1
|
Nazemidashtarjandi S, Larsen B, Cheng K, Faulkner S, Peppas NA, Parekh SH, Zoldan J. Near-infrared light-responsive hydrogels for on-demand dual delivery of proangiogenic growth factors. Acta Biomater 2024; 183:61-73. [PMID: 38838911 PMCID: PMC11514431 DOI: 10.1016/j.actbio.2024.05.052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/07/2024]
Abstract
Achieving precise spatiotemporal control over the release of proangiogenic factors is crucial for vasculogenesis, the process of de novo blood vessel formation. Although various strategies have been explored, there is still a need to develop cell-laden biomaterials with finely controlled release of proangiogenic factors at specific locations and time points. We report on the developed of a near-infrared (NIR) light-responsive collagen hydrogel comprised of gold nanorods (GNRs)-conjugated liposomes containing proangiogenic growth factors (GFs). We demonstrated that this system enables on-demand dual delivery of GFs at specific sites and over selected time intervals. Liposomes were strategically formulated to encapsulate either platelet-derived growth factor (PDGF) or vascular endothelial growth factor (VEGF), each conjugated to gold nanorods (GNRs) with distinct geometries and surface plasmon resonances at 710 nm (GNR710) and 1064 nm (GNR1064), respectively. Using near infrared (NIR) irradiation and two-photon (2P) luminescence imaging, we successfully demonstrated the independent release of PDGF from GNR710 conjugated liposomes and VEGF from GNR1064-conjugated liposomes. Our imaging data revealed rapid release kinetics, with localized PDGF released in approximately 4 min and VEGF in just 1 and a half minutes following NIR laser irradiation. Importantly, we demonstrated that the release of each GF could be independently triggered using NIR irradiation with the other GF formulation remaining retained within the liposomes. This light-responsive collagen hydrogels holds promise for various applications in regenerative medicine where the establishment of a guided vascular network is essential for the survival and integration of engineered tissues. STATEMENT OF SIGNIFICANCE: In this study, we have developed a light-responsive system with gold nanorods (GNRs)-conjugated liposomes in a collagen hydrogel, enabling precise dual delivery of proangiogenic growth factors (GFs) at specific locations and timepoints. Liposomes, containing platelet-derived growth factor (PDGF) or vascular endothelial growth factor (VEGF), release independently under near- infrared irradiation. This approach allows external activation of desired GF release, ensuring high cell viability. Each GF can be triggered independently, retaining the other within the liposomes. Beyond its application in establishing functional vascular networks, this dual delivery system holds promise as a universal platform for delivering various combinations of two or more GFs.
Collapse
Affiliation(s)
- Saeed Nazemidashtarjandi
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Bryce Larsen
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Kristie Cheng
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Sara Faulkner
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates; Department of Chemical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Sapun H Parekh
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates
| | - Janet Zoldan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX 78711, United Sates.
| |
Collapse
|
2
|
Madonna R, Barachini S, Ghelardoni S, Lu L, Shen WF, De Caterina R. Vasostatins: new molecular targets for atherosclerosis, post-ischaemic angiogenesis, and arteriogenesis. Cardiovasc Res 2024; 120:132-139. [PMID: 38242632 DOI: 10.1093/cvr/cvae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/25/2023] [Accepted: 11/30/2023] [Indexed: 01/21/2024] Open
Abstract
The chromogranin-secretogranin secretory proteins-granins-are acidic proteins localized in granules of endocrine cells and neurons. The chromogranin family includes chromogranins A (CgA) and B, as well as secretogranin II (once called chromogranin C). Members of this family undergo catalytic proteolysis to produce active peptides. The CgA-derived peptides vasostatin-1 and vasostatin-2, in particular, appear to protect against atherosclerosis, suppressing the expression of vascular cell adhesion molecule-1 and intercellular adhesion molecule-1, as well as exerting vasodilatory effects by enhancing nitric oxide bioavailability. Vasostatin-1 also suppresses vasoconstriction and abnormal angiogenesis. Vasostatin-1 and vasostatin-2 may be novel therapeutic targets for atherosclerosis and coronary heart disease, also protecting the myocardium against ischaemic damage.
