexed: 01/09/2023]
Abstract
Studies demonstrate that microRNA-126 plays a critical role in promoting angiogenesis. However, its effects on angiogenesis following ischemic stroke are unclear. Here, we explored the effect of microRNA-126-3p and microRNA-126-5p on angiogenesis and neurogenesis after brain ischemia. We demonstrated that both microRNA (miRNA)-126-3p and microRNA-126-5p increased the proliferation, migration, and tube formation of human umbilical vein endothelial cells (HUVECs) compared with the scrambled miRNA control (p < 0.05). Transferring microRNA-126 into a mouse middle cerebral artery occlusion model via lentivirus, we found that microRNA-126 overexpression increased the number of CD31+/BrdU+ (5-bromo-2'-deoxyuridine-positive) proliferating endothelial cells and DCX+/BrdU+ neuroblasts in the ischemic mouse brain, improved neurobehavioral outcomes (p < 0.05), and reduced brain atrophy volume (p < 0.05) compared with control mice. Western blot results showed that AKT and ERK signaling pathways were activated in the lentiviral-microRNA-126-treated group (p < 0.05). Both PCR and western blot results demonstrated that tyrosine-protein phosphatase non-receptor type 9 (PTPN9) was decreased in the lentiviral-microRNA-126-treated group (p < 0.05). Dual-luciferase gene reporter assay also showed that PTPN9 was the direct target of microRNA-126-3p and microRNA-126-5p in the ischemic brain. We demonstrated that microRNA-126-3p and microRNA-126-5p promoted angiogenesis and neurogenesis in ischemic mouse brain, and further improved neurobehavioral outcomes. Our mechanistic study further showed that microRNA-126 mediated angiogenesis through directly inhibiting its target PTPN9 and activating AKT and ERK signaling pathways.
Collapse
Affiliation(s)
- Meijie Qu
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Jiaji Pan
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Liping Wang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Panting Zhou
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaying Song
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Shuhong Wang
- Department of Geriatrics, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China
| | - Lu Jiang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Jieli Geng
- Department of Neurology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Zhijun Zhang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yongting Wang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Yaohui Tang
- Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| | - Guo-Yuan Yang
- Department of Neurology, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200025, China; Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China.
| |
Collapse
|
35
|
A meta-analysis of dysregulated miRNAs in coronary heart disease. Life Sci 2018; 215:170-181. [PMID: 30423308 DOI: 10.1016/j.lfs.2018.11.016] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2018] [Revised: 10/24/2018] [Accepted: 11/07/2018] [Indexed: 12/14/2022]
Abstract
AIMS To combine the results of dysregulated miRNAs in individual coronary heart disease (CHD) studies and to identify potential miRNA biomarkers. MAIN METHODS MiRNA profiling studies of CHD were extracted from Pubmed, Embase, Web of Science and China National Knowledge Internet (CNKI) databases if they met the inclusion criteria. The meta-analysis was conducted using a random effects model to identify the effect of each multiple-reported miRNA. We also performed subgroup analysis according to miRNA detecting methods, tissues and subtypes of CHD. Sensitivity analysis was performed on the sample size. Bioinformatic analysis was performed to identify the potential biomatic functions. All results were represented as log10 odds ratios (logORs). KEY FINDINGS A total of 239 miRNAs were reported to be dysregulated in all 25 studies analyzed herein, and meta-analysis identified 48 statistically significant miRNAs. Bioinformatic analysis showed they were closely related with CHD. The most reported up-regulated miRNA was miR-122-5p (logOR: 2.7924, P < 0.001). A total of 7, 6, 4 and 9 miRNAs were detected to be differentially expressed in myocardial infarction (MI), unstable angia (UA), stable angina (SA) and pre-CHD subjects, respectively. 32 miRNAs were dysregulated in blood sample. The dysregulation of miR-133a-3p in whole blood and plasma/serum was contrary. In sensitivity analysis, 37 out of 48 (77.08%) miRNAs were consistently dysregulated. SIGNIFICANCE A total of 48 dysregulated miRNAs were confirmed in this meta-analysis. MiR-122-5p and miR-133a-3p may be valuable biomarkers for CHD.
