1
|
Li S, Li F, Wang Y, Li W, Wu J, Hu X, Tang T, Liu X. Multiple delivery strategies of nanocarriers for myocardial ischemia-reperfusion injury: current strategies and future prospective. Drug Deliv 2024; 31:2298514. [PMID: 38147501 PMCID: PMC10763895 DOI: 10.1080/10717544.2023.2298514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 11/30/2023] [Indexed: 12/28/2023] Open
Abstract
Acute myocardial infarction, characterized by high morbidity and mortality, has now become a serious health hazard for human beings. Conventional surgical interventions to restore blood flow can rapidly relieve acute myocardial ischemia, but the ensuing myocardial ischemia-reperfusion injury (MI/RI) and subsequent heart failure have become medical challenges that researchers have been trying to overcome. The pathogenesis of MI/RI involves several mechanisms, including overproduction of reactive oxygen species, abnormal mitochondrial function, calcium overload, and other factors that induce cell death and inflammatory responses. These mechanisms have led to the exploration of antioxidant and inflammation-modulating therapies, as well as the development of myocardial protective factors and stem cell therapies. However, the short half-life, low bioavailability, and lack of targeting of these drugs that modulate these pathological mechanisms, combined with liver and spleen sequestration and continuous washout of blood flow from myocardial sites, severely compromise the expected efficacy of clinical drugs. To address these issues, employing conventional nanocarriers and integrating them with contemporary biomimetic nanocarriers, which rely on passive targeting and active targeting through precise modifications, can effectively prolong the duration of therapeutic agents within the body, enhance their bioavailability, and augment their retention at the injured myocardium. Consequently, these approaches significantly enhance therapeutic effectiveness while minimizing toxic side effects. This article reviews current drug delivery systems used for MI/RI, aiming to offer a fresh perspective on treating this disease.
Collapse
Affiliation(s)
- Shengnan Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Fengmei Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Yan Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Wenqun Li
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Junyong Wu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xiongbin Hu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Tiantian Tang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| | - Xinyi Liu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
- Institution of Clinical Pharmacy, Central South University, Changsha, China
| |
Collapse
|
2
|
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
|
3
|
Ren H, Xiang S, Liu A, Wang Q, Zhou N, Hu Z. A noval noninvasive targeted therapy for osteosarcoma: the combination of LIFU and ultrasound-magnetic-mediated SPIO/TP53/PLGA nanobubble. Front Bioeng Biotechnol 2024; 12:1418903. [PMID: 39007051 PMCID: PMC11239426 DOI: 10.3389/fbioe.2024.1418903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 06/10/2024] [Indexed: 07/16/2024] Open
Abstract
Purpose Osteosarcoma (OS) is the most common type of primary malignant bone tumor. Transducing a functional TP53 gene can effectively inhibit OS cell activity. Poly lactic acid-glycolic acid (PLGA) nanobubbles (NBs) mediated by focused ultrasound (US) can introduce exogenous genes into target cells in animal models, but this technique relies on the passive free diffusion of agents across the body. The inclusion of superparamagnetic iron oxide (SPIO) in microbubbles allows for magnetic-based tissue localization. A low-intensity-focused ultrasound (LIFU) instrument was developed at our institute, and different intensities of LIFU can either disrupt the NBs (RLI-LIFU) or exert cytocidal effects on the target tissues (RHI-LIFU). Based on these data, we performed US-magnetic-mediated TP53-NB destruction and investigated its ability to inhibit OS growth when combined with LIFU both in vitro and in vivo. Methods Several SPIO/TP53/PLGA (STP) NB variants were prepared and characterized. For the in vitro experiments, HOS and MG63 cells were randomly assigned into five treatment groups. Cell proliferation and the expression of TP53 were detected by CCK8, qRT-PCR and Western blotting, respectively. In vivo, tumor-bearing nude mice were randomly assigned into seven treatment groups. The iron distribution of Perls' Prussian blue-stained tissue sections was determined by optical microscopy. TUNEL-DAPI was performed to examine apoptosis. TP53 expression was detected by qRT-PCR and immunohistochemistry. Results SPIO/TP53/PLGA NBs with a particle size of approximately 200 nm were prepared successfully. For in vitro experiments, ultrasound-targeted transfection of TP53 overexpression in OS cells and efficient inhibition of OS proliferation have been demonstrated. Furthermore, in a tumor-bearing nude mouse model, RLI-LIFU-magnetic-mediated SPIO/TP53/PLGA NBs increased the transfection efficiency of the TP53 plasmid, resulting in apoptosis. Adding RHI-LIFU to the treatment regimen significantly increased the apoptosis of OS cells in vivo. Conclusion Combining LIFU and US-magnetic-mediated SPIO/TP53/PLGA NB destruction is potentially a novel noninvasive and targeted therapy for OS.
Collapse
Affiliation(s)
- Honglei Ren
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedic Surgery, ChongQing Red Cross Hospital (People's Hospital of JiangBei District), Chongqing, China
| | - Shanlin Xiang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Aiguo Liu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
- Department of Orthopedic Surgery, The First Affiliated Hospital of Henan University, Kaifeng, China
| | - Qian Wang
- Department of Orthopedics, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Nian Zhou
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhenming Hu
- Department of Orthopedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Orthopedic Surgery, The University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Sun Z, Cai Y, Chen Y, Jin Q, Zhang Z, Zhang L, Li Y, Huang L, Wang J, Yang Y, Lv Q, Han Z, Xie M, Zhu X. Ultrasound-targeted microbubble destruction promotes PDGF-primed bone mesenchymal stem cell transplantation for myocardial protection in acute Myocardial Infarction in rats. J Nanobiotechnology 2023; 21:481. [PMID: 38102643 PMCID: PMC10725038 DOI: 10.1186/s12951-023-02204-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Accepted: 11/07/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND Ultrasound-targeted microbubble destruction (UTMD) has emerged as a promising strategy for the targeted delivery of bone marrow mesenchymal stem cells (MSCs) to the ischemic myocardium. However, the limited migration capacity and poor survival of MSCs remains a major therapeutic barrier. The present study was performed to investigate the synergistic effect of UTMD with platelet-derived growth factor BB (PDGF-BB) on the homing of MSCs for acute myocardial infarction (AMI). METHODS MSCs from male donor rats were treated with PDGF-BB, and a novel microbubble formulation was prepared using a thin-film hydration method. In vivo, MSCs with or without PDGF-BB pretreatment were transplanted by UTMD after inducing AMI in experimental rats. The therapeutic efficacy of PDGF-BB-primed MSCs on myocardial apoptosis, angiogenesis, cardiac function and scar repair was estimated. The effects and molecular mechanisms of PDGF-BB on MSC migration and survival were explored in vitro. RESULTS The results showed that the biological effects of UTMD increased the local levels of stromal-derived factor-1 (SDF-1), which promoted the migration of transplanted MSCs to the ischemic region. Compared with UTMD alone, UTMD combined with PDGF-BB pretreatment significantly increased the cardiac homing of MSCs, which subsequently reduced myocardial apoptosis, promoted neovascularization and tissue repair, and increased cardiac function 30 days after MI. The vitro results demonstrated that PDGF-BB enhanced MSC migration and protected these cells from H2O2-induced apoptosis. Mechanistically, PDGF-BB pretreatment promoted MSC migration and inhibited H2O2-induced MSC apoptosis via activation of the phosphatidylinositol 3-kinase/serine-threonine kinase (PI3K/Akt) pathway. Furthermore, crosstalk between PDGF-BB and stromal-derived factor-1/chemokine receptor 4 (SDF-1/CXCR4) is involved in the PI3K/AKT signaling pathway. CONCLUSION The present study demonstrated that UTMD combined with PDGF-BB treatment could enhance the homing ability of MSCs, thus alleviating AMI in rats. Therefore, UTMD combined with PDGF-BB pretreatment may offer exciting therapeutic opportunities for strengthening MSC therapy in ischemic diseases.
Collapse
Grants
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 81701716; 82230066, 81922033, 81727805, 82171961, 81801715, 81801716 National Natural Science Foundation of China
- 2018CFB568 National Natural Science Foundation of Hubei
Collapse
Affiliation(s)
- Zhenxing Sun
- Anhui Medical University, Hefei, 230031, China
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yu Cai
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yihan Chen
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qiaofeng Jin
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Ziming Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Li Zhang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yuman Li
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Lei Huang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Jing Wang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Yali Yang
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Qing Lv
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China
| | - Zhengyang Han
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
- Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Mingxing Xie
- Department of Ultrasound Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Province Key Laboratory of Molecular Imaging, 430022, Wuhan, China.
| | - Xiangming Zhu
- Anhui Medical University, Hefei, 230031, China.
