1
|
Islam P, Abosalha A, Schaly S, Boyajian JL, Santos M, Makhlouf S, Renesteen E, Kassab A, Shum-Tim C, Shum-Tim D, Prakash S. Baculovirus Expressing Tumor Growth Factor-β1 (TGFβ1) Nanoshuttle Augments Therapeutic Effects for Vascular Wound Healing: Design and In Vitro Analysis. ACS Pharmacol Transl Sci 2024; 7:3419-3428. [PMID: 39539270 PMCID: PMC11555499 DOI: 10.1021/acsptsci.4c00509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/20/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
One of the major challenges in vascular tissue regeneration is effective wound healing that can be resolved by an innovative targeted nanoshuttle that delivers growth factors to blood vessels. This study investigates the production and efficacy of transforming growth factor-β1 (TGFβ1) gene delivery using poly(lactic-co-glycolic acid) (PLGA) baculovirus (BV) nanoshuttles (NSs). They exhibited an encapsulation efficiency of 86.23% ± 0.65% and a negative zeta potential of -29.57 ± 1.27 mV. In vitro studies in human umbilical vein endothelial cells (HUVECs) revealed that a 12 h incubation period optimized virus transduction. The safety and superior intracellular uptake of NSs and BVs in HUVECs were observed. The NSs carrying 100 and 400 MOI exhibited the highest cell proliferation rates in HUVECs. These sustained-release NSs significantly improved vascular cell migration and wound closure compared to free TGFβ1 carrying BV and can be a groundbreaking find in regenerative medicine, cardiovascular diseases, and chronic ulcer conditions.
Collapse
Affiliation(s)
- Paromita Islam
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Ahmed Abosalha
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
- Pharmaceutical
Technology Department, Faculty of Pharmacy, Tanta University, Tanta
Al-Geish St., the Medical Campus, Tanta 31527, Egypt
| | - Sabrina Schaly
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jacqueline L. Boyajian
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Madison Santos
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Stephanie Makhlouf
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Editha Renesteen
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Amal Kassab
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Cedrique Shum-Tim
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
- Mila−Quebec
AI Institute, McGill University, 6666 Saint-Urbain Street, Montreal, Quebec H2S 3H1, Canada
| | - Dominique Shum-Tim
- Division
of Cardiac Surgery, Royal Victoria Hospital, McGill University Health
Centre, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3G 2M1, Canada
| | - Satya Prakash
- Biomedical
Technology and Cell Therapy Research Laboratory, Department of Biomedical
Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec H3A 2B4, Canada
| |
Collapse
|
2
|
Choudhury P, Kandula N, Kosuru R, Adena SKR. Nanomedicine: A great boon for cardiac regenerative medicine. Eur J Pharmacol 2024; 982:176969. [PMID: 39218342 DOI: 10.1016/j.ejphar.2024.176969] [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: 04/14/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Cardiovascular disease (CVD) represents a significant global health challenge, remaining the leading cause of illness and mortality worldwide. The adult heart's limited regenerative capacity poses a major obstacle in repairing extensive damage caused by conditions like myocardial infarction. In response to these challenges, nanomedicine has emerged as a promising field aimed at improving treatment outcomes through innovative drug delivery strategies. Nanocarriers, such as nanoparticles (NPs), offer a revolutionary approach by facilitating targeted delivery of therapeutic agents directly to the heart. This precise delivery system holds immense potential for treating various cardiac conditions by addressing underlying mechanisms such as inflammation, oxidative stress, cell death, extracellular matrix remodeling, prosurvival signaling, and angiogenic pathways associated with ischemia-reperfusion injury. In this review, we provide a concise summary of the fundamental mechanisms involved in cardiac remodeling and regeneration. We explore how nanoparticle-based drug delivery systems can effectively target the afore-mentioned mechanisms. Furthermore, we discuss clinical trials that have utilized nanoparticle-based drug delivery systems specifically designed for cardiac applications. These trials demonstrate the potential of nanomedicine in clinical settings, paving the way for future advancements in cardiac therapeutics through precise and efficient drug delivery. Overall, nanomedicine holds promise in revolutionizing the treatment landscape of cardiovascular diseases by offering targeted and effective therapeutic strategies that address the complex pathophysiology of cardiac injuries.
Collapse
Affiliation(s)
- Priyanka Choudhury
- Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, Milwaukee, WI, 53226, USA
| | - Nirupama Kandula
- Department of Microbiology, GSL Medical College, Rajahmahendravaram, Andhra Pradesh, 533296, India
| | - Ramoji Kosuru
- Versiti Blood Research Institute, Milwaukee, WI, 53226, USA
| | - Sandeep Kumar Reddy Adena
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (Banaras Hindu University), Varanasi, Uttar Pradesh, 221005, India.
| |
Collapse
|
3
|
Zhang W, Qian S, Chen J, Jian T, Wang X, Zhu X, Dong Y, Fan G. Photo-Crosslinked Pro-Angiogenic Hydrogel Dressing for Wound Healing. Int J Mol Sci 2024; 25:9948. [PMID: 39337435 PMCID: PMC11432402 DOI: 10.3390/ijms25189948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Severe burns are one of the most devastating injuries, in which sustained inflammation and ischemia often delay the healing process. Pro-angiogenic growth factors such as vascular endothelial growth factor (VEGF) have been widely studied for promoting wound healing. However, the short half-life and instability of VEGF limit its clinical applications. In this study, we develop a photo-crosslinked hydrogel wound dressing from methacrylate hyaluronic acid (MeHA) bonded with a pro-angiogenic prominin-1-binding peptide (PR1P). The materials were extruded in wound bed and in situ formed a wound dressing via exposure to short-time ultraviolet radiation. The study shows that the PR1P-bonded hydrogel significantly improves VEGF recruitment, tubular formation, and cell migration in vitro. Swelling, Scanning Electron Microscope, and mechanical tests indicate the peptide does not affect the overall mechanical and physical properties of the hydrogels. For in vivo studies, the PR1P-bonded hydrogel dressing enhances neovascularization and accelerates wound closure in both deep second-degree burn and full-thickness excisional wound models. The Western blot assay shows such benefits can be related to the activation of the VEGF-Akt signaling pathway. These results suggest this photo-crosslinked hydrogel dressing efficiently promotes VEGF recruitment and angiogenesis in skin regeneration, indicating its potential for clinical applications in wound healing.
Collapse
Affiliation(s)
- Wang Zhang
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Shuyi Qian
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jia Chen
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai 201210, China
| | - Tianshen Jian
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai 201210, China
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xuechun Wang
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Xianmin Zhu
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai 201210, China
- Shanghai Academy of Sciences & Technology Institute of Model Animals Transformation, Shanghai 201203, China
| | - Yixiao Dong
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai 201210, China
- Shanghai Academy of Sciences & Technology Institute of Model Animals Transformation, Shanghai 201203, China
| | - Guoping Fan
- Shanghai Institute for Advanced Immunochemical Studies (SIAIS), ShanghaiTech University, Shanghai 201210, China; (W.Z.); (S.Q.); (J.C.); (T.J.); (X.W.); (X.Z.)
- Shanghai Clinical Research and Trial Center, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
4
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
5
|
Lacinski RA, Dziadowicz SA, Stewart A, Chaharbakhshi E, Akhter H, Pisquiy JJ, Victory JH, Hardham JB, Chew C, Prorock A, Bao Y, Sol-Church K, Hobbs GR, Klein E, Nalesnik MA, Hu G, de Oliveira A, Santiago SP, Lindsey BA. Nanosphere pharmacodynamics improves safety of immunostimulatory cytokine therapy. iScience 2024; 27:108836. [PMID: 38303687 PMCID: PMC10831265 DOI: 10.1016/j.isci.2024.108836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/04/2023] [Accepted: 01/03/2024] [Indexed: 02/03/2024] Open
Abstract
Systemic administration of interleukin (IL)-12 induces potent anti-tumor immune responses in preclinical cancer models through the systemic activation of effector immune cells and release of proinflammatory cytokines. IL-12-loaded PLGA nanospheres (IL12ns) are hypothesized to improve therapeutic efficacy and thwart unwanted side effects observed in previous human clinical trials. Through the investigation of peripheral blood and local tissue immune responses in healthy BALB/c mice, the immune-protective pharmacodynamics of IL12ns were suggested. Nanospheres increased pro-inflammatory plasma cytokines/chemokines (IFN-γ, IL-6, TNF-α, and CXCL10) without inducing maladaptive transcriptomic signatures in circulating peripheral immune cells. Gene expression profiling revealed activation of pro-inflammatory signaling pathways in systemic tissues, the likely source of these effector cytokines. These data support that nanosphere pharmacodynamics, including shielding IL-12 from circulating immune cells, depositing peripherally in systemic immune tissues, and then slowly eluting bioactive cytokine, thereafter, are essential to safe immunostimulatory therapy.