Collapse
|
36
|
Gene therapy in cardiovascular diseases: A review of recent updates. J Cell Biochem 2018; 119:9645-9654. [DOI: 10.1002/jcb.27303] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 06/27/2018] [Indexed: 11/07/2022]
|
37
|
Heuslein JL, Gorick CM, McDonnell SP, Song J, Annex BH, Price RJ. Exposure of Endothelium to Biomimetic Flow Waveforms Yields Identification of miR-199a-5p as a Potent Regulator of Arteriogenesis. MOLECULAR THERAPY-NUCLEIC ACIDS 2018; 12:829-844. [PMID: 30153567 PMCID: PMC6118158 DOI: 10.1016/j.omtn.2018.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Revised: 07/31/2018] [Accepted: 08/01/2018] [Indexed: 02/07/2023]
Abstract
Arteriogenesis, the growth of endogenous collateral arteries bypassing arterial occlusion(s), is a fundamental shear stress-induced adaptation with implications for treating peripheral arterial disease (PAD). Nonetheless, endothelial mechano-signaling during arteriogenesis is incompletely understood. Here we tested the hypothesis that a mechanosensitive microRNA, miR-199a-5p, regulates perfusion recovery and collateral arteriogenesis following femoral arterial ligation (FAL) via control of monocyte recruitment and pro-arteriogenic gene expression. We have previously shown that collateral artery segments exhibit distinctly amplified arteriogenesis if they are exposed to reversed flow following FAL in the mouse. We performed a genome-wide analysis of endothelial cells exposed to a biomimetic reversed flow waveform. From this analysis, we identified mechanosensitive miR-199a-5p as a novel candidate regulator of collateral arteriogenesis. In vitro, miR-199a-5p inhibited pro-arteriogenic gene expression (IKKβ, Cav1) and monocyte adhesion to endothelium. In vivo, following FAL in mice, miR-199a-5p overexpression impaired foot perfusion and arteriogenesis. In contrast, a single intramuscular anti-miR-199a-5p injection elicited a robust therapeutic response, including complete foot perfusion recovery, markedly augmented arteriogenesis (>3.4-fold increase in segment conductance), and improved gastrocnemius tissue composition. Finally, we found plasma miR-199a-5p to be elevated in human PAD patients with intermittent claudication compared to a risk factor control population. Through our transformative analysis of endothelial mechano-signaling in response to a biomimetic amplified arteriogenesis flow waveform, we have identified miR-199a-5p as both a potent regulator of arteriogenesis and a putative target for treating PAD.
Collapse
Affiliation(s)
- Joshua L Heuslein
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Catherine M Gorick
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Stephanie P McDonnell
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Ji Song
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Brian H Annex
- Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA
| | - Richard J Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA; Robert M. Berne Cardiovascular Research Center, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
38
|
Atanasov G, Dietel C, Feldbrügge L, Benzing C, Krenzien F, Brandl A, Katou S, Schierle K, Robson SC, Splith K, Wiltberger G, Reutzel-Selke A, Jonas S, Pascher A, Bahra M, Pratschke J, Schmelzle M. Angiogenic miRNAs, the angiopoietin axis and related TIE2-expressing monocytes affect outcomes in cholangiocarcinoma. Oncotarget 2018; 9:29921-29933. [PMID: 30042823 PMCID: PMC6057457 DOI: 10.18632/oncotarget.25699] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/12/2018] [Indexed: 12/13/2022] Open
Abstract
Background Tumour angiogenesis is modulated on both an epigenetic and protein level and has potential implications for immune cell responses. However, the importance of related angiogenic biomarkers in cholangiocarcinoma (CCA) is unknown. This study assessed human CCA samples for the expression of angiogenesis-associated microRNAs, angiopoietins (Angs) and monocytes expressing the Ang-receptor, TIE2, with regards to prognostic significance after liver resection. Methods Angiogenic miRNAs were analysed in frozen samples of intrahepatic CCA (iCC; n = 43) and hilar CCA (HC; n = 45). Ang-1 and Ang-2, as well as TIE2-expressing monocytes (TEMs), were detected in paraffin-embedded iCC sections (n = 88). MiRNA expression and the abundance of TEMs and Angs were correlated with clinicopathological characteristics and survival. Results MiR-126 was downregulated in 76.7% of all CCA samples, with high relative expression associated with smaller tumours and reduced lymph node metastasis. High Ang-1 expression was associated with less lymphangiosis carcinomatosa and better histological grading (all p < 0.05). The absence of TEMs in iCC correlated with elevated CA19-9 levels. High relative miR-126 and low miR-128 levels were associated with improved survival in iCC and HC, respectively (all p < 0.05). High miR-126, low miR-128 and TEMs were independent prognostic factors for recurrence-free and overall survival (all p < 0.05). Conclusions These results suggest that angiogenic miRNAs, Angs and TEMs are of prognostic value in CCA. In addition to the possible functional links between angiogenic miRNA expression profiles, Angs and immune-cell responses by TEMs, these data have clinical implications as novel diagnostic tools.