- Department of Ultrasound, The First Affiliated Hospital of Wannan Medical College, Wuhu, 241001, China.
| |
Collapse
|
5
|
Cheng L, Zhang D, Yan W. Ultrasound‑targeted microbubble destruction‑mediated overexpression of Sirtuin 3 inhibits the progression of ovarian cancer. Oncol Rep 2021; 46:220. [PMID: 34396428 PMCID: PMC8377464 DOI: 10.3892/or.2021.8171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 11/26/2020] [Indexed: 12/12/2022] Open
Abstract
Ultrasound-targeted microbubble destruction (UTMD) has recently been developed as a promising noninvasive tool for organ- and tissue-specific gene or drug delivery. The aim of the present study was to explore the role of UTMD-mediated Sirtuin 3 (SIRT3) overexpression in the malignant behaviors of human ovarian cancer (HOC) cells. Reverse transcription-quantitative PCR was performed to detect SIRT3 mRNA expression levels in normal human ovarian epithelial cells and HOC cell lines; low SIRT3 expression was found in HOC cell lines, and the SKOV3 cell line was used in the following experiments. The SIRT3-microbubble (MB) was prepared, and the effects of ultrasound-treated SIRT3-MB on biological processes of SKOV3 cells were determined. The proliferation, migration, invasion and apoptosis of SKOV3 cells were measured after SIRT3 upregulation by UTMD. Xenograft tumors in nude mice were induced to observe tumor growth in vivo. Upregulation of SIRT3 inhibited the malignant behaviors of SKOV3 cells, whereas UTMD-mediated SIRT3 upregulation further inhibited proliferation, epithelial-mesenchymal transition, invasion and migration, and induced apoptosis of SKOV3 cells, and it also inhibited tumor formation and growth in vivo. Moreover, the present study identified hypoxia inducible factor-1α (HIF-1α) as a target of SIRT3. The present study provided evidence that UTMD-mediated overexpression of SIRT3 may suppress HOC progression through the inhibition of HIF-1α.
Collapse
Affiliation(s)
- Li Cheng
- Department of Electrical Diagnosis, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Dongmei Zhang
- Department of Electrical Diagnosis, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130021, P.R. China
| | - Wei Yan
- Department of Electrical Diagnosis, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
6
|
Abstract
With the increasing insight into molecular mechanisms of cardiovascular disease, a promising solution involves directly delivering genes, cells, and chemicals to the infarcted myocardium or impaired endothelium. However, the limited delivery efficiency after administration fails to reach the therapeutic dose and the adverse off-target effect even causes serious safety concerns. Controlled drug release via external stimuli seems to be a promising method to overcome the drawbacks of conventional drug delivery systems (DDSs). Microbubbles and magnetic nanoparticles responding to ultrasound and magnetic fields respectively have been developed as an important component of novel DDSs. In particular, several attempts have also been made for the design and fabrication of dual-responsive DDS. This review presents the recent advances in the ultrasound and magnetic fields responsive DDSs in cardiovascular application, followed by their current problems and future reformation.
Collapse
|
7
|
Wei X, Zheng Y, Zhang W, Tan J, Zheng H. Ultrasound‑targeted microbubble destruction‑mediated Galectin‑7‑siRNA promotes the homing of bone marrow mesenchymal stem cells to alleviate acute myocardial infarction in rats. Int J Mol Med 2020; 47:677-687. [PMID: 33416139 PMCID: PMC7797467 DOI: 10.3892/ijmm.2020.4830] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 10/19/2020] [Indexed: 12/13/2022] Open
Abstract
Bone marrow mesenchymal stem cells (BMSCs) are accepted as a form of cellular therapy to improve cardiac function following acute myocardial infarction (AMI). The present study was performed to investigate the synergistic effect of ultrasound-targeted microbubble destruction (UTMD)-mediated Galectin-7-small interfering (si)RNA with the homing of BMSCs for AMI. The rat model of AMI was established, followed by identification of BMSCs. Rats with AMI received BMSC transplantation, BMSC transplantation + UTMD + siRNA negative control, or BMSC transplantation + UTMD + Galectin-7-siRNA. The cardiac function, hemodynamics indexes, degree of myocardial fiber injury and expression of apoptosis-related proteins in myocardial tissues of rats were detected. The homing of BMSCs was observed, and the indexes of myocardial microenvironment and the TGF-β/Smads pathway-related proteins in myocardial tissues were determined. AMI rats treated with UTMD-mediated Galectin-7-siRNA exhibited improved cardiac function and hemodynamics-related indices, decreased myocardial fiber injury and apoptotic cells, as well as enhanced homing ability of BMSCs, improved myocardial microenvironment, and suppressed TGF-β1/Smads pathway activation. In conclusion, the present study demonstrated that UTMD-mediated Galectin-7-siRNA treatment could enhance the homing ability of BMSCs, thus alleviating AMI in rats.
Collapse
Affiliation(s)
- Xin Wei
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Yan Zheng
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Weilin Zhang
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| | - Jing Tan
- Department of Cardiology, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Hong Zheng
- Department of Ultrasound, People's Hospital of Deyang City, Deyang, Sichuan 618000, P.R. China
| |
Collapse
|
8
|
Jang KW, Tu TW, Rosenblatt RB, Burks SR, Frank JA. MR-guided pulsed focused ultrasound improves mesenchymal stromal cell homing to the myocardium. J Cell Mol Med 2020; 24:13278-13288. [PMID: 33067927 PMCID: PMC7701528 DOI: 10.1111/jcmm.15944] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/28/2020] [Accepted: 09/14/2020] [Indexed: 12/26/2022] Open
Abstract
Image-guided pulsed focused ultrasound (pFUS) is a non-invasive technique that can increase tropism of intravenously (IV)-infused mesenchymal stromal cells (MSC) to sonicated tissues. MSC have shown promise for cardiac regenerative medicine strategies but can be hampered by inefficient homing to the myocardium. This study sonicated the left ventricles (LV) in rats with magnetic resonance imaging (MRI)-guided pFUS and examined both proteomic responses and subsequent MSC tropism to treated myocardium. T2-weighted MRI was used for pFUS targeting of the entire LV. pFUS increased numerous pro- and anti-inflammatory cytokines, chemokines, and trophic factors and cell adhesion molecules in the myocardial microenvironment for up to 48 hours post-sonication. Cardiac troponin I and N-terminal pro-B-type natriuretic peptide were elevated in the serum and myocardium. Immunohistochemistry revealed transient hypoxia and immune cell infiltration in pFUS-targeted regions. Myocardial tropism of IV-infused human MSC following pFUS increased twofold-threefold compared with controls. Proteomic and histological changes in myocardium following pFUS suggested a reversible inflammatory and hypoxic response leading to increased tropism of MSC. MR-guided pFUS could represent a non-invasive modality to improve MSC therapies for cardiac regenerative medicine approaches.
Collapse
Affiliation(s)
- Kee W Jang
- Frank Laboratory, Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD, USA.,Office of Product Evaluation and Quality, Center for Devices and Radiological Health, U.S. Food and Drug Administration, Silver Spring, MD, USA
| | - Tsang-Wei Tu
- Frank Laboratory, Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD, USA.,Molecular Imaging Laboratory, Department of Radiology, Howard University, Washington, DC, USA.,Center for Neuroscience and Regenerative Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Robert B Rosenblatt
- Frank Laboratory, Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD, USA
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, National Institutes of Health, Bethesda, MD, USA.,National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
9
|
Chen C, Geng L, Xu X, Kong W, Hou Y, Yao G, Feng X, Zhang H, Liang J. Comparative proteomics analysis of plasma protein in patients with neuropsychiatric systemic lupus erythematosus. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:579. [PMID: 32566606 PMCID: PMC7290550 DOI: 10.21037/atm.2020.04.58] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Background The aim of this study was to evaluate serum biomarkers of systemic lupus erythematous (SLE) patients, with and without neuropsychiatric (NP) manifestation by high-resolution proteomic analysis. Methods SLE patients with NP (NPSLE, n=9), SLE patients without NP (non-NPSLE, n=9) and healthy controls (HC, n=9) were prospectively enrolled in this study, and their plasma samples were collected and pooled into 3 NPSLE, 3 non-NPSLE and 3 HC samples for discovery profile. The TMT-LC-MS/MS-based proteomics approach was used to identify the differential proteome among the three matched groups, and the data were analyzed by bioinformatics tools, including Gene Ontology (GO) categories, Kyoto Encyclopedia of Genes and Genomes enrichment analysis, to explore canonical pathways and networks involved in the pathogenesis of NPSLE. To validation of differentially expressed proteomics results, four proteins were measured by ELISA. Results There were altogether 223 differentially expressed proteins in NPSLE groups compared with healthy controls (HC), of which 96 proteins increased while 127 proteins decreased. Compared with non-NPSLE, there were only 49 differentially expressed proteins in NPSLE groups, of which 37 proteins increased while 12 proteins decreased. The significantly changed pathway that those proteins are involved in was complement and coagulation cascades in NPSLE group compared with health controls. However, we didn't find significantly changed pathway between NPSLE group and non-NPSLE group. Five proteins were found significantly changed in all group-comparisons with consistent tendencies using Venn analysis, including Vitamin D binding protein (VDBP), C-reactive protein (CRP), KRT16, IGHV4-4 and CTRP3. Four proteins including CTRP3, VDBP, PAPPA and TRYP2 were selected to estimate the validity of the proteomics approach by ELISA. The expression levels of CTRP3 and TRYP2 were significantly changed in NPSLE patients compared with either HC or non-NPSLE patients. Conclusions Our research has successfully established serum protein profiles of NPSLE and non-NPSLE patients through TMT technology and screened out five proteins significantly changed in group-comparisons with consistent tendencies. The pathway of complement and coagulation cascades may participate in pathogenesis of NPSLE and non-NPSLE.