Collapse
Affiliation(s)
- Rosalinda Madonna
- Department of Surgical, Medical, and Molecular Pathology and Critical Care, University of Pisa and Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| | - Serena Barachini
- Department of Clinical and Experimental Medicine, Laboratory for Cell Therapy, University of Pisa, Pisa, Italy
| | - Sandra Ghelardoni
- Department of Pathology, Laboratory of Biochemistry, University of Pisa, Pisa, Italy
| | - Lin Lu
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai 200025, People's Republic of China
| | - Wei-Feng Shen
- Department of Cardiovascular Medicine, Rui Jin Hospital, Shanghai Jiaotong University School of Medicine, 197 Rui Jin Road II, Shanghai 200025, People's Republic of China
| | - Raffaele De Caterina
- Department of Surgical, Medical, and Molecular Pathology and Critical Care, University of Pisa and Pisa University Hospital, Via Paradisa 2, 56124 Pisa, Italy
| |
Collapse
|
3
|
Longoni A, Major GS, Jiang S, Farrugia BL, Kieser DC, Woodfield TBF, Rnjak-Kovacina J, Lim KS. Pristine gelatin incorporation as a strategy to enhance the biofunctionality of poly(vinyl alcohol)-based hydrogels for tissue engineering applications. Biomater Sci 2023; 12:134-150. [PMID: 37933486 DOI: 10.1039/d3bm01172k] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2023]
Abstract
Synthetic polymers, such as poly(vinyl alcohol) (PVA), are popular biomaterials for the fabrication of hydrogels for tissue engineering and regenerative medicine (TERM) applications, as they provide excellent control over the physico-chemical properties of the hydrogel. However, their bioinert nature is known to limit cell-biomaterial interactions by hindering cell infiltration, blood vessel recruitment and potentially limiting their integration with the host tissue. Efforts in the field have therefore focused on increasing the biofunctionality of synthetic hydrogels, without limiting the advantages associated with their tailorability and controlled release capacity. The aim of this study was to investigate the suitability of pristine gelatin to enhance the biofunctionality of tyraminated PVA (PVA-Tyr) hydrogels, by promoting cell infiltration and host blood vessel recruitment for TERM applications. Pure PVA-Tyr hydrogels and PVA-Tyr hydrogels incorporated with vascular endothelial growth factor (VEGF), a well-known pro-angiogenic stimulus, were used for comparison. Incorporating increasing concentrations of VEGF (0.01-10 μg mL-1) or gelatin (0.01-5 wt%) did not influence the physical properties of PVA-Tyr hydrogels. However, their presence within the polymer network (>0.1 μg mL-1 VEGF and >0.1 wt% gelatin) promoted endothelial cell interactions with the hydrogels. The covalent binding of unmodified gelatin or VEGF to the PVA-Tyr network did not hamper their inherent bioactivity, as they both promoted angiogenesis in a chick chorioallantoic membrane (CAM) assay, performing comparably with the unbound VEGF control. When the PVA-Tyr hydrogels were implanted subcutaneously in mice, it was observed that cell infiltration into the hydrogels was possible in the absence of gelatin or VEGF at 1- or 3-weeks post-implantation, highlighting a clear difference between in vitro an in vivo cell-biomaterial interaction. Nevertheless, the presence of gelatin or VEGF was necessary to enhance blood vessel recruitment and infiltration, although no significant difference was observed between these two biological molecules. Overall, this study highlights the potential of gelatin as a standalone pro-angiogenic cue to enhance biofunctionality of synthetic hydrogels and provides promise for their use in a variety of TERM applications.
Collapse
Affiliation(s)
- Alessia Longoni
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, New Zealand.
| | - Gretel S Major
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, New Zealand.
| | - Shaoyuan Jiang
- Graduate School of Biomedical Engineering, UNSW Sydney, Sydney 2052, Australia
| | - Brooke L Farrugia
- School of Biomedical Engineering, University of Melbourne, Australia
| | - David C Kieser
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, New Zealand.
| | - Tim B F Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, New Zealand.
| | | | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, University of Otago Christchurch, New Zealand.
- Light-Activated Biomaterials Group, School of Medical Sciences, University of Sydney, Australia
| |
Collapse
|
4
|
Wang J, Song Y, Xie W, Zhao J, Wang Y, Yu W. Therapeutic angiogenesis based on injectable hydrogel for protein delivery in ischemic heart disease. iScience 2023; 26:106577. [PMID: 37192972 PMCID: PMC10182303 DOI: 10.1016/j.isci.2023.106577] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023] Open
Abstract
Ischemic heart disease (IHD) remains the leading cause of death and disability worldwide and leads to myocardial necrosis and negative myocardial remodeling, ultimately leading to heart failure. Current treatments include drug therapy, interventional therapy, and surgery. However, some patients with severe diffuse coronary artery disease, complex coronary artery anatomy, and other reasons are unsuitable for these treatments. Therapeutic angiogenesis stimulates the growth of the original blood vessels by using exogenous growth factors to generate more new blood vessels, which provides a new treatment for IHD. However, direct injection of these growth factors can cause a short half-life and serious side effects owing to systemic spread. Therefore, to overcome this problem, hydrogels have been developed for temporally and spatially controlled delivery of single or multiple growth factors to mimic the process of angiogenesis in vivo. This paper reviews the mechanism of angiogenesis, some important bioactive molecules, and natural and synthetic hydrogels currently being applied for bioactive molecule delivery to treat IHD. Furthermore, the current challenges of therapeutic angiogenesis in IHD and its potential solutions are discussed to facilitate real translation into clinical applications in the future.