Collapse
Affiliation(s)
- Georgi Atanasov
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Corinna Dietel
- Department of Visceral, Transplantation, Thoracic and Vascular Surgery, University Hospital Leipzig, Leipzig, Germany
| | - Linda Feldbrügge
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Christian Benzing
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Felix Krenzien
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Andreas Brandl
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Shadi Katou
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Katrin Schierle
- Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | - Simon C Robson
- The Transplant Institute and Division of Gastroenterology, Beth Israel Deaconess Medical Center, Harvard University, Boston, MA, USA
| | - Katrin Splith
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Georg Wiltberger
- Department of General, Visceral and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
| | - Anja Reutzel-Selke
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Sven Jonas
- Department of General and Visceral Surgery, 310Klinik Nürnberg, Nürnberg, Germany
| | - Andreas Pascher
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marcus Bahra
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Johann Pratschke
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Moritz Schmelzle
- Department of Surgery, Charité-Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
39
|
Jin H, Li DY, Chernogubova E, Sun C, Busch A, Eken SM, Saliba-Gustafsson P, Winter H, Winski G, Raaz U, Schellinger IN, Simon N, Hegenloh R, Matic LP, Jagodic M, Ehrenborg E, Pelisek J, Eckstein HH, Hedin U, Backlund A, Maegdefessel L. Local Delivery of miR-21 Stabilizes Fibrous Caps in Vulnerable Atherosclerotic Lesions. Mol Ther 2018; 26:1040-1055. [PMID: 29503197 PMCID: PMC6080193 DOI: 10.1016/j.ymthe.2018.01.011] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 01/22/2023] Open
Abstract
miRNAs are potential regulators of carotid artery stenosis and concordant vulnerable atherosclerotic plaques. Hence, we analyzed miRNA expression in laser captured micro-dissected fibrous caps of either ruptured or stable plaques (n = 10 each), discovering that miR-21 was significantly downregulated in unstable lesions. To functionally evaluate miR-21 in plaque vulnerability, miR-21 and miR-21/apolipoprotein-E double-deficient mice (Apoe-/-miR-21-/-) were assessed. miR-21-/- mice lacked sufficient smooth muscle cell proliferation in response to carotid ligation injury. When exposing Apoe-/-miR-21-/- mice to an inducible plaque rupture model, they presented with more atherothrombotic events (93%) compared with miR-21+/+Apoe-/- mice (57%). We discovered that smooth muscle cell fate in experimentally induced advanced lesions is steered via a REST-miR-21-REST feedback signaling pathway. Furthermore, Apoe-/-miR-21-/- mice presented with more pronounced atherosclerotic lesions, greater foam cell formation, and substantially higher levels of arterial macrophage infiltration. Local delivery of a miR-21 mimic using ultrasound-targeted microbubbles into carotid plaques rescued the vulnerable plaque rupture phenotype. In the present study, we identify miR-21 as a key modulator of pathologic processes in advanced atherosclerosis. Targeted, lesion site-specific overexpression of miR-21 can stabilize vulnerable plaques.
Collapse
Affiliation(s)
- Hong Jin
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Daniel Y Li
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | | | - Changyan Sun
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Albert Busch
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Suzanne M Eken
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | | | - Hanna Winter
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Greg Winski
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Uwe Raaz
- University Heart Center, Göttingen, Germany
| | | | - Nancy Simon
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Renate Hegenloh
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Ljubica Perisic Matic
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | - Maja Jagodic
- Department of Clinical Neuroscience, Karolinska Institute, Stockholm, Sweden
| | - Ewa Ehrenborg
- Department of Medicine, Karolinska Institute, Stockholm, Sweden
| | - Jaroslav Pelisek
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Hans-Henning Eckstein
- Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany
| | - Ulf Hedin
- Department of Molecular Medicine and Surgery, Karolinska Institute, Stockholm, Sweden
| | | | - Lars Maegdefessel
- Department of Medicine, Karolinska Institute, Stockholm, Sweden; Department of Vascular and Endovascular Surgery, Technical University Munich, Munich, Germany; German Center for Cardiovascular Research (DZHK), Munich, Germany.