Collapse
Affiliation(s)
- Chen Chen
- Department of Clinical Nutrition, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Linyu Geng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xue Xu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Wei Kong
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Yayi Hou
- Institute of Brain Sciences, Medical School, Nanjing University, Nanjing 210093, China
| | - Genhong Yao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Xuebing Feng
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Huayong Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| | - Jun Liang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing 210008, China
| |
Collapse
|
10
|
Sun Z, Xie Y, Lee RJ, Chen Y, Jin Q, Lv Q, Wang J, Yang Y, Li Y, Cai Y, Wang R, Han Z, Zhang L, Xie M. Myocardium-targeted transplantation of PHD2 shRNA-modified bone mesenchymal stem cells through ultrasound-targeted microbubble destruction protects the heart from acute myocardial infarction. Theranostics 2020; 10:4967-4982. [PMID: 32308762 PMCID: PMC7163444 DOI: 10.7150/thno.43233] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Accepted: 03/22/2020] [Indexed: 01/09/2023] Open
Abstract
Ultrasound-targeted microbubble destruction (UTMD) is a promising approach to facilitate the precise delivery of bone marrow stem cells (BMSCs) to the ischemic myocardium. However, stem cell therapy for ischemic myocardium is challenging due to the poor survival of transplanted stem cells under severe ischemic conditions. In this study, we investigated whether myocardium-targeted transplantation of prolyl hydroxylase domain protein 2 (PHD2) shRNA-modified BMSCs by UTMD increases the viability of grafted cells, and enhances their cardioprotective effects in acute myocardial infarction. Methods: BMSCs were transduced with lentiviral PHD2 shRNA, and a novel microbubble formulation was prepared by a thin-film hydration method. In rats, BMSCs with or without PHD2 shRNA modification were transplanted by UTMD after inducing acute myocardium infarction. Effects of PHD2 shRNA on BMSC survival, myocardial apoptosis, angiogenesis, and cardiac function were evaluated. In vitro, anti-apoptotic effects and its mechanisms of PHD2 silencing on BMSC and BMSC-conditioned medium on H9C2 cell were detected. Results: PHD2 shRNA-modified BMSC transplantation by UTMD resulted in increased BMSC survival, reduced myocardial apoptosis, reduced infarct size, increased vascular density, and improved cardiac function compared to the control vector-modified BMSC transplantation by UTMD. PHD2 silencing increased BMSC survival through a HIF-1α-dependent mechanism. The decrease in cardiomyocyte apoptosis by conditioned medium from PHD2 shRNA-treated BMSCs was due to an increase in the expression of insulin-like growth factor (IGF)-1. Conclusions: The delivery of PHD2 shRNA-modified BMSCs by UTMD promoted grafted cell homing and activity, and increased myocardial angiogenesis in the infarcted heart, leading to improved cardiac function. This combination may provide a promising strategy for enhancing the effectiveness of stem cell therapy after acute myocardial infarction.
Collapse
|
11
|
Liu DD, Ullah M, Concepcion W, Dahl JJ, Thakor AS. The role of ultrasound in enhancing mesenchymal stromal cell-based therapies. Stem Cells Transl Med 2020; 9:850-866. [PMID: 32157802 PMCID: PMC7381806 DOI: 10.1002/sctm.19-0391] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 02/17/2020] [Indexed: 12/18/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) have been a popular platform for cell‐based therapy in regenerative medicine due to their propensity to home to damaged tissue and act as a repository of regenerative molecules that can promote tissue repair and exert immunomodulatory effects. Accordingly, a great deal of research has gone into optimizing MSC homing and increasing their secretion of therapeutic molecules. A variety of methods have been used to these ends, but one emerging technique gaining significant interest is the use of ultrasound. Sound waves exert mechanical pressure on cells, activating mechano‐transduction pathways and altering gene expression. Ultrasound has been applied both to cultured MSCs to modulate self‐renewal and differentiation, and to tissues‐of‐interest to make them a more attractive target for MSC homing. Here, we review the various applications of ultrasound to MSC‐based therapies, including low‐intensity pulsed ultrasound, pulsed focused ultrasound, and extracorporeal shockwave therapy, as well as the use of adjunctive therapies such as microbubbles. At a molecular level, it seems that ultrasound transiently generates a local gradient of cytokines, growth factors, and adhesion molecules that facilitate MSC homing. However, the molecular mechanisms underlying these methods are far from fully elucidated and may differ depending on the ultrasound parameters. We thus put forth minimal criteria for ultrasound parameter reporting, in order to ensure reproducibility of studies in the field. A deeper understanding of these mechanisms will enhance our ability to optimize this promising therapy to assist MSC‐based approaches in regenerative medicine.
Collapse
Affiliation(s)
- Daniel D Liu
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California
| | - Mujib Ullah
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California
| | - Waldo Concepcion
- Department of Surgery, Stanford University, Palo Alto, California
| | - Jeremy J Dahl
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California
| | - Avnesh S Thakor
- Interventional Regenerative Medicine and Imaging Laboratory, Department of Radiology, Stanford University, Palo Alto, California
| |
Collapse
|
12
|
Aydin O, Lorsung R, Chandran P, Cohen G, Burks SR, Frank JA. The Proteomic Effects of Pulsed Focused Ultrasound on Tumor Microenvironments of Murine Melanoma and Breast Cancer Models. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:3232-3245. [PMID: 31530419 PMCID: PMC7456468 DOI: 10.1016/j.ultrasmedbio.2019.08.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Revised: 06/18/2019] [Accepted: 08/20/2019] [Indexed: 05/08/2023]
Abstract
Non-ablative pulsed focused ultrasound (pFUS) targets non-thermal forces that activate local molecular and cellular immune responses. Optimal parameters to stimulate immunotherapeutic tumor microenvironments (TME) and responses in different tumor types remain uninvestigated. Flank B16 murine melanoma and 4T1 breast tumors received 1 MHz pFUS at 1-8 MPa peak negative pressures (PNP) and were analyzed 24 hr post-sonication. Necrosis or hemorrhage were unaltered in both tumors, but pFUS induced DNA strand breaks in tumor cells at PNP ≥6 MPa. pFUS at >4 MPa suppressed anti-inflammatory cytokines in B16 tumors. pFUS to 4T1 tumors decreased anti-inflammatory cytokines and increased pro-inflammatory cytokines and cell adhesion molecules. pFUS at 6 MPa increased calreticulin and alterations in check-point proteins along with tumoral and splenic immune cell changes that could be consistent with a shift towards an anti-TME. pFUS-induced TME alterations shows promise in generating anti-tumor immune responses, but non-uniform responses between tumor types require additional investigation to assess pFUS as a suitable anti-tumor therapy.