Collapse
Affiliation(s)
- Ryan A. Lacinski
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Sebastian A. Dziadowicz
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Amanda Stewart
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Chaharbakhshi
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Halima Akhter
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - John J. Pisquiy
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Jack H. Victory
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Joshua B. Hardham
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Claude Chew
- Advanced Technology Cores, Baylor College of Medicine, Houston, TX 77030, USA
| | - Alyson Prorock
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Yongde Bao
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Katia Sol-Church
- Genome Analysis & Technology Core, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Gerald R. Hobbs
- Department of Orthopaedics, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Edwin Klein
- Division of Laboratory Animal Resources, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260, USA
| | - Michael A. Nalesnik
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA 15260, USA
| | - Gangqing Hu
- Department of Microbiology, Immunology, and Cell Biology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
- Bioinformatics Core, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Ana de Oliveira
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA 22904, USA
| | - Stell P. Santiago
- Department of Pathology, West Virginia University School of Medicine, Morgantown, WV 26505, USA
| | - Brock A. Lindsey
- Department of Orthopaedic Surgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| |
Collapse
|
6
|
Dang-Luong PT, Nguyen HP, Le-Tuan L, Cao XT, Tran-Anh V, Quang HV. Nanocarrier systems loaded with IR780, iron oxide nanoparticles and chlorambucil for cancer theragnostics. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2024; 15:180-189. [PMID: 38352718 PMCID: PMC10862130 DOI: 10.3762/bjnano.15.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024]
Abstract
Theragnostics has become a popular term nowadays, since it enables both diagnosis and therapy at the same time while only using one carrier platform. Therefore, formulating a nanocarrier system that could serve as theragnostic agent by using simple techniques would be an advantage during production. In this project, we aimed to develop a nanocarrier that can be loaded with the chemotherapeutic medication chlorambucil and magnetic resonance imaging agents (e.g., iron oxide nanoparticles and near-infrared fluorophore IR780) for theragnostics. Poly(lactic-co-glycolic acid) was combined with the aforementioned ingredients to generate poly(vinyl alcohol)-based nanoparticles (NPs) using the single emulsion technique. Then the NPs were coated with F127 and F127-folate by simple incubation for five days. The nanoparticles have the hydrodynamic size of approx. 250 nm with negative charge. Similar to chlorambucil and IR780, iron oxide loadings were observed for all three kinds of NPs. The release of chlorambucil was quicker at pH 5.4 than at pH 7.4 at 37 °C. The F127@NPs and F127-folate@NPs demonstrated much greater cell uptake and toxicity up to 72 h after incubation. Our in vitro results of F127@NPs and F127-folate@NPs have demonstrated the ability of these systems to serve as medication and imaging agent carriers for cancer treatment and diagnostics, respectively.
Collapse
Affiliation(s)
| | - Hong-Phuc Nguyen
- NTT Hi-tech institute, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Loc Le-Tuan
- NTT Hi-tech institute, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| | - Xuan-Thang Cao
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 700000, Vietnam
| | - Vy Tran-Anh
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Vietnam
| | - Hieu Vu Quang
- Department of Biotechnology, NTT Hi-tech institute, Nguyen Tat Thanh University, Ho Chi Minh City, 700000, Vietnam
| |
Collapse
|
7
|
Beheshtizadeh N, Gharibshahian M, Bayati M, Maleki R, Strachan H, Doughty S, Tayebi L. Vascular endothelial growth factor (VEGF) delivery approaches in regenerative medicine. Biomed Pharmacother 2023; 166:115301. [PMID: 37562236 DOI: 10.1016/j.biopha.2023.115301] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/28/2023] [Accepted: 08/05/2023] [Indexed: 08/12/2023] Open
Abstract
The utilization of growth factors in the process of tissue regeneration has garnered significant interest and has been the subject of extensive research. However, despite the fervent efforts invested in recent clinical trials, a considerable number of these studies have produced outcomes that are deemed unsatisfactory. It is noteworthy that the trials that have yielded the most satisfactory outcomes have exhibited a shared characteristic, namely, the existence of a mechanism for the regulated administration of growth factors. Despite the extensive exploration of drug delivery vehicles and their efficacy in delivering certain growth factors, the development of a reliable predictive approach for the delivery of delicate growth factors like Vascular Endothelial Growth Factor (VEGF) remains elusive. VEGF plays a crucial role in promoting angiogenesis; however, the administration of VEGF demands a meticulous approach as it necessitates precise localization and transportation to a specific target tissue. This process requires prolonged and sustained exposure to a low concentration of VEGF. Inaccurate administration of drugs, either through off-target effects or inadequate delivery, may heighten the risk of adverse reactions and potentially result in tumorigenesis. At present, there is a scarcity of technologies available for the accurate encapsulation of VEGF and its subsequent sustained and controlled release. The objective of this review is to present and assess diverse categories of VEGF administration mechanisms. This paper examines various systems, including polymeric, liposomal, hydrogel, inorganic, polyplexes, and microfluidic, and evaluates the appropriate dosage of VEGF for multiple applications.
Collapse
Affiliation(s)
- Nima Beheshtizadeh
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Iran; Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran.
| | - Maliheh Gharibshahian
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran; Regenerative Medicine group (REMED), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Mohammad Bayati
- Department of Phytochemistry, Medicinal Plants and Drugs Research Institute, Shahid Beheshti University, Tehran, Iran
| | - Reza Maleki
- Department of Chemical Technologies, Iranian Research Organization for Science and Technology (IROST), P.O. Box 33535111, Tehran, Iran.
| | - Hannah Strachan
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Sarah Doughty
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI 53233, USA
| |
Collapse
|
8
|
Hydrogel-Based Tissue-Mimics for Vascular Regeneration and Tumor Angiogenesis. Regen Med 2023. [DOI: 10.1007/978-981-19-6008-6_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
|
9
|
Hoseinzadeh A, Ghoddusi Johari H, Anbardar MH, Tayebi L, Vafa E, Abbasi M, Vaez A, Golchin A, Amani AM, Jangjou A. Effective treatment of intractable diseases using nanoparticles to interfere with vascular supply and angiogenic process. Eur J Med Res 2022; 27:232. [PMID: 36333816 PMCID: PMC9636835 DOI: 10.1186/s40001-022-00833-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 09/30/2022] [Indexed: 11/06/2022] Open
Abstract
Angiogenesis is a vital biological process involving blood vessels forming from pre-existing vascular systems. This process contributes to various physiological activities, including embryonic development, hair growth, ovulation, menstruation, and the repair and regeneration of damaged tissue. On the other hand, it is essential in treating a wide range of pathological diseases, such as cardiovascular and ischemic diseases, rheumatoid arthritis, malignancies, ophthalmic and retinal diseases, and other chronic conditions. These diseases and disorders are frequently treated by regulating angiogenesis by utilizing a variety of pro-angiogenic or anti-angiogenic agents or molecules by stimulating or suppressing this complicated process, respectively. Nevertheless, many traditional angiogenic therapy techniques suffer from a lack of ability to achieve the intended therapeutic impact because of various constraints. These disadvantages include limited bioavailability, drug resistance, fast elimination, increased price, nonspecificity, and adverse effects. As a result, it is an excellent time for developing various pro- and anti-angiogenic substances that might circumvent the abovementioned restrictions, followed by their efficient use in treating disorders associated with angiogenesis. In recent years, significant progress has been made in different fields of medicine and biology, including therapeutic angiogenesis. Around the world, a multitude of research groups investigated several inorganic or organic nanoparticles (NPs) that had the potential to effectively modify the angiogenesis processes by either enhancing or suppressing the process. Many studies into the processes behind NP-mediated angiogenesis are well described. In this article, we also cover the application of NPs to encourage tissue vascularization as well as their angiogenic and anti-angiogenic effects in the treatment of several disorders, including bone regeneration, peripheral vascular disease, diabetic retinopathy, ischemic stroke, rheumatoid arthritis, post-ischemic cardiovascular injury, age-related macular degeneration, diabetic retinopathy, gene delivery-based angiogenic therapy, protein delivery-based angiogenic therapy, stem cell angiogenic therapy, and diabetic retinopathy, cancer that may benefit from the behavior of the nanostructures in the vascular system throughout the body. In addition, the accompanying difficulties and potential future applications of NPs in treating angiogenesis-related diseases and antiangiogenic therapies are discussed.
Collapse
Affiliation(s)
- Ahmad Hoseinzadeh
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamed Ghoddusi Johari
- Thoracic and Vascular Surgery Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Surgery, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, 53233, USA
| | - Ehsan Vafa
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Milad Abbasi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Golchin
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Clinical Biochemistry and Applied Cell Sciences, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Ali Mohammad Amani
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Jangjou
- Department of Emergency Medicine, School of Medicine, Namazi Teaching Hospital, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Tanaka M, Izumiya M, Haniu H, Ueda K, Ma C, Ueshiba K, Ideta H, Sobajima A, Uchiyama S, Takahashi J, Saito N. Current Methods in the Study of Nanomaterials for Bone Regeneration. NANOMATERIALS 2022; 12:nano12071195. [PMID: 35407313 PMCID: PMC9000656 DOI: 10.3390/nano12071195] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 03/21/2022] [Accepted: 03/30/2022] [Indexed: 12/18/2022]
Abstract
Nanomaterials show great promise as bone regeneration materials. They can be used as fillers to strengthen bone regeneration scaffolds, or employed in their natural form as carriers for drug delivery systems. A variety of experiments have been conducted to evaluate the osteogenic potential of bone regeneration materials. In vivo, such materials are commonly tested in animal bone defect models to assess their bone regeneration potential. From an ethical standpoint, however, animal experiments should be minimized. A standardized in vitro strategy for this purpose is desirable, but at present, the results of studies conducted under a wide variety of conditions have all been evaluated equally. This review will first briefly introduce several bone regeneration reports on nanomaterials and the nanosize-derived caveats of evaluations in such studies. Then, experimental techniques (in vivo and in vitro), types of cells, culture media, fetal bovine serum, and additives will be described, with specific examples of the risks of various culture conditions leading to erroneous conclusions in biomaterial analysis. We hope that this review will create a better understanding of the evaluation of biomaterials, including nanomaterials for bone regeneration, and lead to the development of versatile assessment methods that can be widely used in biomaterial development.