| |
Collapse
|
40
|
AlJaroudi WA, Lloyd SG, Hage FG. Multi-modality imaging: Bird's eye view from the 2017 American Heart Association Scientific Sessions. J Nucl Cardiol 2018; 25:678-684. [PMID: 29362982 DOI: 10.1007/s12350-018-1195-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/05/2018] [Indexed: 02/08/2023]
Abstract
This review summarizes key imaging studies that were presented in the American Heart Association Scientific Sessions 2017 related to the fields of nuclear cardiology, cardiac computed tomography, cardiac magnetic resonance, and echocardiography. The aim of this bird's eye view is to inform readers about multiple studies reported at the meeting from these different imaging modalities. While such a review is most useful for those that did not attend the conference, we find that a general overview may also be useful to those that did since it is often difficult to get exposure to many abstracts at large meetings. The review, therefore, aims to help readers stay updated on the newest imaging studies presented at the meeting and will hopefully stimulate new ideas for future research in imaging.
Collapse
Affiliation(s)
- Wael A AlJaroudi
- Division of Cardiovascular Medicine, Clemenceau Medical Center, Beirut, Lebanon
| | - Steven G Lloyd
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Lyons Harrison Research Building 306, 1900 University BLVD, Birmingham, AL, 35294, USA
- Section of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA
| | - Fadi G Hage
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Lyons Harrison Research Building 306, 1900 University BLVD, Birmingham, AL, 35294, USA.
- Section of Cardiology, Birmingham Veterans Affairs Medical Center, Birmingham, AL, USA.
| |
Collapse
|
41
|
Porter TR, Mulvagh SL, Abdelmoneim SS, Becher H, Belcik JT, Bierig M, Choy J, Gaibazzi N, Gillam LD, Janardhanan R, Kutty S, Leong-Poi H, Lindner JR, Main ML, Mathias W, Park MM, Senior R, Villanueva F. Clinical Applications of Ultrasonic Enhancing Agents in Echocardiography: 2018 American Society of Echocardiography Guidelines Update. J Am Soc Echocardiogr 2018; 31:241-274. [DOI: 10.1016/j.echo.2017.11.013] [Citation(s) in RCA: 142] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
42
|
Matkar PN, Singh KK, Rudenko D, Kim YJ, Kuliszewski MA, Prud'homme GJ, Hedley DW, Leong-Poi H. Novel regulatory role of neuropilin-1 in endothelial-to-mesenchymal transition and fibrosis in pancreatic ductal adenocarcinoma. Oncotarget 2018; 7:69489-69506. [PMID: 27542226 PMCID: PMC5342493 DOI: 10.18632/oncotarget.11060] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 07/18/2016] [Indexed: 12/15/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by an intense fibrotic reaction termed tumor desmoplasia, which is in part responsible for its aggressiveness. Endothelial cells have been shown to display cellular plasticity in the form of endothelial-to-mesenchymal transition (EndMT) that serves as an important source of fibroblasts in pathological disorders, including cancer. Angiogenic co-receptor, neuropilin-1 (NRP-1) actively binds TGFβ1, the primary mediator of EndMT and is involved in oncogenic processes like epithelial-to-mesenchymal transition (EMT). NRP-1 and TGFβ1 signaling have been shown to be aberrantly up-regulated in PDAC. We report herein a positive correlation between NRP-1 levels, EndMT and fibrosis in human PDAC xenografts. Loss of NRP-1 in HUVECs limited TGFβ1-induced EndMT as demonstrated by gain of endothelial and loss of mesenchymal markers, while maintaining endothelial cell architecture. Knockdown of NRP-1 down-regulated TGFβ canonical signaling (pSMAD2) and associated pro-fibrotic genes. Overexpression of NRP-1 exacerbated TGFβ1-induced EndMT and up-regulated TGFβ signaling and expression of pro-fibrotic genes. In vivo, loss of NRP-1 attenuated tumor perfusion and size, accompanied by reduction in EndMT and fibrosis. This study defines a previously unrecognized role of NRP-1 in regulating TGFβ1-induced EndMT and fibrosis, and advocates NRP-1 as a therapeutic target to reduce tumor fibrosis and PDAC progression.