Collapse
Affiliation(s)
- Omer Aydin
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
- Erciyes University, School of Engineering, Department of Biomedical Engineering, 38039, Talas, Kayseri, Turkey
- To whom correspondence may be addressed. ;
| | - Rebecca Lorsung
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Parwathy Chandran
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Gadi Cohen
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Scott R. Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Joseph A. Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
- National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
- To whom correspondence may be addressed. ;
| |
Collapse
|
13
|
Sun T, Gao F, Li X, Cai Y, Bai M, Li F, Du L. A combination of ultrasound-targeted microbubble destruction with transplantation of bone marrow mesenchymal stem cells promotes recovery of acute liver injury. Stem Cell Res Ther 2018; 9:356. [PMID: 30594241 PMCID: PMC6311028 DOI: 10.1186/s13287-018-1098-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Revised: 10/29/2018] [Accepted: 12/02/2018] [Indexed: 12/13/2022] Open
Abstract
Background Bone marrow mesenchymal stem cells (BMSCs) can provide an additional source of therapeutic stem cells for regeneration of liver cells during acute liver injury (ALI). However, the insufficient hepatic homing by the transplanted BMSCs limits their applications. Ultrasound-targeted microbubble destruction (UTMD) has been reported to promote the homing of transplanted stem cells into the ischemic myocardium. In this study, we investigated whether UTMD promotes the hepatic homing of BMSCs in ALI rats and evaluated the therapeutic effect. Methods BMSCs were isolated from the femurs and tibias of Sprague-Dawley (SD) rats. The isolated BMSCs were stably transfected with a lentivirus expressing enhanced green fluorescent protein (EGFP) that can be visualized and quantified in vivo after transplantation. Both tumor necrosis factor α (TNF-α) and stromal cell-derived factor 1 (SDF-1) were used to verify the appropriate ultrasound parameters. The ALI rats were divided into four groups: control, BMSCs, UTMD, and UTMD + BMSCs. The protein and mRNA expression levels of SDF-1, intercellular cell adhesion molecule (ICAM-1), vascular cell adhesion molecule 1 (VCAM-1), hepatocyte growth factor (HGF), and monocyte chemotactic protein 1 (MCP-1) in the exposed livers were analyzed at 48 h after treatment. ALI recovery was determined by serum biochemical parameters and histology. Results The isolated rat BMSCs demonstrated a good proliferation potential that was both osteogenic and adipogenic in differentiation and expressed cluster of differentiation (CD) 29 and CD90, but not CD45 or CD11b/c. After BMSC and/or UTMD treatment, the number of GFP-labeled BMSCs in the UTMD + BMSCs group was significantly higher than that of the BMSCs group (9.8 ± 2.3 vs. 5.2 ± 1.1/per high-power field). Furthermore, the expression of GFP mRNA was performed for evaluation of the homing rate of BMSCs in injury sites as well. In addition, the expression levels of SDF-1, ICAM-1, VCAM-1, HGF, and MCP-1 were higher (p < 0.01) in UTMD+BMSCs group. The serum levels of biomarkers were significantly lower in the UTMD + BMSCs group, and the apoptotic rate of hepatocytes in the UTMD + BMSCs group was markedly lower than that of the BMSCs group (all p < 0.05). The hepatic pathology was significantly alleviated in the UTMD + BMSCs group. Conclusions UTMD treatment efficiently induced a favorable microenvironment for cell engraftment, resulting in improvement of hepatic homing of BMSCs, which was probably mediated through upregulation of the expression of adhesion molecules and cytokines. UTMD treatment appeared to be an effective and noninvasive approach to achieve better efficacy of BMSC-based therapy for repairing a severely injured liver. Electronic supplementary material The online version of this article (10.1186/s13287-018-1098-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Ting Sun
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Feng Gao
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Xin Li
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Yingyu Cai
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Min Bai
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Fan Li
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
| | - Lianfang Du
- Department of Medical Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
14
|
Qian L, Thapa B, Hong J, Zhang Y, Zhu M, Chu M, Yao J, Xu D. The present and future role of ultrasound targeted microbubble destruction in preclinical studies of cardiac gene therapy. J Thorac Dis 2018; 10:1099-1111. [PMID: 29607187 DOI: 10.21037/jtd.2018.01.101] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Multiple limitations for cardiac pharmacologic therapies like intolerance, individual variation in effectiveness, side effects, and high cost still remain, despite the recent progress in diagnosis and health support. Gene therapy is poised to be an attractive alternative in various ways for the future, refractory cardiac diseases being one aspect of it. As a novel therapy to deliver the objective gene to organs of living animals, ultrasound targeted microbubble destruction (UTMD) has therapeutic potential in cardiovascular disorders. UTMD, which binds microbubbles with DNA or RNA carriers into the shell and destroys the located microbubbles with low frequency and high mechanical index ultrasound can release target agents to specific organs. UTMD has the ability to transfect markedly through sonoporation, cavitation and other effects by way of intravenous injection that is minimally invasive and highly specific for gene deliverance. Here, we have summarized the present role of UTMD in pre-clinical studies of cardiac gene therapy which covers myocardial infarction, regeneration, ischaemia/reperfusion injury, hypertension, diabetic cardiomyopathy, adriamycin cardiomyopathy and some discussion for further studies.
Collapse
Affiliation(s)
- Lijun Qian
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Barsha Thapa
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jian Hong
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Yanmei Zhang
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Menglin Zhu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Ming Chu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Jing Yao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Di Xu
- Department of Geriatrics, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
15
|
Jang KW, Tu TW, Nagle ME, Lewis BK, Burks SR, Frank JA. Molecular and histological effects of MR-guided pulsed focused ultrasound to the rat heart. J Transl Med 2017; 15:252. [PMID: 29237455 PMCID: PMC5729396 DOI: 10.1186/s12967-017-1361-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2017] [Accepted: 12/06/2017] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Image-guided high intensity focused ultrasound has been used as an extracorporeal cardiac pacing tool and to enhance homing of stem cells to targeted tissues. However, molecular changes in the myocardium after sonication have not been widely investigated. Magnetic-resonance (MR)-guided pulsed focused ultrasound (pFUS) was targeted to the rat myocardium over a range of pressures and the microenvironmental and histological effects were evaluated over time. METHODS Eight-to-ten-week-old Sprague-Dawley rats received T2-weighted MR images to target pFUS to the left ventricular and septum without cardiac or respiratory gating. Rats were sonicated through the thoracic wall at peak negative pressures (PNP) from 1 to 8 MPa at a center frequency of 1 MHz, 10 ms pulse duration and 1 Hz pulse repetition frequency for 100 pulses per focal target. Following pFUS, myocardium was harvested over 24 h and subjected to imaging, proteomic, and histological measurements. RESULTS pFUS to the myocardium increased expression of cytokines, chemokines, and trophic factors characterized by an initial increase in tumor necrosis factor (TNF)-α followed by increases in pro- and anti-inflammatory factors that returned to baseline by 24 h. Immediately after pFUS, there was a transient (< 1 h) increase in N-terminal pro b-type natriuretic peptide (NT-proBNP) without elevation of other cardiac injury markers. A relationship between PNP and expression of TNF-α and NT-proBNP was observed with significant changes (p < 0.05 ANOVA) ≥ 4 MPa compared to untreated controls. Contrast-enhanced ex vivo T1-weighted MRI revealed vascular leakage in sonicated myocardium that was accompanied by the presence of albumin upon immunohistochemistry. Histology revealed infiltration of neutrophils and macrophages without morphological myofibril changes in sonicated tissue accompanied by pulmonary hemorrhage at PNP > 4 MPa. CONCLUSIONS MR-guided pFUS to myocardium induced transient proteomic and histological changes. The temporal proteomic changes in the myocardium indicate a short-lived sterile inflammatory response consistent with ischemia or contusion. Further study of myocardial function and strain is needed to determine if pFUS could be developed as an experimental model of cardiac injury and chest trauma.
Collapse
Affiliation(s)
- Kee W Jang
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Dr., Bethesda, MD, 20892, USA.
| | - Tsang-Wei Tu
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Matthew E Nagle
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Bobbi K Lewis
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Dr., Bethesda, MD, 20892, USA
| | - Joseph A Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, 10 Center Dr., Bethesda, MD, 20892, USA.,National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
16
|
Song Z, Wang Z, Shen J, Xu S, Hu Z. Nerve growth factor delivery by ultrasound-mediated nanobubble destruction as a treatment for acute spinal cord injury in rats. Int J Nanomedicine 2017; 12:1717-1729. [PMID: 28280337 PMCID: PMC5340249 DOI: 10.2147/ijn.s128848] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Background Spinal cord injuries (SCIs) can cause severe disability or death. Treatment options include surgical intervention, drug therapy, and stem cell transplantation. However, the efficacy of these methods for functional recovery remains unsatisfactory. Purpose This study was conducted to explore the effect of ultrasound (US)-mediated destruction of poly(lactic-co-glycolic acid) (PLGA) nanobubbles (NBs) expressing nerve growth factor (NGF) (NGF/PLGA NBs) on nerve regeneration in rats following SCI. Materials and methods Adult male Sprague Dawley rats were randomly divided into four treatment groups after Allen hit models of SCI were established. The groups were normal saline (NS) group, NGF and NBs group, NGF and US group, and NGF/PLGA NBs and US group. Histological changes after SCI were observed by hematoxylin and eosin staining. Neuron viability was determined by Nissl staining. Terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end labeling staining was used to examine cell apoptosis. NGF gene and protein expressions were detected by quantitative reverse transcription polymerase chain reaction and Western blotting. Green fluorescent protein expression in the spinal cord was examined using an inverted fluorescence microscope. The recovery of neural function was determined using the Basso, Beattie, and Bresnahan test. Results NGF therapy using US-mediated NGF/PLGA NBs destruction significantly increased NGF expression, attenuated histological injury, decreased neuron loss, inhibited neuronal apoptosis in injured spinal cords, and increased BBB scores in rats with SCI. Conclusion US-mediated NGF/PLGA NBs destruction effectively transfects the NGF gene into target tissues and has a significant effect on the injured spinal cord. The combination of US irradiation and gene therapy through NGF/PLGA NBs holds great promise for the future of nanomedicine and the development of noninvasive treatment options for SCI and other diseases.