Collapse
Affiliation(s)
- Junke Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
- Qingdao Medical College, Qingdao University, Qingdao, Shandong 266071, China
| | - Yancheng Song
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26000, China
| | - Wenjie Xie
- Department of Clinical Laboratory, The Affiliated Hospital of Qingdao University, Shandong, Qingdao, Shandong 26000, China
| | - Jiang Zhao
- Department of Urology, The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Ying Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong 26000, China
- Corresponding author
| | - Wenzhou Yu
- Department of Cardiovascular Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong 26003, China
- Corresponding author
| |
Collapse
|
5
|
Deng J, Zhang X, Yin M, Cao W, Zhang B, Liu Q, Hou X, Wang H, Shi C. Modified CFBP-bFGF targeting to ischemic brain promoted the functional recovery of cerebral ischemia. J Control Release 2023; 353:462-474. [PMID: 36493946 DOI: 10.1016/j.jconrel.2022.12.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/11/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022]
Abstract
The cerebral ischemia was one of the most common causes of disability and death worldwide. Basic fibroblast growth factor (bFGF) was reported to have neuroprotective function as well as promoting angiogenesis in the ischemic brain, but the targeting delivery of bFGF to ischemic brain was still difficult. In present study, a specific peptide was used to modify bFGF to construct recombinant CFBP-bFGF, and CFBP-bFGF could specifically deliver to ischemic brain through binding with the upregulated protein-connective tissue growth factor (CTGF). When CFBP-bFGF was used in rats with cerebral ischemia by intravenous injection, local concentration of the bFGF in ischemic brain was significantly increased. In addition, enhanced neurons survival, increased angiogenesis, decreased neuroinflammation were observed, that improved the motor functional recovery of cerebral ischemic injury. These results demonstrated that the targeting delivery of CFBP-bFGF would be a potential therapeutic approach for cerebral ischemia.
Collapse
Affiliation(s)
- Jin Deng
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China; Department of Neurology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266000, Shandong, China
| | - Xiaojing Zhang
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Mengmeng Yin
- The Second Department of Neurology, Qingdao Central Hospital, 127 South Siliu Road, Qingdao 266042, Shandong, China
| | - Wenxuan Cao
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China
| | - Bing Zhang
- Department of Neurology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266000, Shandong, China
| | - Qi Liu
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China; Department of Neurology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266000, Shandong, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100190, China..
| | - Haiping Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao 266000, Shandong, China.
| | - Chunying Shi
- Department of Human Anatomy, Histology and Embryology, School of Basic Medicine, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
6
|
Gionet-Gonzales MA, Gresham RCH, Griffin KH, Casella A, Wohlgemuth RP, Ramos-Rodriguez DH, Lowen J, Smith LR, Leach JK. Mesenchymal stromal cell spheroids in sulfated alginate enhance muscle regeneration. Acta Biomater 2023; 155:271-281. [PMID: 36328130 PMCID: PMC11970561 DOI: 10.1016/j.actbio.2022.10.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 09/26/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
The therapeutic efficacy of mesenchymal stromal cells (MSCs) for tissue regeneration is critically linked to the potency of the complex mixture of growth factors, cytokines, exosomes, and other biological cues that they secrete. The duration of cell-based approaches is limited by rapid loss of cells upon implantation, motivating the need to prolong cell viability and extend the therapeutic influence of the secretome. We and others demonstrated that the secretome is upregulated when MSCs are formed into spheroids. Although the efficacy of the MSC secretome has been characterized in the literature, no studies have reported the therapeutic benefit of in situ sequestration of the secretome within a wound site using engineered biomaterials. We previously demonstrated the capacity of sulfated alginate hydrogels to sequester components of the MSC secretome for prolonged presentation in vitro, yet the efficacy of this platform has not been evaluated in vivo. In this study, we used sulfated alginate hydrogels loaded with MSC spheroids to aid in the regeneration of a rat muscle crush injury. We hypothesized that the use of sulfated alginate to bind therapeutically relevant growth factors from the MSC spheroid secretome would enhance muscle regeneration by recruiting host cells into the tissue site. The combination of sulfated alginate and MSC spheroids resulted in decreased collagen deposition, improved myogenic marker expression, and increased neuromuscular junctions 2 weeks after injury. These data indicate that MSC spheroids delivered in sulfated alginate represent a promising approach for decreased fibrosis and increased functional regeneration of muscle. STATEMENT OF SIGNIFICANCE: The therapeutic efficacy of mesenchymal stromal cells (MSCs) for tissue regeneration is attributed to the complex diversity of the secretome. Cell-based approaches are limited by rapid cell death, motivating the need to extend the availability of the secretome. We previously demonstrated that sulfated alginate hydrogels sequester components of the MSC secretome for prolonged presentation in vitro, yet no studies have reported the in situ sequestration of the secretome. Herein, we transplanted MSC spheroids in sulfated alginate hydrogels to promote muscle regeneration. MSC spheroids in sulfated alginate decreased collagen deposition, improved myogenic marker expression, and increased neuromuscular junctions. These data indicate that MSC spheroids delivered in sulfated alginate represent a promising approach for decreasing fibrosis and increasing functional muscle regeneration.