Collapse
Affiliation(s)
- Pratiek N Matkar
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Krishna Kumar Singh
- Division of Vascular Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada.,Division of Cardiac Surgery, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| | - Dmitriy Rudenko
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Yu Jin Kim
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Michael A Kuliszewski
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - Gerald J Prud'homme
- Division of Laboratory Medicine, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada
| | - David W Hedley
- Division of Medical Oncology and Hematology, Ontario Cancer Institute, Campbell Family Cancer Research Institute, Princess Margaret Cancer Centre, Toronto, Canada
| | - Howard Leong-Poi
- Division of Cardiology, Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Canada.,Institute of Medical Science, University of Toronto, Toronto, Canada
| |
Collapse
|
43
|
Johnson MG, Konicke K, Kristianto J, Gustavson A, Garbo R, Wang X, Yuan B, Blank RD. Endothelin signaling regulates mineralization and posttranscriptionally regulates SOST in TMOb cells via miR 126-3p. Physiol Rep 2017; 5:5/4/e13088. [PMID: 28235973 PMCID: PMC5328763 DOI: 10.14814/phy2.13088] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 11/24/2022] Open
Abstract
Previously, our laboratory identified ECE‐1, encoding endothelin‐converting enzyme‐1 (ECE‐1), as a positional candidate for a pleiotropic quantitative trait locus affecting femoral size, shape, and biomechanical performance. We hypothesized that endothelin‐1 (ET‐1) signaling promotes osteogenesis. Exposure of immortalized mouse osteoblast (TMOb) cells to big ET‐1 increased mineralization. Following big ET‐1 treatment, we measured the secretion of insulin‐like‐growth factor‐1 (IGF1), dickkopf‐homolog‐1 protein 1 (DKK1), and sclerostin (SOST). In each case, big ET‐1 signaling changed secretion in a manner that favored increased osteogenic activity. Treatment with ECE‐1, endothelin receptor A (EDNRA), or WNT receptor antagonists inhibited the big ET‐1‐mediated increase in mineralization. In the presence of big ET‐1, message levels of Runx2, Igf1, Dkk1, and Sost are uncoupled from protein production, suggesting posttranscriptional regulation. To evaluate the role of big ET‐1 in normal bone physiology, we inhibited EDNRA signaling during mineralization in the absence of exogenous ET‐1. EDNRA blockade reduced mineralization, decreased IGF1, and increased DKK1 and SOST secretion, responses opposite to those induced by exogenous big ET‐1. Pharmacological and siRNA knockdown to inhibit ECE‐1 reduced mineralization and IGF1 secretion with decreasing DKK1 and decreasing or stable SOST secretion, suggesting a further, unknown role of ECE‐1 in osteoblast maturation. Previously we identified miR 126‐3p as a potential ET‐1‐responsive regulator of SOST in murine cells. Overexpression of miR126‐3p increased mineralization in TMOb cells and decreased SOST secretion. Osteoblasts express the ET‐1 signaling pathway and ET‐1 signaling is necessary for normal osteoblast differentiation and mineralization, acting through regulation of miRs that target osteogenic molecules.