Collapse
Affiliation(s)
- Zhaojun Song
- Department of Orthopedics, The First Affiliated Hospital
| | - Zhigang Wang
- Institution of Ultrasound Imaging, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, People's Republic of China
| | - Jieliang Shen
- Department of Orthopedics, The First Affiliated Hospital
| | - Shengxi Xu
- Department of Orthopedics, The First Affiliated Hospital
| | - Zhenming Hu
- Department of Orthopedics, The First Affiliated Hospital
| |
Collapse
|
17
|
Tang H, Zheng Y, Chen Y. Materials Chemistry of Nanoultrasonic Biomedicine. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2017; 29:1604105. [PMID: 27991697 DOI: 10.1002/adma.201604105] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 10/12/2016] [Indexed: 06/06/2023]
Abstract
As a special cross-disciplinary research frontier, nanoultrasonic biomedicine refers to the design and synthesis of nanomaterials to solve some critical issues of ultrasound (US)-based biomedicine. The concept of nanoultrasonic biomedicine can also overcome the drawbacks of traditional microbubbles and promote the generation of novel US-based contrast agents or synergistic agents for US theranostics. Here, we discuss the recent developments of material chemistry in advancing the nanoultrasonic biomedicine for diverse US-based bio-applications. We initially introduce the design principles of novel nanoplatforms for serving the nanoultrasonic biomedicine, from the viewpoint of synthetic material chemistry. Based on these principles and diverse US-based bio-application backgrounds, the representative proof-of-concept paradigms on this topic are clarified in detail, including nanodroplet vaporization for intelligent/responsive US imaging, multifunctional nano-contrast agents for US-based multi-modality imaging, activatable synergistic agents for US-based therapy, US-triggered on-demand drug releasing, US-enhanced gene transfection, US-based synergistic therapy on combating the cancer and potential toxicity issue of screening various nanosystems suitable for nanoultrasonic biomedicine. It is highly expected that this novel nanoultrasonic biomedicine and corresponding high performance in US imaging and therapy can significantly promote the generation of new sub-discipline of US-based biomedicine by rationally integrating material chemistry and theranostic nanomedicine with clinical US-based biomedicine.
Collapse
Affiliation(s)
- Hailin Tang
- Department of Diagnostic Ultrasound, Tongde Hospital of Zhejiang Province, Hangzhou, 310012, P. R. China
| | - Yuanyi Zheng
- Shanghai Institute of Ultrasound in Medicine, Shanghai Jiaotong University Affiliated, Shanghai Sixth People's Hospital, Shanghai, 200233, P. R. China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China
| |
Collapse
|
18
|
Ultrasound-targeted microbubble destruction enhances delayed BMC delivery and attenuates post-infarction cardiac remodelling by inducing engraftment signals. Clin Sci (Lond) 2016; 130:2105-2120. [PMID: 27609823 DOI: 10.1042/cs20160085] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 09/05/2016] [Indexed: 12/26/2022]
Abstract
Delayed administration of bone marrow cells (BMCs) at 2-4 weeks after successful reperfusion in patients with acute myocardial infarction (MI) does not improve cardiac function. The reduction in engraftment signals observed following this time interval might impair the effects of delayed BMC treatment. In the present study, we aimed to determine whether ultrasound-targeted microbubble destruction (UTMD) treatment could increase engraftment signals, enhance the delivery of delayed BMCs and subsequently attenuate post-infarction cardiac remodelling. A myocardial ischaemia/reperfusion (I/R) model was induced in Wistar rats via left coronary ligation for 45 min followed by reperfusion. Western blotting revealed that engraftment signals peaked at 7 days post-I/R and were dramatically lower at 14 days post-I/R. The lower engraftment signals at 14 days post-I/R could be triggered by UTMD treatment at a mechanical index of 1.0-1.9. The troponin I levels in the 1.9 mechanical index group were higher than in the other groups. Simultaneous haematoxylin and eosin staining and fluorescence revealed that the number of engrafted BMCs in the ischaemic zone was greater in the group treated with both UTMD and delayed BMC transplantation than in the control groups (P<0.05). Both UTMD and delayed BMC transplantation improved cardiac function and decreased cardiac fibrosis at 4 weeks after treatment, as compared with control groups (both P<0.05). Histopathology demonstrated that UTMD combined with delayed BMC transplantation increased capillary density, myocardial cell proliferation and c-kit+ cell proliferation. These findings indicated that UTMD treatment could induce engraftment signals and enhance homing of delayed BMCs to ischaemic myocardium, attenuating post-infarction cardiac remodelling by promoting neovascularization, cardiomyogenesis and expansion of cardiac c-kit+ cells.
Collapse
|
19
|
Santiesteban DY, Kubelick K, Dhada KS, Dumani D, Suggs L, Emelianov S. Monitoring/Imaging and Regenerative Agents for Enhancing Tissue Engineering Characterization and Therapies. Ann Biomed Eng 2016; 44:750-72. [PMID: 26692081 PMCID: PMC4956083 DOI: 10.1007/s10439-015-1509-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 11/11/2015] [Indexed: 01/07/2023]
Abstract
The past three decades have seen numerous advances in tissue engineering and regenerative medicine (TERM) therapies. However, despite the successes there is still much to be done before TERM therapies become commonplace in clinic. One of the main obstacles is the lack of knowledge regarding complex tissue engineering processes. Imaging strategies, in conjunction with exogenous contrast agents, can aid in this endeavor by assessing in vivo therapeutic progress. The ability to uncover real-time treatment progress will help shed light on the complex tissue engineering processes and lead to development of improved, adaptive treatments. More importantly, the utilized exogenous contrast agents can double as therapeutic agents. Proper use of these Monitoring/Imaging and Regenerative Agents (MIRAs) can help increase TERM therapy successes and allow for clinical translation. While other fields have exploited similar particles for combining diagnostics and therapy, MIRA research is still in its beginning stages with much of the current research being focused on imaging or therapeutic applications, separately. Advancing MIRA research will have numerous impacts on achieving clinical translations of TERM therapies. Therefore, it is our goal to highlight current MIRA progress and suggest future research that can lead to effective TERM treatments.
Collapse
Affiliation(s)
- Daniela Y Santiesteban
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, 1 University Station, C0800, Austin, TX, 78712, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Kelsey Kubelick
- School of Electrical and Computer Engineering, Georgia Institute of Technology, 777 Atlantic Drive NW, Atlanta, GA, 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Kabir S Dhada
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, 1 University Station, C0800, Austin, TX, 78712, USA
| | - Diego Dumani
- School of Electrical and Computer Engineering, Georgia Institute of Technology, 777 Atlantic Drive NW, Atlanta, GA, 30332, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Laura Suggs
- Department of Biomedical Engineering, University of Texas at Austin, 107 W. Dean Keeton, BME Building, 1 University Station, C0800, Austin, TX, 78712, USA.
| | - Stanislav Emelianov
- School of Electrical and Computer Engineering, Georgia Institute of Technology, 777 Atlantic Drive NW, Atlanta, GA, 30332, USA.
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Emory University School of Medicine, 313 Ferst Dr NW, Atlanta, GA, 30332, USA.
| |
Collapse
|
20
|
Tebebi PA, Burks SR, Kim SJ, Williams RA, Nguyen BA, Venkatesh P, Frenkel V, Frank JA. Cyclooxygenase-2 or tumor necrosis factor-α inhibitors attenuate the mechanotransductive effects of pulsed focused ultrasound to suppress mesenchymal stromal cell homing to healthy and dystrophic muscle. Stem Cells 2016; 33:1173-86. [PMID: 25534849 DOI: 10.1002/stem.1927] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Revised: 11/05/2014] [Accepted: 11/21/2014] [Indexed: 12/13/2022]
Abstract
Maximal homing of infused stem cells to diseased tissue is critical for regenerative medicine. Pulsed focused ultrasound (pFUS) is a clinically relevant platform to direct stem cell migration. Through mechanotransduction, pFUS establishes local gradients of cytokines, chemokines, trophic factors (CCTF) and cell adhesion molecules (CAM) in treated skeletal muscle that subsequently infused mesenchymal stromal cells (MSC) can capitalize to migrate into the parenchyma. Characterizing molecular responses to mechanical pFUS effects revealed tumor necrosis factor-alpha (TNFα) drives cyclooxygenase-2 (COX2) signaling to locally increase CCTF/CAM that are necessary for MSC homing. pFUS failed to increase chemoattractants and induce MSC homing to treated muscle in mice pretreated with ibuprofen (nonspecific COX inhibitor) or etanercept (TNFα inhibitor). pFUS-induced MSC homing was also suppressed in COX2-knockout mice, demonstrating ibuprofen blocked the mechanically induced CCTF/CAM by acting on COX2. Anti-inflammatory drugs, including ibuprofen, are administered to muscular dystrophy (MD) patients, and ibuprofen also suppressed pFUS-induced homing to muscle in a mouse model of MD. Drug interactions with cell therapies remain unexplored and are not controlled for during clinical cell therapy trials. This study highlights potentially negative drug-host interactions that suppress stem cell homing and could undermine cell-based approaches for regenerative medicine.