Collapse
Affiliation(s)
| | - Robert C H Gresham
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | - Katherine H Griffin
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA; School of Veterinary Medicine, UC Davis, Davis, CA, USA
| | - Alena Casella
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | - Ross P Wohlgemuth
- Department of Neurobiology, Physiology and Behavior, UC Davis, Davis, CA, USA
| | | | - Jeremy Lowen
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA
| | - Lucas R Smith
- Department of Neurobiology, Physiology and Behavior, UC Davis, Davis, CA, USA; Department of Physical Medicine and Rehabilitation, UC Davis Health, Sacramento, CA, USA
| | - J Kent Leach
- Department of Orthopaedic Surgery, School of Medicine, UC Davis Health, Sacramento, CA, USA; Department of Biomedical Engineering, UC Davis, Davis, CA, USA.
| |
Collapse
|
7
|
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: 10] [Impact Index Per Article: 3.3] [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
|
8
|
Santra M, Liu YC, Jhanji V, Yam GHF. Human SMILE-Derived Stromal Lenticule Scaffold for Regenerative Therapy: Review and Perspectives. Int J Mol Sci 2022; 23:ijms23147967. [PMID: 35887309 PMCID: PMC9315730 DOI: 10.3390/ijms23147967] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
A transparent cornea is paramount for vision. Corneal opacity is one of the leading causes of blindness. Although conventional corneal transplantation has been successful in recovering patients’ vision, the outcomes are challenged by a global lack of donor tissue availability. Bioengineered corneal tissues are gaining momentum as a new source for corneal wound healing and scar management. Extracellular matrix (ECM)-scaffold-based engineering offers a new perspective on corneal regenerative medicine. Ultrathin stromal laminar tissues obtained from lenticule-based refractive correction procedures, such as SMall Incision Lenticule Extraction (SMILE), are an accessible and novel source of collagen-rich ECM scaffolds with high mechanical strength, biocompatibility, and transparency. After customization (including decellularization), these lenticules can serve as an acellular scaffold niche to repopulate cells, including stromal keratocytes and stem cells, with functional phenotypes. The intrastromal transplantation of these cell/tissue composites can regenerate native-like corneal stromal tissue and restore corneal transparency. This review highlights the current status of ECM-scaffold-based engineering with cells, along with the development of drug and growth factor delivery systems, and elucidates the potential uses of stromal lenticule scaffolds in regenerative therapeutics.
Collapse
Affiliation(s)
- Mithun Santra
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
| | - Yu-Chi Liu
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Vishal Jhanji
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
| | - Gary Hin-Fai Yam
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
9
|
Bupphathong S, Quiroz C, Huang W, Chung PF, Tao HY, Lin CH. Gelatin Methacrylate Hydrogel for Tissue Engineering Applications—A Review on Material Modifications. Pharmaceuticals (Basel) 2022; 15:ph15020171. [PMID: 35215284 PMCID: PMC8878046 DOI: 10.3390/ph15020171] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 11/26/2022] Open
Abstract
To recreate or substitute tissue in vivo is a complicated endeavor that requires biomaterials that can mimic the natural tissue environment. Gelatin methacrylate (GelMA) is created through covalent bonding of naturally derived polymer gelatin and methacrylic groups. Due to its biocompatibility, GelMA receives a lot of attention in the tissue engineering research field. Additionally, GelMA has versatile physical properties that allow a broad range of modifications to enhance the interaction between the material and the cells. In this review, we look at recent modifications of GelMA with naturally derived polymers, nanomaterials, and growth factors, focusing on recent developments for vascular tissue engineering and wound healing applications. Compared to polymers and nanoparticles, the modifications that embed growth factors show better mechanical properties and better cell migration, stimulating vascular development and a structure comparable to the natural-extracellular matrix.
Collapse
Affiliation(s)
- Sasinan Bupphathong
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan; (S.B.); (H.-Y.T.)
| | - Carlos Quiroz
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
| | - Wei Huang
- Department of Orthodontics, Rutgers School of Dental Medicine, Newark, NJ 07103, USA;
| | - Pei-Feng Chung
- School of Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 110, Taiwan;
| | - Hsuan-Ya Tao
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan; (S.B.); (H.-Y.T.)
| | - Chih-Hsin Lin
- Graduate Institute of Nanomedicine and Medical Engineering, Taipei Medical University, Taipei 110, Taiwan; (S.B.); (H.-Y.T.)
- Correspondence:
| |
Collapse
|
10
|
Fu B, Wang X, Chen Z, Jiang N, Guo Z, Zhang Y, Zhang S, Liu X, Liu L. Improved myocardial performance in infarcted rat heart by injection of disulfide-cross-linked chitosan hydrogels loaded with basic fibroblast growth factor. J Mater Chem B 2022; 10:656-665. [PMID: 35014648 DOI: 10.1039/d1tb01961a] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Myocardial infarction (MI) has been considered as the leading cause of cardiovascular-related deaths worldwide. Basic fibroblast growth factor (bFGF) is a member of the fibroblast growth factor family that promotes angiogenesis after MI; however, it has poor clinical efficacy due to proteolytic degradation, low drug accumulation, and severe drug-induced side effects. In this study, an injectable disulfide-cross-linked chitosan hydrogel loaded with bFGF was prepared via a thiol-disulfide exchange reaction for MI treatment. The thiol-disulfide exchange reaction between pyridyl disulfide-modified carboxymethyl chitosan (CMCS-S-S-Py) and reduced BSA (rBSA) was carried out under physiological conditions (37 °C and pH 7.4). The mechanical properties of the disulfide-cross-linked chitosan hydrogel were evaluated based on the molar ratio of the pyridyl disulfide groups of CMCS-S-S-Py and the thiol groups of rBSA. The disulfide-cross-linked chitosan hydrogel showed good swelling performance, rapid glutathione-triggered degradation behavior and well-defined cell proliferation towards NIH 3T3 fibroblast cells. In the process of establishing a rat MI model, the squeezing heart method was used to make the operation more accurate and the mortality of rats was decreased by using a ventilator. The disulfide-cross-linked chitosan hydrogel loaded with bFGF (bFGF-hydrogel) was injected into a peri-infarcted area of cardiac tissue immediately following MI. Echocardiography demonstrated that the left ventricular functions were improved by the bFGF-hydrogel after 28 days of treatment. Histological results revealed that the hydrogel significantly reduced the fibrotic area of MI, and this was further improved by the bFGF-hydrogel treatment. TUNEL and immunohistochemical staining results showed that the bFGF-hydrogel had a more synergistic effect on antiapoptosis and proangiogenesis than using either bFGF or the hydrogel alone.