Collapse
Affiliation(s)
- Michael G Johnson
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin .,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Kathryn Konicke
- Medical Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin.,Division of Endocrinology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jasmin Kristianto
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Anne Gustavson
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Rachel Garbo
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Xiaohu Wang
- Medical Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin.,Division of Endocrinology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Baozhi Yuan
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Division of Endocrinology, Department of Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Robert D Blank
- Geriatrics Research, Education and Clinical Center, William S. Middleton Veterans Affairs Hospital, Madison, Wisconsin.,Medical Service, Clement J. Zablocki Veterans Affairs Medical Center, Milwaukee, Wisconsin.,Division of Endocrinology, Department of Medicine, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
44
|
Exosomal miR-126 as a circulating biomarker in non-small-cell lung cancer regulating cancer progression. Sci Rep 2017; 7:15277. [PMID: 29127370 PMCID: PMC5681649 DOI: 10.1038/s41598-017-15475-6] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2016] [Accepted: 10/27/2017] [Indexed: 12/16/2022] Open
Abstract
Lung cancer is one of the leading causes of cancer-related deaths. It is diagnosed mostly at the locally advanced or metastatic stage. Recently, micro RNAs (miRs) and their distribution in circulation have been implicated in physiological and pathological processes. In this study, miR-126 was evaluated in serum, exosome and exosome-free serum fractions in non-small cell lung cancer (NSCLC) patients at early and advanced stages, and compared with healthy controls. Down-regulation of miR-126 was found in serum of advanced stage NSCLC patients. In healthy controls, circulating miR-126 was equally distributed between exosomes and exosome-free serum fractions. Conversely, in both early and advanced stage NSCLC patients, miR-126 was mainly present in exosomes. Different fractions of miR-126 in circulation may reflect different conditions during tumour formation. Incubation of exosomes from early and advanced NSCLC patients induced blood vessel formation and malignant transformation in human bronchial epithelial cells. On the other hand, exosome-enriched miR-126 from normal endothelial cells inhibited cell growth and induces loss of malignancy of NSCLC cells. These findings suggest a role of exo-miRs in the modulation of the NSCLC microenvironmental niche. Exosome-delivered miRs thus hold a substantial promise as a diagnostics biomarker as well as a personalized therapeutic modality.
Collapse
|
45
|
Upregulation of miR-126-3p promotes human saphenous vein endothelial cell proliferation in vitro and prevents vein graft neointimal formation ex vivo and in vivo. Oncotarget 2017; 8:106790-106806. [PMID: 29290989 PMCID: PMC5739774 DOI: 10.18632/oncotarget.22365] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 08/27/2017] [Indexed: 11/25/2022] Open
Abstract
Poor long-term patency of vein grafts remains an obstacle in coronary artery bypass grafting (CABG) surgery using an autologous saphenous vein graft. Recent studies have revealed that miR-126-3p promotes vascular integrity and angiogenesis. We aimed to identify the role of miR-126-3p in the setting of vein graft disease and investigate the value of miR-126-3p agomir as a future gene therapy in vein graft failure. Expression analysis of circulating miR-126-3p in plasma from CABG patients established the basic clues that miR-126-3p participates in CABG. The in vitro results indicated that elevated miR-126-3p expression significantly improved proliferation and migration in human saphenous vein endothelial cells (HSVECs) by targeting sprouty-related protein-1 (SPRED-1) and phosphatidylinositol-3-kinase regulatory subunit 2 (PIK3R2), but not in human saphenous vein smooth muscle cells (HSVSMCs). Moreover, the therapeutic potential of miR-126-3p agomir was demonstrated in cultured human saphenous vein (HSV) ex vivo. Finally, local delivery of miR-126-3p agomir was confirmed to enhance reendothelialization and attenuate neointimal formation in a rat vein arterialization model. In conclusion, we provide evidence that upregulation of miR-126-3p by agomir possesses potential clinical value in the prevention and treatment of autologous vein graft restenosis in CABG.
Collapse
|
46
|
Dron JS, Ho R, Hegele RA. Recent Advances in the Genetics of Atherothrombotic Disease and Its Determinants. Arterioscler Thromb Vasc Biol 2017; 37:e158-e166. [DOI: 10.1161/atvbaha.117.309934] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jacqueline S. Dron
- From the Department of Biochemistry (J.S.D, R.H., R.A.H.), Robarts Research Institute (J.S.D., R.H., R.A.H.), and Department of Medicine (R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Rosettia Ho
- From the Department of Biochemistry (J.S.D, R.H., R.A.H.), Robarts Research Institute (J.S.D., R.H., R.A.H.), and Department of Medicine (R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Robert A. Hegele
- From the Department of Biochemistry (J.S.D, R.H., R.A.H.), Robarts Research Institute (J.S.D., R.H., R.A.H.), and Department of Medicine (R.A.H.), Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
47
|
Juni RP, Abreu RC, da Costa Martins PA. Regulation of microvascularization in heart failure - an endothelial cell, non-coding RNAs and exosome liaison. Noncoding RNA Res 2017; 2:45-55. [PMID: 30159420 PMCID: PMC6096416 DOI: 10.1016/j.ncrna.2017.01.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2016] [Accepted: 01/26/2017] [Indexed: 12/22/2022] Open
Abstract
Heart failure is a complex syndrome involving various pathophysiological processes. An increasing body of evidence shows that the myocardial microvasculature is essential for the homeostasis state and that a decompensated heart is associated with microvascular dysfunction as a result of impaired endothelial angiogenic capacity. The intercellular communication between endothelial cells and cardiomyocytes through various signaling molecules, such as vascular endothelial growth factor, nitric oxide, and non-coding RNAs is an important determinant of cardiac microvascular function. Non-coding RNAs are transported from endothelial cells to cardiomyocytes, and vice versa, regulating microvascular properties and angiogenic processes in the heart. Small-exocytosed vesicles, called exosomes, which are secreted by both cell types, can mediate this intercellular communication. The purpose of this review is to highlight the contribution of the microvasculature to proper heart function maintenance by focusing on the interaction between cardiac endothelial cells and myocytes with a specific emphasis on non-coding RNAs (ncRNAs) in this form of cell-to-cell communication. Finally, the potential of ncRNAs as targets for angiogenesis therapy will also be discussed.