Collapse
Affiliation(s)
- Pamela A Tebebi
- Department of Biomedical Engineering, Catholic University of America, Washington, District of Columbia, USA; Frank Lab, Radiology and Imaging Sciences Department, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Burks SR, Nguyen BA, Tebebi PA, Kim SJ, Bresler MN, Ziadloo A, Street JM, Yuen PST, Star RA, Frank JA. Pulsed focused ultrasound pretreatment improves mesenchymal stromal cell efficacy in preventing and rescuing established acute kidney injury in mice. Stem Cells 2016; 33:1241-53. [PMID: 25640064 DOI: 10.1002/stem.1965] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 10/28/2014] [Accepted: 11/08/2014] [Indexed: 12/13/2022]
Abstract
Animal studies have shown that mesenchymal stromal cell (MSC) infusions improve acute kidney injury (AKI) outcomes when administered early after ischemic/reperfusion injury or within 24 hours after cisplatin administration. These findings have spurred several human clinical trials to prevent AKI. However, no specific therapy effectively treats clinically obvious AKI or rescues renal function once advanced injury is established. We investigated if noninvasive image-guided pulsed focused ultrasound (pFUS) could alter the kidney microenvironment to enhance homing of subsequently infused MSC. To examine the efficacy of pFUS-enhanced cell homing in disease, we targeted pFUS to kidneys to enhance MSC homing after cisplatin-induced AKI. We found that pFUS enhanced MSC homing at 1 day post-cisplatin, prior to renal functional deficits, and that enhanced homing improved outcomes of renal function, tubular cell death, and regeneration at 5 days post-cisplatin compared to MSC alone. We then investigated whether pFUS+MSC therapy could rescue established AKI. MSC alone at 3 days post-cisplatin, after renal functional deficits were obvious, significantly improved 7-day survival of animals. Survival was further improved by pFUS and MSC. pFUS prior to MSC injections increased IL-10 production by MSC that homed to kidneys and generated an anti-inflammatory immune cell profile in treated kidneys. This study shows pFUS is a neoadjuvant approach to improve MSC homing to diseased organs. pFUS with MSC better prevents AKI than MSC alone and allows rescue therapy in established AKI, which currently has no meaningful therapeutic options.
Collapse
Affiliation(s)
- Scott R Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA; Imaging Sciences Training Program, Clinical Center and National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Wang G, Zhuo Z, Yang B, Wu S, Xu Y, Liu Z, Tan K, Xia H, Wang X, Zou L, Gan L, Gao Y. Enhanced Homing Ability and Retention of Bone Marrow Stromal Cells to Diabetic Nephropathy by Microbubble-Mediated Diagnostic Ultrasound Irradiation. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:2977-2989. [PMID: 26318561 DOI: 10.1016/j.ultrasmedbio.2015.07.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 06/22/2015] [Accepted: 07/06/2015] [Indexed: 06/04/2023]
Abstract
Bone marrow stromal cell (BMSC) transplantation can successfully treat diabetic nephropathy (DN), but the lack of a specific homing place for intravenously injected cells limits the effective implementation of stem cell therapies. The migration and survival of transplanted BMSCs are determined by inflammatory reactions in the local kidney micro-environment. We tested the hypothesis that microbubble-mediated diagnostic ultrasound irradiation could provide a suitable micro-environment for BMSC delivery and retention in DN therapy. In this study, red fluorescent protein-labeled BMSCs were administered combined with microbubbles to streptozotocin-induced DN rats 4 wk after diabetes onset. We observed enhanced BMSC homing and retention in microbubble-mediated diagnostic ultrasound-irradiated kidneys compared with the contralateral kidneys on days 1 and 3 post-treatment. The results from immunohistochemical analysis, Western blot and enzyme-linked immunosorbent assay indicated that the local and transient expression of various chemo-attractants (i.e., cytokines, integrins and trophic factors) found to promote BMSC homing was much higher than observed in non-treated kidneys. The local capillary endothelium rupture observed by transmission electron microscopy may account for local micro-environment changes. Histopathologic analysis revealed no signs of kidney damage. These results confirmed that renal micro-environment changes caused by appropriate microbubble-mediated diagnostic ultrasound irradiation may promote BMSC homing ability to the diabetic kidney without renal toxicity and cell damage. This non-invasive and effective technique may be a promising method for BMSC transplantation therapy.
Collapse
Affiliation(s)
- Gong Wang
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zhongxiong Zhuo
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Bin Yang
- Department of Ultrasound Diagnostics, Jinling Hospital, Nanjing University School of Medicine, Nanjing, Jiangsu, China
| | - Shengzheng Wu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Zheng Liu
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Kaibin Tan
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Hongmei Xia
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Xiaoyan Wang
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Linru Zou
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Ling Gan
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China
| | - Yunhua Gao
- Department of Ultrasound, Xinqiao Hospital, The Third Military Medical University, Chongqing, China.
| |
Collapse
|
23
|
Liu Y, Li L, Su Q, Liu T, Ma Z, Yang H. Ultrasound-Targeted Microbubble Destruction Enhances Gene Expression of microRNA-21 in Swine Heart via Intracoronary Delivery. Echocardiography 2015; 32:1407-16. [PMID: 25613289 DOI: 10.1111/echo.12876] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Ultrasound-targeted microbubble destruction (UTMD) has proved to be a promising method for gene delivery. However, the feasibility and efficacy of UTMD-mediated gene delivery to the heart of large animals remain unclear. The present study was to explore the probability of increasing the transfection of microRNA-21 (miR-21) in swine heart by UTMD, and to search for the most suitable transfection conditions. METHODS We first optimized ultrasound intensity for successful miR-21 delivery. After intravenous injection of miR-21/microbubble mixture (miR-21/MB), transthoracic ultrasound irradiation (US) was applied from the left anterior chest using different intensities (1, 2, and 3 W/cm(2)). Then the efficacy of UTMD-mediated miR-21 delivery into myocardium via intracoronary injection was explored. Solution of miR-21/MB was infused intravenously or intracoronarily with US over the heart. Swine undergoing phosphate-buffered saline (PBS) injection, miR-21/MB injection via ear vein or coronary artery without US served as the control. The dynamic changes of left ventricular ejection fraction (LVEF) and serum troponin I (cTnI) after UTMD were detected, then the left ventricular myocardium was harvested for hematoxylin and eosin (H&E) staining 4 days later; the expression levels of miR-21 and programmed cell death 4 (PDCD4) were detected by quantitative real time polymerase chain reaction (qRT-PCR) and Western blot, respectively. RESULTS Results showed that pulse ultrasound at an intensity of 2 W/cm(2) and a 50% duty ratio for 20 minutes, there was no increase in serum cTnI, no histological sign of myocardial damage, and no noted cardiac dysfunction with relatively higher miR-21 expression (P < 0.05). Compared to miR-21/MB alone, UTMD significantly increased gene expression in myocardium regardless of the delivery routes (P < 0.05). Interestingly, the transfection efficiency was found to be a little bit higher with intracoronary injection than that with intravenous injection, though the dose for intracoronary injection was half of the intravenous injection (P < 0.05). CONCLUSION Under suitable conditions, UTMD can efficiently enhance gene expression in swine heart regardless of the delivery routes. The intravenous injection might be superior to intracoronary injection with less invasiveness and lower requirement of the technique. And for those undergoing percutaneous coronary intervention, intracoronary injection seems to be another alternative.