Collapse
Affiliation(s)
- Bo Fu
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300051, P. R. China. .,Tianjin Medical University, Tianjin 300203, P. R. China
| | - Xiaobei Wang
- Department of Materials Engineering, North China Institute of Aerospace Engineering, Langfang 065000, P. R. China
| | - Zhengda Chen
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300051, P. R. China. .,Tianjin Medical University, Tianjin 300203, P. R. China
| | - Nan Jiang
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300051, P. R. China.
| | - Zhigang Guo
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300051, P. R. China.
| | - Yuhui Zhang
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300051, P. R. China.
| | - Shaopeng Zhang
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300051, P. R. China.
| | - Xiankun Liu
- Department of Cardiovascular Surgery, Tianjin Chest Hospital, Tianjin 300051, P. R. China.
| | - Li Liu
- Key Laboratory of Functional Polymer Materials, Ministry of Education, Institute of Polymer Chemistry, College of Chemistry, Nankai University, Tianjin 300071, P. R. China.
| |
Collapse
|
11
|
Isosteviol improves cardiac function and promotes angiogenesis after myocardial infarction in rats. Cell Tissue Res 2021; 387:275-285. [PMID: 34820705 DOI: 10.1007/s00441-021-03559-9] [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: 05/21/2021] [Accepted: 11/12/2021] [Indexed: 10/19/2022]
Abstract
Isosteviol has been indicated as a cardiomyocyte protector. However, the underlying mechanism remains unclear. Thus, we sought to confirm the protective effect of isosteviol after myocardial infarction in a model of permanent coronary artery occlusion and investigate the potential proangiogenic activity in vitro and in vivo. A 4-week permanent coronary artery occlusion rat model was generated, and the protective effect of isosteviol was evaluated by echocardiographic imaging and hemodynamics assays. The coronary capillary density was tested by immunochemistry and micro-computed tomography (μCT) imaging. The effect of isosteviol on endothelial cells was determined in human umbilical vein endothelial cells (HUVECs) in vitro and Tg (kdrl: EGFP) zebrafish in vivo. We also examined the expression of related transcription factors by real-time polymerase chain reaction (RT-qPCR). Isosteviol increased ejection fraction (EF), fractional shortening (FS), cardiac systolic index (CI), maximum rate of increase of left ventricular pressure (Max dp/dt), and left ventricular systolic pressure (LVSP) by 32%, 40%, 25%, 26%, and 10%, respectively, in permanent coronary artery occlusion rats. Interestingly, it also promoted coronary capillary density by 2.5-fold. In addition, isosteviol promoted the proliferation and branching of HUVECs in vitro. It also rescued intersegmental vessel (ISV) development and improved endothelial cell proliferation by approximately fivefold (4-6) in zebrafish embryos in vivo. Isosteviol also upregulated the expression of hypoxia inducible factor-1α (HIF-1α) and vascular endothelial growth factor A (VEGFA) in zebrafish by fourfold and 3.5-fold, respectively. Our findings suggest that isosteviol is a proangiogenic agent and that this activity is related to its protective effects against myocardial ischemia. After using the permanent coronary artery occlusion model, we demonstrated that isosteviol promotes angiogenesis directly and increases capillary density in myocardial ischemia rats. Isosteviol promotes angiogenesis in zebrafish in vivo and increases vascular endothelial cell proliferation in HUVECs and zebrafish. The angiogenesis activity of isosteviol may be correlated with VEGFA and HIF-1α signaling.
Collapse
|
12
|
Analysis of CGF Biomolecules, Structure and Cell Population: Characterization of the Stemness Features of CGF Cells and Osteogenic Potential. Int J Mol Sci 2021; 22:ijms22168867. [PMID: 34445573 PMCID: PMC8396261 DOI: 10.3390/ijms22168867] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/12/2021] [Accepted: 08/15/2021] [Indexed: 01/08/2023] Open
Abstract
Concentrated Growth Factors (CGF) represent new autologous (blood-derived biomaterial), attracting growing interest in the field of regenerative medicine. In this study, the chemical, structural, and biological characterization of CGF was carried out. CGF molecular characterization was performed by GC/MS to quantify small metabolites and by ELISA to measure growth factors and matrix metalloproteinases (MMPs) release; structural CGF characterization was carried out by SEM analysis and immunohistochemistry; CGF has been cultured, and its primary cells were isolated for the identification of their surface markers by flow cytometry, Western blot, and real-time PCR; finally, the osteogenic differentiation of CGF primary cells was evaluated through matrix mineralization by alizarin red staining and through mRNA quantification of osteogenic differentiation markers by real-time PCR. We found that CGF has a complex inner structure capable of influencing the release of growth factors, metabolites, and cells. These cells, which could regulate the production and release of the CGF growth factors, show stem features and are able to differentiate into osteoblasts producing a mineralized matrix. These data, taken together, highlight interesting new perspectives for the use of CGF in regenerative medicine.