Collapse
Affiliation(s)
- Manabu Tanaka
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Makoto Izumiya
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Hisao Haniu
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
- Correspondence: (M.T.); (H.H.); Tel.: +81-266-23-8000 (M.T.); +81-263-37-3555 (H.H.)
| | - Katsuya Ueda
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Chuang Ma
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
| | - Koki Ueshiba
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
- Biomedical Engineering Division, Graduate School of Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan
| | - Hirokazu Ideta
- Biomedical Engineering Division, Graduate School of Medicine, Science and Technology, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan;
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Atsushi Sobajima
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
- Department of Orthopedics (Lower Limbs), Social Medical Care Corporation Hosei-kai Marunouchi Hospital, 1-7-45 Nagisa, Matsumoto, Nagano 390-8601, Japan
| | - Shigeharu Uchiyama
- Department of Orthopedic Surgery, Okaya City Hospital, 4-11-33 Honcho, Okaya, Nagano 394-8512, Japan;
| | - Jun Takahashi
- Department of Orthopedic Surgery, Shinshu University School of Medicine, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (A.S.); (J.T.)
| | - Naoto Saito
- Institute for Biomedical Sciences, Interdisciplinary Cluster for Cutting Edge Research, Shinshu University, 3-1-1 Asahi, Matsumoto, Nagano 390-8621, Japan; (M.I.); (K.U.); (C.M.); (K.U.); (N.S.)
| |
Collapse
|
11
|
Gonciar D, Mocan T, Agoston-Coldea L. Nanoparticles Targeting the Molecular Pathways of Heart Remodeling and Regeneration. Pharmaceutics 2022; 14:pharmaceutics14040711. [PMID: 35456545 PMCID: PMC9028351 DOI: 10.3390/pharmaceutics14040711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/13/2022] [Accepted: 03/22/2022] [Indexed: 12/10/2022] Open
Abstract
Cardiovascular diseases are the main cause of death worldwide, a trend that will continue to grow over the next decade. The heart consists of a complex cellular network based mainly on cardiomyocytes, but also on endothelial cells, smooth muscle cells, fibroblasts, and pericytes, which closely communicate through paracrine factors and direct contact. These interactions serve as valuable targets in understanding the phenomenon of heart remodeling and regeneration. The advances in nanomedicine in the controlled delivery of active pharmacological agents are remarkable and may provide substantial contribution to the treatment of heart diseases. This review aims to summarize the main mechanisms involved in cardiac remodeling and regeneration and how they have been applied in nanomedicine.
Collapse
Affiliation(s)
- Diana Gonciar
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| | - Teodora Mocan
- Physiology Department, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania
- Department of Nanomedicine, Regional Institute of Gastroenterology and Hepatology, Cluj-Napoca 400162, Romania
- Correspondence:
| | - Lucia Agoston-Coldea
- 2nd Department of Internal Medicine, Faculty of Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, Cluj-Napoca 400000, Romania; (D.G.); (L.A.-C.)
| |
Collapse
|
12
|
Messerschmidt VL, Chintapula U, Bonetesta F, Laboy-Segarra S, Naderi A, Nguyen KT, Cao H, Mager E, Lee J. In vivo Evaluation of Non-viral NICD Plasmid-Loaded PLGA Nanoparticles in Developing Zebrafish to Improve Cardiac Functions. Front Physiol 2022; 13:819767. [PMID: 35283767 PMCID: PMC8906778 DOI: 10.3389/fphys.2022.819767] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Accepted: 01/07/2022] [Indexed: 12/12/2022] Open
Abstract
In the era of the advanced nanomaterials, use of nanoparticles has been highlighted in biomedical research. However, the demonstration of DNA plasmid delivery with nanoparticles for in vivo gene delivery experiments must be carefully tested due to many possible issues, including toxicity. The purpose of the current study was to deliver a Notch Intracellular Domain (NICD)-encoded plasmid via poly(lactic-co-glycolic acid) (PLGA) nanoparticles and to investigate the toxic environmental side effects for an in vivo experiment. In addition, we demonstrated the target delivery to the endothelium, including the endocardial layer, which is challenging to manipulate gene expression for cardiac functions due to the beating heart and rapid blood pumping. For this study, we used a zebrafish animal model and exposed it to nanoparticles at varying concentrations to observe for specific malformations over time for toxic effects of PLGA nanoparticles as a delivery vehicle. Our nanoparticles caused significantly less malformations than the positive control, ZnO nanoparticles. Additionally, the NICD plasmid was successfully delivered by PLGA nanoparticles and significantly increased Notch signaling related genes. Furthermore, our image based deep-learning analysis approach evaluated that the antibody conjugated nanoparticles were successfully bound to the endocardium to overexpress Notch related genes and improve cardiac function such as ejection fraction, fractional shortening, and cardiac output. This research demonstrates that PLGA nanoparticle-mediated target delivery to upregulate Notch related genes which can be a potential therapeutic approach with minimum toxic effects.
Collapse
Affiliation(s)
- Victoria L Messerschmidt
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Fabrizio Bonetesta
- Department of Biological Sciences, University of North Texas, Denton, TX, United States
| | - Samantha Laboy-Segarra
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Amir Naderi
- Department of Electrical Engineering and Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hung Cao
- Department of Electrical Engineering and Computer Science, University of California, Irvine, Irvine, CA, United States
| | - Edward Mager
- Department of Biological Sciences, University of North Texas, Denton, TX, United States
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
13
|
Cakir SN, Whitehead KM, Hendricks HKL, de Castro Brás LE. Novel Techniques Targeting Fibroblasts after Ischemic Heart Injury. Cells 2022; 11:cells11030402. [PMID: 35159212 PMCID: PMC8834471 DOI: 10.3390/cells11030402] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 12/12/2022] Open
Abstract
The great plasticity of cardiac fibroblasts allows them to respond quickly to myocardial injury and to contribute to the subsequent cardiac remodeling. Being the most abundant cell type (in numbers) in the heart, and a key participant in the several phases of tissue healing, the cardiac fibroblast is an excellent target for treating cardiac diseases. The development of cardiac fibroblast-specific approaches have, however, been difficult due to the lack of cellular specific markers. The development of genetic lineage tracing tools and Cre-recombinant transgenics has led to a huge acceleration in cardiac fibroblast research. Additionally, the use of novel targeted delivery approaches like nanoparticles and modified adenoviruses, has allowed researchers to define the developmental origin of cardiac fibroblasts, elucidate their differentiation pathways, and functional mechanisms in cardiac injury and disease. In this review, we will first characterize the roles of fibroblasts in the different stages of cardiac repair and then examine novel techniques targeting fibroblasts post-ischemic heart injury.
Collapse
Affiliation(s)
- Sirin N Cakir
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Kaitlin M Whitehead
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Hanifah K L Hendricks
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| | - Lisandra E de Castro Brás
- Department of Physiology, The Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
14
|
Kerimoğlu O, Özer-Önder S, Alarçin E, Karsli S. Formulation and evaluation of the vascular endothelial growth factor loaded polycaprolactone nanoparticles. BRAZ J PHARM SCI 2022. [DOI: 10.1590/s2175-97902022e19660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
15
|
Weng T, Wang J, Yang M, Zhang W, Wu P, You C, Han C, Wang X. Nanomaterials for the delivery of bioactive factors to enhance angiogenesis of dermal substitutes during wound healing. BURNS & TRAUMA 2022; 10:tkab049. [PMID: 36960274 PMCID: PMC8944711 DOI: 10.1093/burnst/tkab049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 09/14/2021] [Indexed: 11/14/2022]
Abstract
Dermal substitutes provide a template for dermal regeneration and reconstruction. They constitutes an ideal clinical treatment for deep skin defects. However, rapid vascularization remains as a major hurdle to the development and application of dermal substitutes. Several bioactive factors play an important regulatory role in the process of angiogenesis and an understanding of the mechanism of achieving their effective delivery and sustained function is vital. Nanomaterials have great potential for tissue engineering. Effective delivery of bioactive factors (including growth factors, peptides and nucleic acids) by nanomaterials is of increasing research interest. This paper discusses the process of dermal substitute angiogenesis and the roles of related bioactive factors in this process. The application of nanomaterials for the delivery of bioactive factors to enhance angiogenesis and accelerate wound healing is also reviewed. We focus on new systems and approaches for delivering bioactive factors for enhancing angiogenesis in dermal substitutes.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Jialiang Wang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Min Yang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Wei Zhang
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Pan Wu
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Chuangang You
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | - Chunmao Han
- Department of Burns & Wound Care Centre, the Second Affiliated Hospital of Zhejiang University School of Medicine Hangzhou 310002, China
- Key Laboratory of The Diagnosis and Treatment of Severe Trauma and Burn of Zhejiang Province, Hangzhou 310002,China
| | | |
Collapse
|
16
|
Li T, Zhang T. The Application of Nanomaterials in Angiogenesis. Curr Stem Cell Res Ther 2021; 16:74-82. [PMID: 32066364 DOI: 10.2174/1574888x15666200211102203] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 12/16/2019] [Accepted: 01/07/2020] [Indexed: 02/08/2023]
Abstract
Induction of angiogenesis has enormous potential in the treatment of ischemic diseases and
the promotion of bulk tissue regeneration. However, the poor activity of angiogenic cells and proangiogenic
factors after transplantation is the main problem that imposes its wide applications. Recent
studies have found that the development of nanomaterials has solved this problem to some extent.
Nanomaterials can be mainly classified into inorganic nanomaterials represented by metals, metal oxides
and metal hydroxides, and organic nanomaterials including DNA tetrahedrons, graphene, graphene
oxide, and carbon nanotubes. These nanomaterials can induce the release of angiogenic factors
either directly or indirectly, thereby initiating a series of signaling pathways to induce angiogenesis.
Moreover, appropriate surface modifications of nanomaterial facilitate a variety of functions, such as
enhancing its biocompatibility and biostability. In clinical applications, nanomaterials can promote the
proliferation and differentiation of endothelial cells or mesenchymal stem cells, thereby promoting the
migration of hemangioblast cells to form new blood vessels. This review outlines the role of nanomaterials
in angiogenesis and is intended to provide new insights into the clinical treatment of systemic
and ischemic diseases.