Collapse
Affiliation(s)
- Rio P. Juni
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Ricardo C. Abreu
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
| | - Paula A. da Costa Martins
- Department of Cardiology, CARIM School for Cardiovascular Diseases, Faculty of Health, Medicine and Life Sciences, Maastricht University, Maastricht, The Netherlands
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine, University of Porto, Porto, Portugal
| |
Collapse
|
48
|
Vimalraj S, Sumantran VN, Chatterjee S. MicroRNAs: Impaired vasculogenesis in metal induced teratogenicity. Reprod Toxicol 2017; 70:30-48. [PMID: 28249814 DOI: 10.1016/j.reprotox.2017.02.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 02/14/2017] [Accepted: 02/21/2017] [Indexed: 02/07/2023]
Abstract
Certain metals have been known for their toxic effects on embryos and fetal development. The vasculature in early pregnancy is extremely dynamic and plays an important role in organogenesis. Nascent blood vessels in early embryonic life are considered to be a primary and delicate target for many teratogens since the nascent blood islands follow a tightly controlled program to form vascular plexus around and inside the embryo for resourcing optimal ingredients for its development. The state of the distribution of toxic metals, their transport mechanisms and the molecular events by which they notch extra-embryonic and embryonic vasculatures are illustrated. In addition, pharmacological aspects of toxic metal induced teratogenicity have also been portrayed. The work reviewed state of the current knowledge of specific role of microRNAs (miRNAs) that are differentially expressed in response to toxic metals, and how they interfere with the vasculogenesis that manifests into embryonic anomalies.
Collapse
Affiliation(s)
- Selvaraj Vimalraj
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India.
| | | | - Suvro Chatterjee
- Vascular Biology Lab, AU-KBC Research Centre, Anna University, Chennai, India; Department of Biotechnology, Anna University, Chennai, India.
| |
Collapse
|
49
|
Mofid A, Newman NS, Lee PJH, Abbasi C, Matkar PN, Rudenko D, Kuliszewski MA, Chen HH, Afrasiabi K, Tsoporis JN, Gramolini AO, Connelly KA, Parker TG, Leong-Poi H. Cardiac Overexpression of S100A6 Attenuates Cardiomyocyte Apoptosis and Reduces Infarct Size After Myocardial Ischemia-Reperfusion. J Am Heart Assoc 2017; 6:JAHA.116.004738. [PMID: 28174168 PMCID: PMC5523770 DOI: 10.1161/jaha.116.004738] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background Cardiomyocyte‐specific transgenic mice overexpressing S100A6, a member of the family of EF‐hand calcium‐binding proteins, develop less cardiac hypertrophy, interstitial fibrosis, and myocyte apoptosis after permanent coronary ligation, findings that support S100A6 as a potential therapeutic target after acute myocardial infarction. Our purpose was to investigate S100A6 gene therapy for acute myocardial ischemia‐reperfusion. Methods and Results We first performed in vitro studies to examine the effects of S100A6 overexpression and knockdown in rat neonatal cardiomyocytes. S100A6 overexpression improved calcium transients and protected against apoptosis induced by hypoxia‐reoxygenation via enhanced calcineurin activity, whereas knockdown of S100A6 had detrimental effects. For in vivo studies, human S100A6 plasmid or empty plasmid was delivered to the left ventricular myocardium by ultrasound‐targeted microbubble destruction in Fischer‐344 rats 2 days prior to a 30‐minute ligation of the left anterior descending coronary artery followed by reperfusion. Control animals received no therapy. Pretreatment with S100A6 gene therapy yielded a survival advantage compared to empty‐plasmid and nontreated controls. S100A6‐pretreated animals had reduced infarct size and improved left ventricular systolic function, with less myocyte apoptosis, attenuated cardiac hypertrophy, and less cardiac fibrosis. Conclusions S100A6 overexpression by ultrasound‐targeted microbubble destruction helps ameliorate myocardial ischemia‐reperfusion, resulting in lower mortality and improved left ventricular systolic function post–ischemia‐reperfusion via attenuation of apoptosis, reduction in cardiac hypertrophy, and reduced infarct size. Our results indicate that S100A6 is a potential therapeutic target for acute myocardial infarction.