Collapse
|
13
|
Zhang H, Zhang Z, Zhang H, Chen C, Zhang D, Zhao Y. Protein-Based Hybrid Responsive Microparticles for Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2021; 13:18413-18422. [PMID: 33856190 DOI: 10.1021/acsami.0c19884] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The in-depth development of biological materials, especially natural polymer materials, has injected strong vitality into clinical wound treatment. Here, a new type of controllable responsive microparticles composed of several natural polymer materials was presented for drug release and wound healing. These hybrid microparticles consisted of silk fibroin, gelatin, agarose, and black phosphorus quantum dots (BPQDs) and were loaded with growth factors and antibacterial peptides. Under near-infrared (NIR) irradiation, BPQDs could absorb the NIR light and increase the temperature of the microparticles to the melting point of gelatin. When the gelatin started to melt, the encapsulated drugs were gradually released because of the reversible phase transformation. Both in vitro and in vivo experiments have demonstrated that the BPQD-laden microparticles with a NIR-responsive feature could achieve the desired controllable release of growth factors to promote neovascularization formation. In addition, because antibacterial peptides were also mixed with the secondary hydrogel and encapsulated in the scaffolds, the microparticles are imparted with the antibacterial ability during storage and usage. These characteristics of BPQD-laden natural protein hybrid microparticles make them ideal for drug delivery and wound healing.
Collapse
Affiliation(s)
- Hui Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Zhuohao Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Han Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
| | - Canwen Chen
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Dagan Zhang
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
| | - Yuanjin Zhao
- Department of Clinical Laboratory, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing 210002, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China
- Chemistry and Biomedicine Innovation Center, Nanjing University, Nanjing 210023, People's Republic of China
| |
Collapse
|
14
|
Zhang J, Liu X, Ma K, Chen M, Xu H, Niu X, Gu H, Wang R, Chen X, Sun H. Collagen/heparin scaffold combined with vascular endothelial growth factor promotes the repair of neurological function in rats with traumatic brain injury. Biomater Sci 2021; 9:745-764. [DOI: 10.1039/c9bm01446b] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
The objective of this study was to evaluate the therapy effects of a novel biological scaffold containing heparin, collagen and vascular endothelial growth factor (VEGF) in treating traumatic brain injury (TBI).
Collapse
Affiliation(s)
- Jian Zhang
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Xiaoyin Liu
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Ke Ma
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Miao Chen
- Affiliated Hospital of Traditional Chinese Medicine
- Xinjiang Medical University
- Urumqi
- China
| | - Huiyou Xu
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | | | - Haoran Gu
- The 947th hospital of Chinese People's Liberation Army
- Xinjiang
- China
| | - Renjie Wang
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - Xuyi Chen
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| | - HongTao Sun
- Tianjin Key Laboratory of Neurotrauma Repair
- Institute of Traumatic Brain Injury and Neuroscience
- Characteristic Medical Center of Chinese People's Armed Police Force
- Tianjin 300162
- China
| |
Collapse
|
15
|
Mizuno Y, Taguchi T. Self-assembled dodecyl group-modified gelatin microparticle-based hydrogels with angiogenic properties. NPG ASIA MATERIALS 2020; 12:48. [DOI: 10.1038/s41427-020-0229-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/18/2020] [Accepted: 04/25/2020] [Indexed: 01/05/2025]
Abstract
AbstractSupplying oxygen and nutrients to implanted cells or tissues is an important factor that improves their survivability and function in regenerative medicine. Various efforts have been made to develop angiogenic materials by incorporating and releasing growth factors such as vascular endothelial growth factor (VEGF). However, these exogenous growth factors have a short half-life under physiological conditions. We therefore designed a novel angiogenic microparticle (C12-MP) comprising Alaska pollock-derived gelatin (ApGltn) modified with a dodecyl group (C12-ApGltn) to stimulate endogenous VEGF secretion. The C12-MP suspension formed an injectable hydrogel, the rheological properties and enzymatic degradation of which were evaluated. RAW264 cells, mouse macrophage-like cells, cultured with C12-MPs, secreted significantly more VEGF than the original ApGltn MPs. Based on laser Doppler perfusion imaging, the C12-MP hydrogel clearly induced increased blood perfusion in a subcutaneous mouse model compared with the original ApGltn microparticle (Org-MP) or phosphate-buffered saline controls. Histological studies revealed that the areas of nuclear factor (NF)-κB, CD31, and myeloperoxidase staining showed a greater increase at the site injected with C12-MPs than at the site injected with the original ApGltn microparticles or phosphate-buffered saline. The C12-MP hydrogel is a promising angiogenic material for constructing vascular beds for cell transplantation by promoting endogenous VEGF secretion without additional growth factors.