Collapse
Affiliation(s)
- Tianle Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Tao Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
17
|
Therapeutic approaches targeting molecular signaling pathways common to diabetes, lung diseases and cancer. Adv Drug Deliv Rev 2021; 178:113918. [PMID: 34375681 DOI: 10.1016/j.addr.2021.113918] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 07/23/2021] [Accepted: 08/05/2021] [Indexed: 12/12/2022]
Abstract
Diabetes mellitus (DM), is the most common metabolic disease and is characterized by sustained hyperglycemia. Accumulating evidences supports a strong association between DM and numerous lung diseases including chronic obstructive pulmonary disease (COPD), fibrosis, and lung cancer (LC). The global incidence of DM-associated lung disorders is rising and several ongoing studies, including clinical trials, aim to elucidate the molecular mechanisms linking DM with lung disorders, in particular LC. Several potential mechanisms, including hyperglycemia, hyperinsulinemia, glycation, inflammation, and hypoxia, are cited as plausible links between DM and LC. In addition, studies also propose a connection between the use of anti-diabetic medications and reduction in the incidence of LC. However, the exact cause for DM associated lung diseases especially LC is not clear and is an area under intense investigation. Herein, we review the biological links reported between DM and lung disorders with an emphasis on LC. Furthermore, we report common signaling pathways (eg: TGF-β, IL-6, HIF-1, PDGF) and miRNAs that are dysregulated in DM and LC and serve as molecular targets for therapy. Finally, we propose a nanomedicine based approach for delivering therapeutics (eg: IL-24 plasmid DNA, HuR siRNA) to disrupt signaling pathways common to DM and LC and thus potentially treat DM-associated LC. Finally, we conclude that the effective modulation of commonly regulated signaling pathways would help design novel therapeutic protocols for treating DM patients diagnosed with LC.
Collapse
|
18
|
Messerschmidt VL, Chintapula U, Kuriakose AE, Laboy S, Truong TTD, Kydd LA, Jaworski J, Pan Z, Sadek H, Nguyen KT, Lee J. Notch Intracellular Domain Plasmid Delivery via Poly(Lactic-Co-Glycolic Acid) Nanoparticles to Upregulate Notch Pathway Molecules. Front Cardiovasc Med 2021; 8:707897. [PMID: 34651022 PMCID: PMC8507495 DOI: 10.3389/fcvm.2021.707897] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 08/20/2021] [Indexed: 12/24/2022] Open
Abstract
Notch signaling is a highly conserved signaling system that is required for embryonic development and regeneration of organs. When the signal is lost, maldevelopment occurs and leads to a lethal state. Delivering exogenous genetic materials encoding Notch into cells can reestablish downstream signaling and rescue cellular functions. In this study, we utilized the negatively charged and FDA approved polymer poly(lactic-co-glycolic acid) to encapsulate Notch Intracellular Domain-containing plasmid in nanoparticles. We show that primary human umbilical vein endothelial cells (HUVECs) readily uptake the nanoparticles with and without specific antibody targets. We demonstrated that our nanoparticles are non-toxic, stable over time, and compatible with blood. We further demonstrated that HUVECs could be successfully transfected with these nanoparticles in static and dynamic environments. Lastly, we elucidated that these nanoparticles could upregulate the downstream genes of Notch signaling, indicating that the payload was viable and successfully altered the genetic downstream effects.
Collapse
Affiliation(s)
- Victoria L Messerschmidt
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Aneetta E Kuriakose
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Samantha Laboy
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Thuy Thi Dang Truong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - LeNaiya A Kydd
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Justyn Jaworski
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States
| | - Zui Pan
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX, United States
| | - Hashem Sadek
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, United States.,Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|
19
|
Balakrishnan B. Role of Nanoscale Delivery Systems in Tissue Engineering. CURRENT PATHOBIOLOGY REPORTS 2021. [DOI: 10.1007/s40139-021-00225-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
20
|
Meng X, Xing Y, Li J, Deng C, Li Y, Ren X, Zhang D. Rebuilding the Vascular Network: In vivo and in vitro Approaches. Front Cell Dev Biol 2021; 9:639299. [PMID: 33968926 PMCID: PMC8097043 DOI: 10.3389/fcell.2021.639299] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Accepted: 03/29/2021] [Indexed: 12/25/2022] Open
Abstract
As the material transportation system of the human body, the vascular network carries the transportation of materials and nutrients. Currently, the construction of functional microvascular networks is an urgent requirement for the development of regenerative medicine and in vitro drug screening systems. How to construct organs with functional blood vessels is the focus and challenge of tissue engineering research. Here in this review article, we first introduced the basic characteristics of blood vessels in the body and the mechanism of angiogenesis in vivo, summarized the current methods of constructing tissue blood vessels in vitro and in vivo, and focused on comparing the functions, applications and advantages of constructing different types of vascular chips to generate blood vessels. Finally, the challenges and opportunities faced by the development of this field were discussed.
Collapse
Affiliation(s)
- Xiangfu Meng
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| | - Yunhui Xing
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Jiawen Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Cechuan Deng
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yifei Li
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Xi Ren
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, United States
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Sciences, Hubei University, Wuhan, China
| |
Collapse
|
21
|
Dal Sasso E, Zamuner A, Filippi A, Romanato F, Palmosi T, Vedovelli L, Gregori D, Gómez Ribelles JL, Russo T, Gloria A, Iop L, Gerosa G, Dettin M. Covalent functionalization of decellularized tissues accelerates endothelialization. Bioact Mater 2021; 6:3851-3864. [PMID: 33937589 PMCID: PMC8065253 DOI: 10.1016/j.bioactmat.2021.04.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 03/31/2021] [Accepted: 04/01/2021] [Indexed: 12/17/2022] Open
Abstract
In the field of tissue regeneration, the lack of a stable endothelial lining may affect the hemocompatibility of both synthetic and biological replacements. These drawbacks might be prevented by specific biomaterial functionalization to induce selective endothelial cell (EC) adhesion. Decellularized bovine pericardia and porcine aortas were selectively functionalized with a REDV tetrapeptide at 10−5 M and 10−6 M working concentrations. The scaffold-bound peptide was quantified and REDV potential EC adhesion enhancement was evaluated in vitro by static seeding of human umbilical vein ECs. The viable cells and MTS production were statistically higher in functionalized tissues than in control. Scaffold histoarchitecture, geometrical features, and mechanical properties were unaffected by peptide anchoring. The selective immobilization of REDV was effective in accelerating ECs adhesion while promoting proliferation in functionalized decellularized tissues intended for blood-contacting applications. Covalent functionalization of the decellularized tissues with REDV peptide accelerates endothelialization. New covalent grafting method not inducing collagen cross-linking. Measurements through two photon miscroscopy allow the quantification of biological matrix bound peptide. The decellularized tissues can be changed by chemical procedures to promote specific cellular behaviour with ECM preservation.
Collapse
Affiliation(s)
- Eleonora Dal Sasso
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Annj Zamuner
- Department of Industrial Engineering, University of Padua, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Andrea Filippi
- LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy.,Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, Italy.,Fondazione Bruno Kessler, Trento, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Filippo Romanato
- LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy.,Department of Physics and Astronomy "G. Galilei", University of Padua, Padua, Italy.,Institute of Pediatric Research Città della Speranza, Padua, Italy
| | - Tiziana Palmosi
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Luca Vedovelli
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - Dario Gregori
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy
| | - José Luís Gómez Ribelles
- Center for Biomaterials and Tissue Engineering, CBIT, Universitat Politècnica de València, València, Spain.,Biomedical Research Networking Center on Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Valencia, Spain
| | - Teresa Russo
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Antonio Gloria
- Institute of Polymers, Composites and Biomaterials, National Research Council of Italy, Naples, Italy
| | - Laura Iop
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Gino Gerosa
- Department of Cardiac, Thoracic and Vascular Sciences and Venetian Institute of Molecular Medicine, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| | - Monica Dettin
- Department of Industrial Engineering, University of Padua, Padua, Italy.,LIFELAB Program, Consorzio per la Ricerca Sanitaria, CORIS, Veneto Region, Italy
| |
Collapse
|
22
|
Pretorius D, Serpooshan V, Zhang J. Nano-Medicine in the Cardiovascular System. Front Pharmacol 2021; 12:640182. [PMID: 33746761 PMCID: PMC7969876 DOI: 10.3389/fphar.2021.640182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 01/19/2021] [Indexed: 01/19/2023] Open
Abstract
Nano-medicines that include nanoparticles, nanocomposites, small molecules, and exosomes represent new viable sources for future therapies for the dysfunction of cardiovascular system, as well as the other important organ systems. Nanomaterials possess special properties ranging from their intrinsic physicochemical properties, surface energy and surface topographies which can illicit advantageous cellular responses within the cardiovascular system, making them exceptionally valuable in future clinical translation applications. The success of nano-medicines as future cardiovascular theranostic agents requires a comprehensive understanding of the intersection between nanomaterial and the biomedical fields. In this review, we highlight some of the major types of nano-medicine systems that are currently being explored in the cardiac field. This review focusses on the major differences between the systems, and how these differences affect the specific therapeutic or diagnostic applications. The important concerns relevant to cardiac nano-medicines, including cellular responses, toxicity of the different nanomaterials, as well as cardio-protective and regenerative capabilities are discussed. In this review an overview of the current development of nano-medicines specific to the cardiac field is provided, discussing the diverse nature and applications of nanomaterials as therapeutic and diagnostic agents.