Collapse
Affiliation(s)
- Yangchun Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lang Li
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qiang Su
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Tao Liu
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhiying Ma
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Huafeng Yang
- Department of Cardiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
24
|
Zhang Y, Ye C, Xu Y, Dong X, Li J, Liu R, Gao Y. Ultrasound-mediated microbubble destruction increases renal interstitial capillary permeability in early diabetic nephropathy rats. ULTRASOUND IN MEDICINE & BIOLOGY 2014; 40:1273-1281. [PMID: 24613211 DOI: 10.1016/j.ultrasmedbio.2013.12.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2012] [Revised: 11/30/2013] [Accepted: 12/09/2013] [Indexed: 06/03/2023]
Abstract
Diabetic nephropathy (DN) is defined as persistent proteinuria corresponding to a urinary albumin excretion rate >300 μg/mg in the absence of other non-diabetic renal diseases. The aim of this study was to determine if ultrasound (US)-mediated microbubble (MB) destruction could increase renal interstitial capillary permeability in early DN rats. Diabetes was induced with streptozotocin. DN rats presented with mild micro-albuminuria 30 d after onset of diabetes. DN rats (N = 120) were divided into four groups that received Evans blue (EB) followed by: (i) no treatment (control group); (ii) continuous ultrasonic irradiation for 5 min (frequency = 7.00 MHz, mechanical index = 0.9, peak rarefactional pressure = 2.38 MPa: US group); (iii) microbubble injection (0.05 mL/kg: MB group); and (iv) both ultrasound and microbubble injection (US + MB group). Another 8 DN rats were subjected to ultrasound and microbubbles and then injected with EB after 24 h (recovery group). EB content, EB extravasation and E-selectin mRNA and protein expression significantly increased, and interstitial capillary walls became discontinuous in the US + MB group. Neither hemorrhage nor necrosis was observed on renal histology. Urine samples were collected 24 h post-treatment. There was no hematuria, and the urinary albumin excretion rate did not increase after ultrasound-microbubble interaction detected by urinalysis. EB content returned to the control group level after 24 h, as assessed for the recovery group. In conclusion, ultrasound-mediated microbubble destruction locally increased renal interstitial capillary permeability in DN rats, and should be considered a therapy for enhancing drug and gene delivery to the kidney in the future.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ultrasound, Xinqiao Hospital of the Third Military Medical University, Chongqing, China; Department of Ultrasound, Forty-Fourth Military Hospital, Guiyang, China
| | - Chuan Ye
- Department of Orthopaedics, Affiliated Hospital of Guiyang Medical College, Guiyang, China; Center for Tissue Engineering and Stem Cells, Guiyang Medical College, Guiyang, China
| | - Yali Xu
- Department of Ultrasound, Xinqiao Hospital of the Third Military Medical University, Chongqing, China
| | - Xuexin Dong
- Department of Ultrasound, Forty-Fourth Military Hospital, Guiyang, China
| | - Jianping Li
- Department of Ultrasound, Forty-Fourth Military Hospital, Guiyang, China
| | - Rong Liu
- Department of Ultrasound, Forty-Fourth Military Hospital, Guiyang, China
| | - Yunhua Gao
- Department of Ultrasound, Xinqiao Hospital of the Third Military Medical University, Chongqing, China.
| |
Collapse
|
25
|
New progress in angiogenesis therapy of cardiovascular disease by ultrasound targeted microbubble destruction. BIOMED RESEARCH INTERNATIONAL 2014; 2014:872984. [PMID: 24900995 PMCID: PMC4037580 DOI: 10.1155/2014/872984] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2014] [Accepted: 03/26/2014] [Indexed: 02/08/2023]
Abstract
Angiogenesis plays a vital part in the pathogenesis and treatment of cardiovascular disease and has become one of the hotspots that are being discussed in the past decades. At present, the promising angiogenesis therapies are gene therapy and stem cell therapy. Besides, a series of studies have shown that the ultrasound targeted microbubble destruction (UTMD) was a novel gene delivery system, due to its advantages of noninvasiveness, low immunogenicity and toxicity, repeatability and temporal and spatial target specificity; UTMD has also been used for angiogenesis therapy of cardiovascular disease. In this review, we mainly discuss the combination of UTMD and gene therapy or stem cell therapy which is applied in angiogenesis therapy in recent researches, and outline the future challenges and good prospects of these approaches.
Collapse
|
26
|
Tong J, Ding J, Shen X, Chen L, Bian Y, Ma G, Yao Y, Yang F. Mesenchymal stem cell transplantation enhancement in myocardial infarction rat model under ultrasound combined with nitric oxide microbubbles. PLoS One 2013; 8:e80186. [PMID: 24244646 PMCID: PMC3828189 DOI: 10.1371/journal.pone.0080186] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 09/30/2013] [Indexed: 12/29/2022] Open
Abstract
OBJECTIVE This study evaluated the effects of ultrasound combined with the homemade nitric oxide (NO) micro-bubble destruction on the in vitro proliferation, apoptosis, and migration of mesenchymal stem cells (MSCs). Furthermore, we studied whether or not irradiation of the NO micro-bubble combined with bone-marrow derived MSC infusion had a better effect on treating myocardial infarction. The possible mechanism of MSC delivery into the infarcted myocardium was also investigated. METHODS The murine bone marrow-derived MSCs were isolated, cultured, irradiated, and combined with different concentrations of NO microbubbles. MTT proliferation assay, annexin V-FITC apoptosis detection, migration assay, and RT-PCR were performed 24 h after the irradiation. The NO micro-bubbles was a intravenously injected, followed by the infusion of MSCs, which were labeled by CM-Dil. Myocardium was harvested 48 h later and the distribution of MSCs was observed by laser scanning confocal microscope after frozen sectioning. Echocardiography, histological examination, RT-PCR, and western blotting were performed four weeks after the cell transplantation. RESULTS Ultrasound combined with 1:70 NO micro-bubbles had no significant impact on the proliferation or apoptosis of MSCs. Transwell chamber findings demonstrated that MSCs migrated more efficiently in group that underwent ultrasound combined with 1:70 NO micro-bubbles. The Real-time PCR results indicated that the expression of CXCR4 was much higher in the group undergoing ultrasound combined with 1:70 NO micro-bubbles. The normalized fluorescence intensity greatly increased in the group of US+NO micro-bubbles and the cardiac function was also markedly improved. Immunohistochemical staining showed that the capillary density was much greater in the group of US+NO micro-bubbles as compared to that of the other groups. RT-PCR and western blotting also revealed a higher SDF-1 and VEGF expression in the group of US+NO micro-bubbles. CONCLUSIONS NO micro-bubbles could be used in the cell transplantation, which efficiently promoted the MSC homing into the infarcted myocardium.
Collapse
Affiliation(s)
- Jiayi Tong
- Institute of Cardiology, Southeast University, Nanjing, Jiangsu Province, China
| | - Jiandong Ding
- Institute of Cardiology, Southeast University, Nanjing, Jiangsu Province, China
| | - Xiangbo Shen
- Institute of Cardiology, Southeast University, Nanjing, Jiangsu Province, China
| | - Long Chen
- Institute of Cardiology, Southeast University, Nanjing, Jiangsu Province, China
| | - Yeping Bian
- Jiangsu Province Official Hospital, Nanjing, Jiangsu Province, China
| | - Genshan Ma
- Institute of Cardiology, Southeast University, Nanjing, Jiangsu Province, China
| | - Yuyu Yao
- Institute of Cardiology, Southeast University, Nanjing, Jiangsu Province, China
| | - Fang Yang
- Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu Province, China
| |
Collapse
|
27
|
Ling ZY, Shu SY, Zhong SG, Luo J, Su L, Liu ZZ, Lan XB, Yuan GB, Zheng YY, Ran HT, Wang ZG, Yin YH. Ultrasound targeted microbubble destruction promotes angiogenesis and heart function by inducing myocardial microenvironment change. ULTRASOUND IN MEDICINE & BIOLOGY 2013; 39:2001-2010. [PMID: 23969167 DOI: 10.1016/j.ultrasmedbio.2013.06.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2012] [Revised: 05/13/2013] [Accepted: 06/06/2013] [Indexed: 06/02/2023]
Abstract
The myocardial microenvironment plays a decisive role in the survival, migration and differentiation of stem cells. We studied myocardial micro-environmental changes induced by ultrasound-targeted microbubble destruction (UTMD) and their influence on the transplantation of mesenchymal stem cells (MSCs). Various intensities of ultrasound were applied to the anterior chest in canines with myocardial infarction after intravenous injection of microbubbles. The expression of cytokines and adhesion molecules in the infarcted area of the myocardium was detected after three sessions of UTMD in 1 wk. Real-time quantitative reverse transcription polymerase chain reaction (RTQ-PCR) showed that the expression of vascular cell adhesion molecule-1 (VCAM-1), stromal cell-derived factor-1 (SDF-1) and vascular endothelial growth factor (VEGF) in the 1.5 W/cm(2) and 1 W/cm(2) groups was markedly increased compared with the 0.5 W/cm(2) or the control groups (3.8- to 4.7-fold, p < 0.01), and the expression of interleukin-1β (IL-1β) in the 1.5 W/cm(2) group was increased twofold over the 1.0 W/cm(2) group, whereas the 0.5 W/cm(2) group experienced no significant changes. UTMD at 1.0 W/cm(2) was performed as previously described before mesenchymal stem cell (MSC) transplantation. Myocardial perfusion, angiogenesis and heart function were investigated before and 1 month after MSC transplantation. Coronary angiography and 99mTc-tetrofosmin scintigraphy revealed that myocardial perfusion was markedly improved after UTMD + MSCs treatment (p < 0.05). At echocardiographic analysis, heart function and the wall motion score index were significantly improved by UTMD + MSCs treatment compared with MSCs or UTMD alone and the control. In a canine model of myocardial infarction, therapeutic effects were markedly enhanced by MSC transplantation after the myocardial micro-environmental changes induced by UTMD; therefore, this novel method may be useful as an efficient approach for cellular therapy.