Collapse
|
16
|
Tobiume S, Kaji Y, Nakamura O, Yamaguchi K, Oka K, Yamamoto T. Effects of VEGF on Prefabricated Vascularized Bone Allografts in Rats. J Reconstr Microsurg 2020; 37:405-412. [PMID: 33058099 DOI: 10.1055/s-0040-1718394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
BACKGROUND Massive bone defects after wide resection of malignant bone tumors or a serious injury require treatment using vascularized bone grafts. Although cadaveric bone allografts combined with vascularized bone autografts are currently thought to be ideal in terms of size and durability, this treatment requires the scarification of healthy bone tissue. In a previous study, we attempted to improve this situation by prefabricating a vascularized bone allograft in recipient rats. In this study, we added vascular endothelial growth factor (VEGF)-containing hydroxyapatite/collagen composite (HAp/Col) to a prefabricated vascularized bone allograft to stimulate angiogenesis, which is known to be important for bone formation. METHODS Sprague Dawley rats (n = 50) were used as donors and Wistar rats (n = 50) as recipients. All rats were 9 weeks old. The recipient rats were divided into five groups according to the use of vascular bundles, HAp/Col, and an additive substance (VEGF). The bone allografts collected from the donors were transplanted into the thigh region of the recipients, and a saphenous vein and 10 μg HAp/Col with VEGF were inserted into the bone allografts through the slit. After 4 weeks, the transplanted bone allografts were harvested, and histologic and genetic evaluations were performed in relation to bone formation and resorption. RESULTS The results showed that, compared with the control group, the implantation of the vascular bundles and VEGF-containing HAp/Col significantly stimulated angiogenesis and bone formation in the rats with the bone allografts. However, histological and genetic evaluations of bone resorption revealed that resorption was not observed in any group. CONCLUSION These results suggest that VEGF-containing HAp/Col effectively stimulates angiogenesis and bone formation, but not bone resorption, in prefabricated vascularized bone allografts. This method could therefore become a useful tool for treating large bone defects.
Collapse
Affiliation(s)
- Sachiko Tobiume
- Department of Orthopaedic Surgery, Kagawa University Faculty of Medicine, Miki-cho, Kita-gun, Kagawa, Japan
| | - Yoshio Kaji
- Department of Orthopaedic Surgery, Kagawa University Faculty of Medicine, Miki-cho, Kita-gun, Kagawa, Japan
| | - Osamu Nakamura
- Department of Orthopaedic Surgery, Kagawa University Faculty of Medicine, Miki-cho, Kita-gun, Kagawa, Japan
| | - Konosuke Yamaguchi
- Department of Orthopaedic Surgery, Kagawa University Faculty of Medicine, Miki-cho, Kita-gun, Kagawa, Japan
| | - Kunihiko Oka
- Department of Orthopaedic Surgery, Kagawa University Faculty of Medicine, Miki-cho, Kita-gun, Kagawa, Japan
| | - Tetsuji Yamamoto
- Department of Orthopaedic Surgery, Kagawa University Faculty of Medicine, Miki-cho, Kita-gun, Kagawa, Japan
| |
Collapse
|
17
|
Dinh ND, Kukumberg M, Nguyen AT, Keramati H, Guo S, Phan DT, Ja'Afar NB, Birgersson E, Leo HL, Huang RYJ, Kofidis T, Rufaihah AJ, Chen CH. Functional reservoir microcapsules generated via microfluidic fabrication for long-term cardiovascular therapeutics. LAB ON A CHIP 2020; 20:2756-2764. [PMID: 32609786 DOI: 10.1039/d0lc00296h] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Cardiovascular disease is a chronic disease that leads to impaired cardiac function and requires long-term management to control its progression. Despite the importance of hydrogels for therapeutic applications, a contradiction between the size of a hydrogel and the amount of loaded drug has been encountered when using conventional fabrication methods. In this study, biocompatible reservoir microcapsules (diameter ∼100 μm) with a large liquid core and polymeric shell were fabricated via a one-step phase separation of poly(ethylene glycol)diacrylate (PEGDA) and dextran within pre-gel droplets through microfluidics. By controlling the process of phase separation, high drug-loading efficiency (∼80%) for long-term release (30 days) of vascular endothelial growth factor (VEGF) and platelet-derived growth factor (PDGF) was achieved. Drug molecules were dispersed within the liquid core at a concentration above saturation solubility for sustained delivery via regulation of the shells. Effective therapeutic enhancement of human umbilical vein endothelial cell (HUVEC) and umbilical artery smooth muscle cell (SMC) proliferation and tube formation in vitro promoted rapid cell proliferation and increased the number of migrated cells by ∼1.7 times. Moreover, in vivo blood vessel regeneration for cardiovascular control induced by sustained dual-drug (VEGF and PDGF) delivery to the rat heart was achieved, showing the effectiveness of long-term protein delivery in improving cardiac function and significantly reducing ventricular wall thickness and fibrosis of the infarct region. The ratio of heart tissue scarring was reduced to 11.2% after microcapsule treatment compared with 21.4% after saline treatment in the rat model. By using these reservoir microcapsules, similar sustained delivery of proteins, mRNAs and biologic drugs could be developed for the treatment of a range of long-term chronic diseases and regenerative medicine.