Collapse
Affiliation(s)
- Danielle Pretorius
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vahid Serpooshan
- Emory Children's Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Jianyi Zhang
- Department of Biomedical Engineering, School of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Biomedical Engineering, School of Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
23
|
Barui AK, Nethi SK, Basuthakur P, Jhelum P, Bollu VS, Reddy BR, Chakravarty S, Patra CR. Therapeutic angiogenesis using zinc oxide nanoflowers for the treatment of hind limb ischemia in rat model. Biomed Mater 2021; 16. [PMID: 33657534 DOI: 10.1088/1748-605x/abebd1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 03/03/2021] [Indexed: 11/11/2022]
Abstract
Critical limb ischemia (CLI) is considered as a severe type of peripheral artery diseases (PADs) which occurs due to the inadequate supply of blood to the limb extremities. CLI patients often suffer from extreme cramping pain, impaired wound healing, immobility, cardiovascular complications, amputation of the affected limb and even death. The conventional therapy for the treatment of CLI includes surgical revascularization as well as restoring angiogenesis using growth factor therapy. However, surgical revascularization is suitable for only a minor percentage of CLI patients and it is associated with high perioperative mortality rate. The use of growth factors is also limited in terms of their poor therapeutic angiogenesis potential as observed by the earlier clinical studies, which could be attributed to their poor bio-availability and non-specificity issues. Therefore, to outweigh the aforesaid disadvantages of the conventional strategies, there is an utmost need for the advancement of new alternative therapeutic biomaterials to treat CLI. Since past few decades, various research groups including ours have been involved in developing different pro-angiogenic nanomaterials. Among them, zinc oxide nanoflowers (ZONF), established in our laboratory, are considered as one of the potent nanoparticles to induce therapeutic angiogenesis. In our earlier studies, we have depicted that ZONF promote angiogenesis by inducing the formation of reactive oxygen species (ROS) and nitric oxide (NO) as well as activating Akt/MAPK/eNOS cell signaling pathways in the endothelial cells. Recently, we have also reported the therapeutic potential of ZONF to treat cerebral ischemia through their neuritogenic and neuroprotective properties, exploiting angio-neural cross talk. Considering the excellent pro-angiogenic properties of ZONF and importance of revascularization for the recovery of CLI, in this present study, we have comprehensively explored the therapeutic potential of ZONF in a rat hind limb ischemia model (established by ligating the femoral artery of hind limb), an animal model that mimics CLI in humans. The behavioural studies, laser Doppler perfusion imaging, histopathology, immunofluorescence as well as estimation of serum NO level depicted that the administration of ZONF could ameliorate the ischemic conditions in rats at a faster rate by promoting therapeutic angiogenesis to the ischemic sites. Altogether, the present study offers an alternative nanomedicine approach employing ZONF for the treatment of PADs.
Collapse
Affiliation(s)
- Ayan Kumar Barui
- Biomaterials Group, LST division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad-500007, Hyderabad, Telangana, 500007, INDIA
| | - Susheel Kumar Nethi
- Biomaterials Group, LST Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, AP, Hyderabad, Andhra Pradesh, 500007, INDIA
| | - Papia Basuthakur
- Biomaterials Group, LST Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, AP, Hyderabad, Andhra Pradesh, 500007, INDIA
| | - Priya Jhelum
- Biomaterials Group, LST Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, AP, Hyderabad, Andhra Pradesh, 500007, INDIA
| | - Vishnu Sravan Bollu
- Biomaterials Group, LST Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, AP, Hyderabad, Andhra Pradesh, 500007, INDIA
| | - Bommana Raghunath Reddy
- Biomaterials Group, LST Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, AP, Hyderabad, Andhra Pradesh, 500007, INDIA
| | - Sumana Chakravarty
- Biomaterials Group, LST Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, AP, Hyderabad, Andhra Pradesh, 500007, INDIA
| | - Chitta Ranjan Patra
- Biomaterials Group, LST Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, AP, Hyderabad, Andhra Pradesh, 500007, INDIA
| |
Collapse
|
24
|
Abstract
Nanotechnology could offer a new complementary strategy for the treatment of vascular diseases including coronary, carotid, or peripheral arterial disease due to narrowing or blockage of the artery caused by atherosclerosis. These arterial diseases manifest correspondingly as angina and myocardial infarction, stroke, and intermittent claudication of leg muscles during exercise. The pathogenesis of atherosclerosis involves biological events at the cellular and molecular level, thus targeting these using nanomaterials precisely and effectively could result in a better outcome. Nanotechnology can mitigate the pathological events by enhancing the therapeutic efficacy of the therapeutic agent by delivering it at the point of a lesion in a controlled and efficacious manner. Further, combining therapeutics with imaging will enhance the theranostic ability in atherosclerosis. Additionally, nanoparticles can provide a range of delivery systems for genes, proteins, cells, and drugs, which individually or in combination can address various problems within the arteries. Imaging studies combined with nanoparticles helps in evaluating the disease progression as well as the response to the treatment because imaging and diagnostic agents can be delivered precisely to the targeted destinations via nanocarriers. This review focuses on the use of nanotechnology in theranostics of coronary artery and peripheral arterial disease.
Collapse
|
25
|
Duan J, Chen Z, Liang X, Chen Y, Li H, Tian X, Zhang M, Wang X, Sun H, Kong D, Li Y, Yang J. Construction and application of therapeutic metal-polyphenol capsule for peripheral artery disease. Biomaterials 2020; 255:120199. [DOI: 10.1016/j.biomaterials.2020.120199] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/02/2020] [Accepted: 06/09/2020] [Indexed: 01/10/2023]
|
26
|
Omidi M, Almeida L, Tayebi L. Microfluidic-assisted fabrication of reverse micelle/PLGA hybrid microspheres for sustained vascular endothelial growth factor delivery. Biotechnol Appl Biochem 2020; 68:616-625. [PMID: 32533571 DOI: 10.1002/bab.1971] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Accepted: 06/06/2020] [Indexed: 12/17/2022]
Abstract
In this study, poly (d, l-lactide-co-glycolide) (PLGA) composite microspheres containing anhydrous reverse micelle (R.M.) dipalmitoylphosphatidylcholine (DPPC) nanoparticles loaded vascular endothelial growth factor (VEGF) were produced using microfluidic platforms. The VEGF-loaded R.M. nanoparticles (VRM) were achieved by initial self-assembly and subsequent lipid inversion of the DPPC vesicles. The fabricated VRMs were encapsulated into the PLGA matrix by flow-focusing geometry microfluidic platforms. The encapsulation efficiency, in vitro release profile, and the bioactivity of the produced composite microspheres were investigated. The release study showed that VEGF was slowly released from the PLGA composite microspheres over 28 days with a reduced initial burst (18 ± 4.17% in the first 24 H). The VEGF stability during encapsulation and release period was also investigated, and the results indicated that encapsulated VEGF was well preserved. Also, the bioactivity assay of the PLGA composite microspheres on human umbilical vein endothelial cells was confirmed that the encapsulated VEGF was utterly active. The present monodisperse and controllable VEGF-loaded microspheres with reproducible manner could be widely used in tissue engineering and therapeutic applications.
Collapse
Affiliation(s)
- Meisam Omidi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| | - Luis Almeida
- Marquette University School of Dentistry, Milwaukee, WI, USA
| | - Lobat Tayebi
- Marquette University School of Dentistry, Milwaukee, WI, USA
| |
Collapse
|
27
|
Harada A, Tsutsuki H, Zhang T, Lee R, Yahiro K, Sawa T, Niidome T. Preparation of Biodegradable PLGA-Nanoparticles Used for pH-Sensitive Intracellular Delivery of an Anti-inflammatory Bacterial Toxin to Macrophages. Chem Pharm Bull (Tokyo) 2020; 68:363-368. [PMID: 32238653 DOI: 10.1248/cpb.c19-00917] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Poly(D,L-lactide-co-glycolic) acid (PLGA) is a synthetic copolymer that has been used to design micro/nanoparticles as a carrier for macromolecules, such as protein and nucleic acids, that can be internalized by the endocytosis pathway. However, it is difficult to control the intracellular delivery to target organelles. Here we report an intracellular delivery system of nanoparticles modified with bacterial cytotoxins to the endoplasmic reticulum (ER) and anti-inflammatory activity of the nanoparticles. Subtilase cytotoxin (SubAB) is a bacterial toxin in certain enterohemorrhagic Escherichia coli (EHEC) strains that cleaves the host ER chaperone BiP and suppresses nuclear factor-kappaB (NF-κB) activation and nitric oxide (NO) generation in macrophages at sub-lethal concentration. PLGA-nanoparticles were modified with oligo histidine-tagged (6 × His-tagged) recombinant SubAB (SubAB-PLGA) through a pH-sensitive linkage, and their translocation to the ER in macrophage cell line J774.1 cells, effects on inducible NO synthase (iNOS), and levels of tumor necrosis factor (TNF)-α cytokine induced by lipopolysaccharide (LPS) were examined. Compared with free SubAB, SubAB-PLGA was significantly effective in BiP cleavage and the induction of the ER stress marker C/EBP homologous protein (CHOP) in J774.1 cells. Furthermore, SubAB-PLGA attenuated LPS-stimulated induction of iNOS and TNF-α. Our findings provide useful information for protein delivery to macrophages and may encourage therapeutic applications of nanoparticles to the treatment of inflammatory diseases.