Collapse
Affiliation(s)
- Zhi-Yu Ling
- Department of Cardiology, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Burks SR, Ziadloo A, Kim SJ, Nguyen BA, Frank JA. Noninvasive pulsed focused ultrasound allows spatiotemporal control of targeted homing for multiple stem cell types in murine skeletal muscle and the magnitude of cell homing can be increased through repeated applications. Stem Cells 2013; 31:2551-60. [PMID: 23922277 PMCID: PMC3834159 DOI: 10.1002/stem.1495] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2013] [Revised: 06/18/2013] [Accepted: 07/05/2013] [Indexed: 12/13/2022]
Abstract
Stem cells are promising therapeutics for cardiovascular diseases, and i.v. injection is the most desirable route of administration clinically. Subsequent homing of exogenous stem cells to pathological loci is frequently required for therapeutic efficacy and is mediated by chemoattractants (cell adhesion molecules, cytokines, and growth factors). Homing processes are inefficient and depend on short-lived pathological inflammation that limits the window of opportunity for cell injections. Noninvasive pulsed focused ultrasound (pFUS), which emphasizes mechanical ultrasound-tissue interactions, can be precisely targeted in the body and is a promising approach to target and maximize stem cell delivery by stimulating chemoattractant expression in pFUS-treated tissue prior to cell infusions. We demonstrate that pFUS is nondestructive to murine skeletal muscle tissue (no necrosis, hemorrhage, or muscle stem cell activation) and initiates a largely M2-type macrophage response. We also demonstrate that local upregulation of chemoattractants in pFUS-treated skeletal muscle leads to enhance homing, permeability, and retention of human mesenchymal stem cells (MSC) and human endothelial precursor cells (EPC). Furthermore, the magnitude of MSC or EPC homing was increased when pFUS treatments and cell infusions were repeated daily. This study demonstrates that pFUS defines transient "molecular zip codes" of elevated chemoattractants in targeted muscle tissue, which effectively provides spatiotemporal control and tunability of the homing process for multiple stem cell types. pFUS is a clinically translatable modality that may ultimately improve homing efficiency and flexibility of cell therapies for cardiovascular diseases.
Collapse
Affiliation(s)
- Scott R. Burks
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
- Imaging Sciences Training Program, Clinical Center and National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD 20892
| | - Ali Ziadloo
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Saejeong J. Kim
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Ben A. Nguyen
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
| | - Joseph A. Frank
- Frank Laboratory, Radiology and Imaging Sciences, Clinical Center, National Institutes of Health, Bethesda, MD 20892
- Intramural Research Program, National Institute of Biomedical Imaging and Bioengineering, Bethesda, MD 20892
| |
Collapse
|
29
|
Kidney-targeted transplantation of mesenchymal stem cells by ultrasound-targeted microbubble destruction promotes kidney repair in diabetic nephropathy rats. BIOMED RESEARCH INTERNATIONAL 2013; 2013:526367. [PMID: 23762850 PMCID: PMC3677660 DOI: 10.1155/2013/526367] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 04/19/2013] [Accepted: 04/19/2013] [Indexed: 01/02/2023]
Abstract
We test the hypothesis that ultrasound-targeted microbubble destruction (UTMD) technique increases the renoprotective effect of kidney-targeted transplantation of bone-marrow-derived mesenchymal stem cells (BM-MSCs) in diabetic nephropathy (DN) rats. Diabetes was induced by streptozotocin injection (60 mg/Kg, intraperitoneally) in Sprague-Dawley rats. MSCs were administered alone or in combination with UTMD to DN rats at 4 weeks after diabetes onset. Random blood glucose concentrations were measured at 1, 2, 4, and 8 weeks, and plasma insulin levels, urinary albumin excretion rate (UAER) values, the structures of pancreas and kidney, the expressions of TGF- β 1, synaptopodin, and IL-10 were assessed at 8 weeks after MSCs transplantation. MSCs transplantation decreased blood glucose concentrations and attenuated pancreatic islets/ β cells damage. The permeability of renal interstitial capillaries and VCAM-1 expression increased after UTMD, which enhanced homing and retention of MSCs to kidneys. MSCs transplantation together with UTMD prevented renal damage and decreased UAER values by inhibiting TGF- β 1 expression and upregulating synaptopodin and IL-10 expression. We conclude that MSCs transplantation reverts hyperglycemia; UTMD technique noninvasively increases the homing of MSCs to kidneys and promotes renal repair in DN rats. This noninvasive cell delivery method may be feasible and efficient as a novel approach for personal MSCs therapy to diabetic nephropathy.
Collapse
|
30
|
Abstract
Time and space controlled drug delivery still remains a huge challenge in medicine. A novel approach that could offer a solution is ultrasound guided drug delivery. “Ultrasonic drug delivery” is often based on the use of small gas bubbles (so-called microbubbles) that oscillate and cavitate upon exposure to ultrasound waves. Some microbubbles are FDA approved contrast agents for ultrasound imaging and are nowadays widely investigated as promising drug carriers. Indeed, it has been observed that upon exposure to ultrasound waves, microbubbles may (a) release the encapsulated drugs and (b) simultaneously change the structure of the cell membranes in contact with the microbubbles which may facilitate drug entrance into cells. This review aims to highlight (a) major factors known so far which affect ultrasonic drug delivery (like the structure of the microbubbles, acoustic settings, etc.) and (b) summarizes the recent preclinical progress in this field together with a number of promising new concepts and applications.
Collapse
|
31
|
Rennert RC, Sorkin M, Garg RK, Gurtner GC. Stem cell recruitment after injury: lessons for regenerative medicine. Regen Med 2013; 7:833-50. [PMID: 23164083 DOI: 10.2217/rme.12.82] [Citation(s) in RCA: 116] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Tissue repair and regeneration are thought to involve resident cell proliferation as well as the selective recruitment of circulating stem and progenitor cell populations through complex signaling cascades. Many of these recruited cells originate from the bone marrow, and specific subpopulations of bone marrow cells have been isolated and used to augment adult tissue regeneration in preclinical models. Clinical studies of cell-based therapies have reported mixed results, however, and a variety of approaches to enhance the regenerative capacity of stem cell therapies are being developed based on emerging insights into the mechanisms of progenitor cell biology and recruitment following injury. This article discusses the function and mechanisms of recruitment of important bone marrow-derived stem and progenitor cell populations following injury, as well as the emerging therapeutic applications targeting these cells.
Collapse
Affiliation(s)
- Robert C Rennert
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic & Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, 257 Campus Drive West, Hagey Building GK-201, Stanford, CA 94305-5148, USA
| | | | | | | |
Collapse
|
32
|
Zhao YZ, Du LN, Lu CT, Jin YG, Ge SP. Potential and problems in ultrasound-responsive drug delivery systems. Int J Nanomedicine 2013; 8:1621-33. [PMID: 23637531 PMCID: PMC3635663 DOI: 10.2147/ijn.s43589] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ultrasound is an important local stimulus for triggering drug release at the target tissue. Ultrasound-responsive drug delivery systems (URDDS) have become an important research focus in targeted therapy. URDDS include many different formulations, such as microbubbles, nanobubbles, nanodroplets, liposomes, emulsions, and micelles. Drugs that can be loaded into URDDS include small molecules, biomacromolecules, and inorganic substances. Fields of clinical application include anticancer therapy, treatment of ischemic myocardium, induction of an immune response, cartilage tissue engineering, transdermal drug delivery, treatment of Huntington’s disease, thrombolysis, and disruption of the blood–brain barrier. This review focuses on recent advances in URDDS, and discusses their formulations, clinical application, and problems, as well as a perspective on their potential use in the future.
Collapse
Affiliation(s)
- Ying-Zheng Zhao
- Wenzhou Medical College, Wenzhou City, Zhejiang Province, People's Republic of China
| | | | | | | | | |
Collapse
|