Collapse
Affiliation(s)
- Azadeh Mofid
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Nadav S Newman
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Paul J H Lee
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Cynthia Abbasi
- Department of Physiology, University of Toronto, Ontario, Canada
| | - Pratiek N Matkar
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Dmitriy Rudenko
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Michael A Kuliszewski
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Hao H Chen
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Kolsoom Afrasiabi
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - James N Tsoporis
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | | | - Kim A Connelly
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Thomas G Parker
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| | - Howard Leong-Poi
- Division of Cardiology, Keenan Research Centre for Biomedical Science, Li Ka Shing Knowledge Institute, St. Michael's Hospital, University of Toronto, Ontario, Canada
| |
Collapse
|
50
|
Wang LH, Tsai HC, Cheng YC, Lin CY, Huang YL, Tsai CH, Xu GH, Wang SW, Fong YC, Tang CH. CTGF promotes osteosarcoma angiogenesis by regulating miR-543/angiopoietin 2 signaling. Cancer Lett 2017; 391:28-37. [PMID: 28108312 DOI: 10.1016/j.canlet.2017.01.013] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2016] [Revised: 12/23/2016] [Accepted: 01/11/2017] [Indexed: 12/24/2022]
Abstract
Osteosarcoma is the most common primary solid tumor of bone. It has a high metastatic potential and occurs predominantly in adolescents and young adults. Angiopoietin 2 (Angpt2) is a key regulator in tumor angiogenesis, facilitating tumor growth and metastasis. Connective tissue growth factor (CTGF, also known as CCN2), is a cysteine-rich protein that has been reported to promote metastasis of osteosarcoma. However, the effect of CTGF on Angpt2 regulation and angiogenesis in human osteosarcoma remains largely unknown. We found that overexpression of CTGF in osteosarcoma cells increased Angpt2 production and induced angiogenesis, in vitro and in vivo. Our findings demonstrate that CTGF-enhanced Angpt2 expression and angiogenesis is mediated by the phospholipase C (PLC)/protein kinase C (PKCδ) signaling pathway. Moreover, endogenous microRNA-543 (miR-543) expression was negatively regulated by CTGF via the PLC/PKCδ pathway. We also provide evidence showing clinical significance between CTGF, Angpt2, and miR-543 as well as tumor staging in human osteosarcoma tissue. CTGF may serve as a therapeutic target in the process of osteosarcoma metastasis and angiogenesis.
Collapse
Affiliation(s)
- Li-Hong Wang
- Department of Orthopedics, Dongyang People's Hospital, Wenzhou Medical University, Dongyang, China
| | - Hsiao-Chi Tsai
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Yu-Che Cheng
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chih-Yang Lin
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Yuan-Li Huang
- Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- School of Medicine, China Medical University, Taichung, Taiwan; Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Guo-Hong Xu
- Department of Orthopedics, Dongyang People's Hospital, Wenzhou Medical University, Dongyang, China
| | - Shih-Wei Wang
- Department of Medicine, Mackay Medical College, New Taipei City, Taiwan
| | - Yi-Chin Fong
- Department of Orthopaedic Surgery, China Medical University Beigang Hospital, Yun-Lin County, Taiwan; Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
| | - Chih-Hsin Tang
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan; Department of Biotechnology, College of Health Science, Asia University, Taichung, Taiwan; School of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|