Collapse
Affiliation(s)
- Ayaka Harada
- Faculty of Advanced Science and Technology, Kumamoto University
| | - Hiroyasu Tsutsuki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University
| | - Tianli Zhang
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University
| | - Ruda Lee
- International Research Organization for Advanced Science and Technology, Kumamoto University
| | - Kinnosuke Yahiro
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University
| | - Takuro Niidome
- Faculty of Advanced Science and Technology, Kumamoto University
| |
Collapse
|
28
|
Weng T, Wu P, Zhang W, Zheng Y, Li Q, Jin R, Chen H, You C, Guo S, Han C, Wang X. Regeneration of skin appendages and nerves: current status and further challenges. J Transl Med 2020; 18:53. [PMID: 32014004 PMCID: PMC6996190 DOI: 10.1186/s12967-020-02248-5] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Accepted: 01/28/2020] [Indexed: 12/14/2022] Open
Abstract
Tissue-engineered skin (TES), as an analogue of native skin, is promising for wound repair and regeneration. However, a major drawback of TES products is a lack of skin appendages and nerves to enhance skin healing, structural integrity and skin vitality. Skin appendages and nerves are important constituents for fully functional skin. To date, many studies have yielded remarkable results in the field of skin appendages reconstruction and nerve regeneration. However, patients often complain about a loss of skin sensation and even cutaneous chronic pain. Restoration of pain, temperature, and touch perceptions should now be a major challenge to solve in order to improve patients’ quality of life. Current strategies to create skin appendages and sensory nerve regeneration are mainly based on different types of seeding cells, scaffold materials, bioactive factors and involved signaling pathways. This article provides a comprehensive overview of different strategies for, and advances in, skin appendages and sensory nerve regeneration, which is an important issue in the field of tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tingting Weng
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Pan Wu
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Wei Zhang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Yurong Zheng
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou, 310022, China
| | - Qiong Li
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Ronghua Jin
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Haojiao Chen
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Chuangang You
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Songxue Guo
- Department of Plastic Surgery, Second Affiliated Hospital of Zhejiang University, Hangzhou, 310009, China
| | - Chunmao Han
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China
| | - Xingang Wang
- Department of Burns & Wound Care Center, Second Affiliated Hospital of Zhejiang University, College of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
29
|
O’Dwyer J, Murphy R, Dolan EB, Kovarova L, Pravda M, Velebny V, Heise A, Duffy GP, Cryan SA. Development of a nanomedicine-loaded hydrogel for sustained delivery of an angiogenic growth factor to the ischaemic myocardium. Drug Deliv Transl Res 2019; 10:440-454. [DOI: 10.1007/s13346-019-00684-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
30
|
Immobilization of Fibronectin-Loaded Polyelectrolyte Nanoparticles on Cardiovascular Material Surface to Improve the Biocompatibility. BIOMED RESEARCH INTERNATIONAL 2019; 2019:5478369. [PMID: 31781622 PMCID: PMC6875231 DOI: 10.1155/2019/5478369] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 09/23/2019] [Indexed: 01/02/2023]
Abstract
Vascular stent interventional therapy is the main method for clinical treatment of coronary artery diseases. However, due to the insufficient biocompatibility of cardiovascular materials, the implantation of stents often leads to serious adverse cardiac events. Surface biofunctional modification to improve the biocompatibility of vascular stents has been the focus of current research. In this study, based on the structure and function of extracellular matrix on vascular injury healing, a novel fibronectin-loaded poly-l-lysine/heparin nanoparticles was constructed for stent surface modification. In vitro blood compatibility evaluation results showed that the nanoparticles-modified surface could effectively reduce platelet adhesion and activation. In vitro cellular compatibility evaluation results indicated that the nanocoating may provide adequate efficacy in promoting the adhesion and proliferation of endothelial cells and thereby accelerate endothelialization. This study provides a new approach for the surface biological function modification of vascular stents.
Collapse
|
31
|
Rademakers T, Horvath JM, van Blitterswijk CA, LaPointe VL. Oxygen and nutrient delivery in tissue engineering: Approaches to graft vascularization. J Tissue Eng Regen Med 2019; 13:1815-1829. [PMID: 31310055 PMCID: PMC6852121 DOI: 10.1002/term.2932] [Citation(s) in RCA: 100] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 06/13/2019] [Accepted: 07/01/2019] [Indexed: 12/29/2022]
Abstract
The field of tissue engineering is making great strides in developing replacement tissue grafts for clinical use, marked by the rapid development of novel biomaterials, their improved integration with cells, better-directed growth and differentiation of cells, and improved three-dimensional tissue mass culturing. One major obstacle that remains, however, is the lack of graft vascularization, which in turn renders many grafts to fail upon clinical application. With that, graft vascularization has turned into one of the holy grails of tissue engineering, and for the majority of tissues, it will be imperative to achieve adequate vascularization if tissue graft implantation is to succeed. Many different approaches have been developed to induce or augment graft vascularization, both in vitro and in vivo. In this review, we highlight the importance of vascularization in tissue engineering and outline various approaches inspired by both biology and engineering to achieve and augment graft vascularization.
Collapse
Affiliation(s)
- Timo Rademakers
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Judith M. Horvath
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Clemens A. van Blitterswijk
- Complex Tissue Regeneration, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| | - Vanessa L.S. LaPointe
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology‐Inspired Regenerative MedicineMaastricht UniversityMaastrichtThe Netherlands
| |
Collapse
|
32
|
Yu C, Xing M, Sun S, Guan G, Wang L. In vitro evaluation of vascular endothelial cell behaviors on biomimetic vascular basement membranes. Colloids Surf B Biointerfaces 2019; 182:110381. [PMID: 31351274 DOI: 10.1016/j.colsurfb.2019.110381] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 07/15/2019] [Accepted: 07/18/2019] [Indexed: 12/14/2022]
Abstract
Vascular basement membrane (VBM) is a thin layer of fibrous extracellular matrix linking endothelium, and collagen type IV (COL IV) is its main composition. VBM plays a crucial role in anchoring down the endothelium to its loose connective tissue underneath. For vascular grafts, constructing biomimetic VBMs on the luminal surface is thus an effective approach to improve endothelialization in situ. In the present work, three types of polycaprolactone (PCL) membranes were produced and characterized through cell counting kit-8 (CCK-8) assay, adhesion force and elastic modulus test to examine the influence of fiber diameter and membrane composition on vascular endothelial cell (EC) behaviors. The PCL membranes with finer fibers of 54.77 nm (PCL-54) could biomimic the nanotopography of VBMs more efficiently than 544.64 nm (PCL-544), and they were more suitable for Pig iliac endothelium cells (PIECs) adhesion and proliferation, meanwhile, inducing higher elastic modulus and adhesion force of PIECs. On this foundation, we further immobilized COL IV onto PCL-54 (PCL-COL IV) to biomimic VBMs compositionally. Results showed that PIECs on PCL-COL IV exhibited the highest viability and proliferation. Besides, quantitative data indicated that the elastic modulus of the PIECs on PCL-COL IV (4441.00 Pa) was as two times higher than that on PCL-54 (2312.26 Pa), and the adhesion force grew to 1120.99 pN from 673.58 pN of PIECs on PCL-54. In summary, the PCL-COL IV membranes show high similarity with the native VBMs in terms of structure and composition, suggesting a promising potential for surface modification to vascular grafts for improved endothelialization.
Collapse
Affiliation(s)
- Chenglong Yu
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Meiyi Xing
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Shibo Sun
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China
| | - Guoping Guan
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China.
| | - Lu Wang
- Engineering Research Center of Technical Textile, Ministry of Education, Key Laboratory of Textile Science and Technology of Ministry of Education, Key Laboratory of Textile Industry for Biomedical Textile materials and Technology, College of Textiles, Donghua University, 2999 North Renmin Road, Shanghai, 201620, China.
| |
Collapse
|
33
|
Growth factor delivery: Defining the next generation platforms for tissue engineering. J Control Release 2019; 306:40-58. [DOI: 10.1016/j.jconrel.2019.05.028] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 05/15/2019] [Accepted: 05/17/2019] [Indexed: 12/14/2022]
|
34
|
Abstract
The ability to generate new microvessels in desired numbers and at desired locations has been a long-sought goal in vascular medicine, engineering, and biology. Historically, the need to revascularize ischemic tissues nonsurgically (so-called therapeutic vascularization) served as the main driving force for the development of new methods of vascular growth. More recently, vascularization of engineered tissues and the generation of vascularized microphysiological systems have provided additional targets for these methods, and have required adaptation of therapeutic vascularization to biomaterial scaffolds and to microscale devices. Three complementary strategies have been investigated to engineer microvasculature: angiogenesis (the sprouting of existing vessels), vasculogenesis (the coalescence of adult or progenitor cells into vessels), and microfluidics (the vascularization of scaffolds that possess the open geometry of microvascular networks). Over the past several decades, vascularization techniques have grown tremendously in sophistication, from the crude implantation of arteries into myocardial tunnels by Vineberg in the 1940s, to the current use of micropatterning techniques to control the exact shape and placement of vessels within a scaffold. This review provides a broad historical view of methods to engineer the microvasculature, and offers a common framework for organizing and analyzing the numerous studies in this area of tissue engineering and regenerative medicine. © 2019 American Physiological Society. Compr Physiol 9:1155-1212, 2019.
Collapse
Affiliation(s)
- Joe Tien
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
- Division of Materials Science and Engineering, Boston University, Brookline, Massachusetts, USA
| |
Collapse
|
35
|
Zhao N, Suzuki A, Zhang X, Shi P, Abune L, Coyne J, Jia H, Xiong N, Zhang G, Wang Y. Dual Aptamer-Functionalized in Situ Injectable Fibrin Hydrogel for Promotion of Angiogenesis via Codelivery of Vascular Endothelial Growth Factor and Platelet-Derived Growth Factor-BB. ACS APPLIED MATERIALS & INTERFACES 2019; 11:18123-18132. [PMID: 31026135 PMCID: PMC6542593 DOI: 10.1021/acsami.9b02462] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
In situ injectable hydrogels hold great potential for in vivo applications such as drug delivery and regenerative medicine. However, it is challenging to ensure stable sequestration and sustained release of loaded biomolecules in these hydrogels. As aptamers have high binding affinities and specificities against target biomolecules, we studied the capability of aptamers in functionalizing in situ injectable fibrin (Fn) hydrogels for in vivo delivery of two growth factors including vascular endothelial growth factor (VEGF) and platelet-derived growth factor-BB (PDGF-BB). The results show that aptamer-functionalized fibrinogen (Fg) could form in situ injectable Fn hydrogels with porous structures. The aptamer-functionalized Fn hydrogels could sequester more VEGF and PDGF-BB than the native Fn and release these growth factors in a sustained manner with high bioactivity. After the aptamer-functionalized Fn hydrogels were subcutaneously injected into mice, the codelivery of VEGF and PDGF-BB could promote the growth of mature blood vessels. Therefore, this study has successfully demonstrated that aptamer-functionalized in situ injectable hydrogels hold great potential for in vivo codelivery of multiple growth factors and promotion of angiogenesis .
Collapse
Affiliation(s)
- Nan Zhao
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Akiho Suzuki
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Xiaolong Zhang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Peng Shi
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Lidya Abune
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - James Coyne
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Huizhen Jia
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| | - Na Xiong
- Department of Veterinary and Biomedical Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| | - Ge Zhang
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA
| | - Yong Wang
- Department of Biomedical Engineering, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
36
|
Bietenbeck M, Engel S, Lamping S, Hansen U, Faber C, Ravoo BJ, Yilmaz A. Functionalization of Clinically Approved MRI Contrast Agents for the Delivery of VEGF. Bioconjug Chem 2019; 30:1042-1047. [PMID: 30860371 DOI: 10.1021/acs.bioconjchem.9b00142] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In combining the two clinically approved substances ferumoxytol and VEGF-165 via peptide coupling, we propose a straightforward approach to obtain a potentially ready-to-use theranostic contrast agent for specific cardiovascular diseases. Clinical and preclinical magnetic resonance imaging (MRI) studies have shown that intravenously applied superparamagnetic ferumoxytol nanoparticles accumulate in acute ischemic myocardial tissue. On the other hand, growth factors such as VEGF-165 (vascular endothelial growth factor) play a major role during angiogenesis and vasculogenesis. Promising clinical studies with systemic application of VEGF-165 have been performed in the past. However, following untargeted systemic application, the biological half-life of VEGF-165 was too short to develop its full effect. Therefore, we hypothesized that ferumoxytol particles functionalized with VEGF-165 will accumulate in ischemic myocardial regions and can be detected by MRI, while the prolonged retention of VEGF-165 due to ferumoxytol-coupling will help to prevent adverse tissue remodeling. In addition, strategies such as magnetic targeting can be used to enhance targeted local accumulation. As a precondition for further preclinical research, we confirmed the successful coupling between ferumoxytol and VEGF-165 in detail (TEM, XPS, and IR spectroscopy), characterized the functionalized ferumoxytol particles (DLS, TEM, and MRI) and performed in vitro tests that showed their superior effect on cell growth and survival.
Collapse
Affiliation(s)
- Michael Bietenbeck
- Division of Cardiovascular Imaging, Department of Cardiology I , University Hospital Münster , Albert-Schweitzer-Campus 1 , 48149 Münster , Germany
| | - Sabrina Engel
- Organic Chemistry Institute and Center for Soft Nanoscience , Westfälische Wilhelms-Universität Münster , Corrensstrasse 40 , 48149 Münster , Germany
| | - Sebastian Lamping
- Organic Chemistry Institute and Center for Soft Nanoscience , Westfälische Wilhelms-Universität Münster , Corrensstrasse 40 , 48149 Münster , Germany
| | - Uwe Hansen
- Institute of Musculoskeletal Medicine , University Hospital Münster , Domagkstraße 3 , 48149 Münster , Germany
| | - Cornelius Faber
- Translational Research Imaging Center, Department of Clinical Radiology , University Hospital Münster , Albert-Schweitzer-Campus 1 , 48149 Münster , Germany
| | - Bart Jan Ravoo
- Organic Chemistry Institute and Center for Soft Nanoscience , Westfälische Wilhelms-Universität Münster , Corrensstrasse 40 , 48149 Münster , Germany
| | - Ali Yilmaz
- Division of Cardiovascular Imaging, Department of Cardiology I , University Hospital Münster , Albert-Schweitzer-Campus 1 , 48149 Münster , Germany
| |
Collapse
|
37
|
Gonciar D, Mocan T, Matea CT, Zdrehus C, Mosteanu O, Mocan L, Pop T. Nanotechnology in metastatic cancer treatment: Current Achievements and Future Research Trends. J Cancer 2019; 10:1358-1369. [PMID: 31031845 PMCID: PMC6485233 DOI: 10.7150/jca.28394] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 01/04/2019] [Indexed: 12/20/2022] Open
Abstract
The systemic spread of malignant cells from a primary site, a process termed metastasis represents a global challenge in cancer treatment. There is a real need to develop novel therapy strategies and nanomedicine may have remarkable and valuable contribution through specific and selective delivery of chemotherapeutic agents, through its intrinsic cytotoxic activity or through imaging applications, appealing also in the context of cancer personalized therapy. This review is focused on the applications of nanoparticles in the treatment of metastatic cancer, particularly on the possible effect on cell stabilization, growth inhibition, eventual interaction with adhesion molecules and antiangiogenic effect.
Collapse
Affiliation(s)
- Diana Gonciar
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Teodora Mocan
- Physiology Department, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Cristian Tudor Matea
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Claudiu Zdrehus
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Ofelia Mosteanu
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Lucian Mocan
- Third Surgery Clinic, "Iuliu Hatieganu" University of Medicine and Pharmacy Cluj-Napoca, Romania.,Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| | - Teodora Pop
- Nanomedicine Department, Regional Institute of Gastroenterology and Hepatology "Prof.Dr. Octavian Fodor" Cluj-Napoca , Romania
| |
Collapse
|
38
|
Ding D, Zhu Q. Recent advances of PLGA micro/nanoparticles for the delivery of biomacromolecular therapeutics. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2018; 92:1041-1060. [DOI: 10.1016/j.msec.2017.12.036] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 12/18/2017] [Accepted: 12/30/2017] [Indexed: 01/06/2023]
|
39
|
Noukeu LC, Wolf J, Yuan B, Banerjee S, Nguyen KT. Nanoparticles for Detection and Treatment of Peripheral Arterial Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800644. [PMID: 29952061 DOI: 10.1002/smll.201800644] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Indexed: 06/08/2023]
Abstract
Peripheral arterial disease (PAD) is defined as a slow, progressive disorder of the lower extremity arterial vessels characterized by chronic narrowing that often results in occlusion and is associated with loss of functional capacity. Although the PAD occurrence rate is increasing in the elderly population, outcomes with current treatment strategies are suboptimal. Hence, there is an urgent need to develop new technologies that overcome limitations of traditional modalities for PAD detection and therapy. In this Review, the application of nanotechnology as a tool that bridges the gap in PAD diagnosis and therapy is in focus. Several materials including synthetic, natural, biodegradable, and biocompatible materials are used to develop nanoparticles for PAD diagnostic and/or therapeutic applications. Moreover, various recent research approaches are being explored to diagnose PAD through multimodality imaging with different nanoplatforms. Further efforts include targeted delivery of various therapeutic agents using nanostructures as carriers to treat PAD. Last, but not least, despite being a fairly new field, researchers are exploring the use of nanotheranostics for PAD detection and therapy.
Collapse
Affiliation(s)
- Linda C Noukeu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| | - Joseph Wolf
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| | - Baohong Yuan
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| | - Subhash Banerjee
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| |
Collapse
|
40
|
Czekanska EM, Geng J, Glinka M, White K, Kanczler J, Evans ND, Oreffo ROC, Bradley M. Combinatorial delivery of bioactive molecules by a nanoparticle-decorated and functionalized biodegradable scaffold. J Mater Chem B 2018; 6:4437-4445. [PMID: 32254661 DOI: 10.1039/c8tb00474a] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The combination of supportive biomaterials and bioactive factors to stimulate endogenous progenitor cells is of key interest for the treatment of conditions in which intrinsic bone healing capacities are compromised. To address this need a "scaffold-decoration platform" was developed in which a biocompatible, biotin-functionalised 3D structural polymer network was generated through a solvent blending process, and used to recruit avidin modified nanoparticles within its 3D structure through biotin-avidin conjugation. This was enabled via the generation of a suite of poly(lactic-co-glycolic acid) (PLGA) nanoparticles, encapsulating two bioactive factors, vascular endothelial growth factor (VEGF) and l-ascorbic acid 2-phosphate (AA2P) and conjugated to streptavidin to allow attachment to the bone generating scaffold. The levels of encapsulated and released VEGF and AA2P were tailored to fall within the desired range to promote biological activity as confirmed by an increase in endothelial cell tubule formation and collagen production by osteoblast cells in response to nanoparticle release of VEGF and AA2P, respectively. The release of VEGF from the scaffolds produced a significant effect on vasculature development within the chick chorioallantoic membrane (CAM) angiogenic assay. Similarly, the scaffolds showed strong biological effects in ex vivo assays indicating the potential of this platform for localised delivery of bioactive molecules with applications in both hard and soft tissue engineering.
Collapse
Affiliation(s)
- Ewa M Czekanska
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Faculty of Medicine, Southampton University, Southampton, SO16 6YD, UK.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Zhu W, Cui H, Boualam B, Masood F, Flynn E, Rao RD, Zhang ZY, Zhang LG. 3D bioprinting mesenchymal stem cell-laden construct with core-shell nanospheres for cartilage tissue engineering. NANOTECHNOLOGY 2018; 29:185101. [PMID: 29446757 DOI: 10.1088/1361-6528/aaafa1] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Cartilage tissue is prone to degradation and has little capacity for self-healing due to its avascularity. Tissue engineering, which provides artificial scaffolds to repair injured tissues, is a novel and promising strategy for cartilage repair. 3D bioprinting offers even greater potential for repairing degenerative tissue by simultaneously integrating living cells, biomaterials, and biological cues to provide a customized scaffold. With regard to cell selection, mesenchymal stem cells (MSCs) hold great capacity for differentiating into a variety of cell types, including chondrocytes, and could therefore be utilized as a cartilage cell source in 3D bioprinting. In the present study, we utilize a tabletop stereolithography-based 3D bioprinter for a novel cell-laden cartilage tissue construct fabrication. Printable resin is composed of 10% gelatin methacrylate (GelMA) base, various concentrations of polyethylene glycol diacrylate (PEGDA), biocompatible photoinitiator, and transforming growth factor beta 1 (TGF-β1) embedded nanospheres fabricated via a core-shell electrospraying technique. We find that the addition of PEGDA into GelMA hydrogel greatly improves the printing resolution. Compressive testing shows that modulus of the bioprinted scaffolds proportionally increases with the concentrations of PEGDA, while swelling ratio decreases with the increase of PEGDA concentration. Confocal microscopy images illustrate that the cells and nanospheres are evenly distributed throughout the entire bioprinted construct. Cells grown on 5%/10% (PEGDA/GelMA) hydrogel present the highest cell viability and proliferation rate. The TGF-β1 embedded in nanospheres can keep a sustained release up to 21 d and improve chondrogenic differentiation of encapsulated MSCs. The cell-laden bioprinted cartilage constructs with TGF-β1-containing nanospheres is a promising strategy for cartilage regeneration.
Collapse
Affiliation(s)
- Wei Zhu
- Department of Mechanical and Aerospace Engineering, The George Washington University, Washington DC 20052, United States of America
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Chereddy KK, Vandermeulen G, Préat V. PLGA based drug delivery systems: Promising carriers for wound healing activity. Wound Repair Regen 2018; 24:223-36. [PMID: 26749322 DOI: 10.1111/wrr.12404] [Citation(s) in RCA: 122] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2015] [Accepted: 12/19/2015] [Indexed: 01/10/2023]
Abstract
Wound treatment remains one of the most prevalent and economically burdensome healthcare issues in the world. Current treatment options are limited and require repeated administrations which led to the development of new therapeutics to satisfy the unmet clinical needs. Many potent wound healing agents were discovered but most of them are fragile and/or sensitive to in vivo conditions. Poly(lactic-co-glycolic acid) (PLGA) is a widely used biodegradable polymer approved by food and drug administration and European medicines agency as an excipient for parenteral administrations. It is a well-established drug delivery system in various medical applications. The aim of the current review is to elaborate the applications of PLGA based drug delivery systems carrying different wound healing agents and also present PLGA itself as a wound healing promoter. PLGA carriers encapsulating drugs such as antibiotics, anti-inflammatory drugs, proteins/peptides, and nucleic acids targeting various phases/signaling cycles of wound healing, are discussed with examples. The combined therapeutic effects of PLGA and a loaded drug on wound healing are also mentioned.
Collapse
Affiliation(s)
- Kiran Kumar Chereddy
- Catholic University of Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Gaëlle Vandermeulen
- Catholic University of Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| | - Véronique Préat
- Catholic University of Louvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Brussels, Belgium
| |
Collapse
|
43
|
Desmet CM, Préat V, Gallez B. Nanomedicines and gene therapy for the delivery of growth factors to improve perfusion and oxygenation in wound healing. Adv Drug Deliv Rev 2018; 129:262-284. [PMID: 29448035 DOI: 10.1016/j.addr.2018.02.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Revised: 01/25/2018] [Accepted: 02/03/2018] [Indexed: 12/16/2022]
Abstract
Oxygen plays a key role in wound healing, and hypoxia is a major cause of wound healing impairment; therefore, treatments to improve hemodynamics and increase wound oxygenation are of particular interest for the treatment of chronic wounds. This article describes the roles of oxygen and angiogenesis in wound healing as well as the tools used to evaluate tissue oxygenation and perfusion and then presents a review of nanomedicines and gene therapies designed to improve perfusion and oxygenation and accelerate wound healing.
Collapse
|
44
|
Molybdenum cluster loaded PLGA nanoparticles: An innovative theranostic approach for the treatment of ovarian cancer. Eur J Pharm Biopharm 2018; 125:95-105. [DOI: 10.1016/j.ejpb.2018.01.007] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 12/01/2017] [Accepted: 01/09/2018] [Indexed: 12/30/2022]
|
45
|
Nakhlband A, Eskandani M, Omidi Y, Saeedi N, Ghaffari S, Barar J, Garjani A. Combating atherosclerosis with targeted nanomedicines: recent advances and future prospective. ACTA ACUST UNITED AC 2018; 8:59-75. [PMID: 29713603 PMCID: PMC5915710 DOI: 10.15171/bi.2018.08] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 02/21/2018] [Indexed: 12/19/2022]
Abstract
![]()
Introduction:
Cardiovascular diseases (CVDs) is recognized as the leading cause of mortality worldwide. The increasing prevalence of such disease demands novel therapeutic and diagnostic approaches to overcome associated clinical/social issues. Recent advances in nanotechnology and biological sciences have provided intriguing insights to employ targeted Nanomachines to the desired location as imaging, diagnosis, and therapeutic modalities. Nanomedicines as novel tools for enhanced drug delivery, imaging, and diagnosis strategies have shown great promise to combat cardiovascular diseases.
Methods:
In the current study, we intend to review the most recent studies on the nano-based strategies for improved management of CVDs.
Results:
A cascade of events results in the formation of atheromatous plaque and arterial stenosis. Furthermore, recent studies have shown that nanomedicines have displayed unique functionalities and provided de novo applications in the diagnosis and treatment of atherosclerosis.
Conclusion:
Despite some limitations, nanomedicines hold considerable potential in the prevention, diagnosis, and treatment of various ailments including atherosclerosis. Fewer side effects, amenable physicochemical properties and multi-potential application of such nano-systems are recognized through various investigations. Therefore, it is strongly believed that with targeted drug delivery to atherosclerotic lesions and plaque, management of onset and progression of disease would be more efficient than classical treatment modalities.
Collapse
Affiliation(s)
- Ailar Nakhlband
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Eskandani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Yadollah Omidi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nazli Saeedi
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samad Ghaffari
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jaleh Barar
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Pharmaceutics, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Alireza Garjani
- Research Center for Pharmaceutical Nanotechnology, Biomedicine Institute, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of pharmacology, Faculty of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
46
|
Wang B, Lv X, Chen S, Li Z, Yao J, Peng X, Feng C, Xu Y, Wang H. Use of heparinized bacterial cellulose based scaffold for improving angiogenesis in tissue regeneration. Carbohydr Polym 2018; 181:948-956. [DOI: 10.1016/j.carbpol.2017.11.055] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2017] [Revised: 11/15/2017] [Accepted: 11/15/2017] [Indexed: 12/21/2022]
|
47
|
Nam M, Lee J, Lee KY, Kim J. Sequential Targeted Delivery of Liposomes to Ischemic Tissues by Controlling Blood Vessel Permeability. ACS Biomater Sci Eng 2018; 4:532-538. [PMID: 33418742 DOI: 10.1021/acsbiomaterials.7b00815] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Delivery systems for therapeutic angiogenesis that deliver angiogenic factors to ischemic tissues have recently been fabricated. However, these systems are designed for surgical implantation or multiple local injections which can cause pain and potential physical burden in patients. Here, we propose a minimally invasive sequential nanoparticle-mediated delivery strategy for ischemic tissue using a murine hindlimb ischemic model. Intravenously injected liposomes that encapsulate VEGF, an angiogenic factor, first target the ischemic sites via the enhanced permeability and retention (EPR) effect in early stages of ischemia. VEGF released from the targeted liposomes maintains the blood vessel permeability for a longer period of time compared to the delivery of empty liposomes. This first nanoparticle-mediated delivery of VEGF to the ischemic site enables extending the temporal window of leaky blood vessel up to 7 days so that the second liposomes could be targeted to the ischemic sites via EPR effect. This strategy will provide opportunities for the targeted delivery of other vessel maturation agents loaded in nanoparticles to ischemic tissue.
Collapse
Affiliation(s)
- Myungjoo Nam
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| | - Jangwook Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Kuen Yong Lee
- Department of Bioengineering, Hanyang University, Seoul 04763, Republic of Korea
| | - Jaeyun Kim
- School of Chemical Engineering, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea.,Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea.,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea
| |
Collapse
|
48
|
Sharma A, Vaghasiya K, Gupta P, Gupta UD, Verma RK. Reclaiming hijacked phagosomes: Hybrid nano-in-micro encapsulated MIAP peptide ensures host directed therapy by specifically augmenting phagosome-maturation and apoptosis in TB infected macrophage cells. Int J Pharm 2018; 536:50-62. [DOI: 10.1016/j.ijpharm.2017.11.046] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/04/2017] [Accepted: 11/21/2017] [Indexed: 12/15/2022]
|
49
|
Wang Z, Jia Z, Jiang Y, Li P, Han L, Lu X, Ren F, Wang K, Yuan H. Mussel-inspired nano-building block assemblies for mimicking extracellular matrix microenvironments with multiple functions. Biofabrication 2017; 9:035005. [DOI: 10.1088/1758-5090/aa7fdc] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
50
|
Chen Y, Wu Y, Gao J, Zhang Z, Wang L, Chen X, Mi J, Yao Y, Guan D, Chen B, Dai J. Transdermal Vascular Endothelial Growth Factor Delivery with Surface Engineered Gold Nanoparticles. ACS APPLIED MATERIALS & INTERFACES 2017; 9:5173-5180. [PMID: 28112909 DOI: 10.1021/acsami.6b15914] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Skin injuries caused by burns or radiation remain a serious concern in terms of clinical therapy. Because of the damage to the epidermis or dermis, angiogenesis is needed to repair the skin. Vascular endothelial growth factor (VEGF) is one of the most effective factors for promoting angiogenesis and preventing injury progression, but the delivery of VEGF to lesion sites is limited by the skin barrier. Recently, gold nanoparticle (AuNP)-mediated drug delivery into or through the epidermis and dermis has attracted much attention. However, the efficacy of the AuNP-mediated transdermal drug delivery remains unknown. In this study, gold nanoparticles were conjugated with VEGF and generated a surface by carrying negative charges, showing an ideal transdermal delivery efficacy for VEGF in wound repair. Our findings may provide new avenues for the treatment of cutaneous injuries.
Collapse
Affiliation(s)
- Ying Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Yonghui Wu
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Jining Gao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Zihao Zhang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Linjie Wang
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Xi Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Junwei Mi
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Yuanjiang Yao
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Dongwei Guan
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
| | - Bing Chen
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing 100101, China
| | - Jianwu Dai
- Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing Engineering Research Center for Nanomedicine, College of Preventive Medicine, Chongqing Engineering Research Center for Biomaterials and Regenerative Medicine, Third Military Medical University , Chongqing 400038, China
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences , Beijing 100101, China
| |
Collapse
|