Collapse
Affiliation(s)
- Ngoc-Duy Dinh
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, 04-08, 117583, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Jin C, Cao N, Ni J, Zhao W, Gu B, Zhu W. A Lipid-Nanosphere-Small MyoD Activating RNA-Bladder Acellular Matrix Graft Scaffold [NP(saMyoD)/BAMG] Facilitates Rat Injured Bladder Muscle Repair and Regeneration [NP(saMyoD)/BAMG]. Front Pharmacol 2020; 11:795. [PMID: 32581787 PMCID: PMC7287117 DOI: 10.3389/fphar.2020.00795] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 05/14/2020] [Indexed: 12/18/2022] Open
Abstract
Background Bladder tissue engineering is an excellent alternative to conventional gastrointestinal bladder enlargement in the treatment of various acquired and congenital bladder abnormalities. We constructed a nanosphere-small MyoD activating RNA-bladder acellular matrix graft scaffold NP(saMyoD)/BAMG inoculated with adipose-derived stem cells (ADSC) to explore its effect on smooth muscle regeneration and bladder repair function in a rat augmentation model. Methods We performed many biotechniques, such as reverse transcriptase-polymerase chain reaction (RT-PCR), Western blot, MTT assay, HE staining, masson staining, and immunohistochemistry in our study. Lipid nanospheres were transfected into rat ADSCs after encapsulate saRNA-MyoD as an introduction vector. Lipid nanospheres encapsulated with saRNA-MyoD were transfected into rat ADSCs. The functional transfected rat ADSCs were called ADSC-NP(saMyoD). Then, Rat models were divided into four groups: sham group, ADSC-BAMG group, ADSC-NP(saMyoD)/BAMG group, and ADSC-NP(saMyoD)/SF(VEGF)/BAMG group. Finally, we compared the bladder function of different models by detecting the bladder histology, bladder capacity, smooth muscle function in each group. Results RT-PCR and Western blot results showed that ADSCs transfected with NP(saMyoD) could induce high expression of α-SMA, SM22α, and Desmin. At the same time, MTT analysis showed that NP(saMyoD) did not affect the activity of ADSC cells, suggesting little toxicity. HE staining and immunohistochemistry indicated that the rat bladder repair effect (smooth muscle function, bladder capacities) was better in the ADSC-NP(saMyoD)/BAMG group, ADSC-NP(saMyoD)/SF(VEGF)/BAMG group than in the control group. Conclusions Taken together, our results demonstrate that the NP(saMyoD)/SF(VEGF)/BAMG scaffold seeded with ADSCs could promote bladder morphological regeneration and improved bladder urinary function. This strategy of ADSC-NP(saMyoD)/SF(VEGF)/BAMG may has a potential to repair bladder defects in the future.
Collapse
Affiliation(s)
- Chongrui Jin
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Urology, Shanghai Eastern Urological Reconstruction and Repair Institute, Shanghai, China
| | - Nailong Cao
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Urology, Shanghai Eastern Urological Reconstruction and Repair Institute, Shanghai, China
| | - Jianshu Ni
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Urology, Shanghai Eastern Urological Reconstruction and Repair Institute, Shanghai, China
| | - Weixin Zhao
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, NC, United States
| | - Baojun Gu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Urology, Shanghai Eastern Urological Reconstruction and Repair Institute, Shanghai, China
| | - Weidong Zhu
- Department of Urology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.,Department of Urology, Shanghai Eastern Urological Reconstruction and Repair Institute, Shanghai, China
| |
Collapse
|
19
|
Pei X, Kim H, Lee M, Wang N, Shin J, Lee S, Yoon M, Yang VC, He H. Local delivery of cardiac stem cells overexpressing HIF-1α promotes angiogenesis and muscular tissue repair in a hind limb ischemia model. J Control Release 2020; 322:610-621. [DOI: 10.1016/j.jconrel.2020.03.017] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/23/2020] [Accepted: 03/13/2020] [Indexed: 12/14/2022]
|
20
|
Basiri H, Abouei Mehrizi A, Ghaee A, Farokhi M, Chekini M, Kumacheva E. Carbon Dots Conjugated with Vascular Endothelial Growth Factor for Protein Tracking in Angiogenic Therapy. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:2893-2900. [PMID: 32125865 DOI: 10.1021/acs.langmuir.9b03980] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
One of the challenges of using growth factors for tissue regeneration is to monitor their biodistributions and delivery to injured tissues for minimally invasive detection. In the present study, tracking of human vascular endothelial growth factor (VEGF) was achieved by chemically linking it to photoluminescent carbon dots (CDs). Carbon dots were synthesized by the hydrothermal method and, subsequently, conjugated with VEGF using carbodiimide coupling. ELISA and western blot analysis revealed that VEGF-conjugated CDs preserve the binding affinity of VEGF to its antibodies. We also show that VEGF-conjugated CDs maintain the functionality of VEGF for tube formation and cell migration. The VEGF-conjugated CDs were also used for in vitro imaging of human umbilical vein endothelial cells. The results of this work suggest that cell-penetrating VEGF-conjugated CDs can be used for growth factor protein tracking in therapeutic and tissue engineering applications.
Collapse
Affiliation(s)
- Hamideh Basiri
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran 1439957131, Iran
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Ali Abouei Mehrizi
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran 1439957131, Iran
| | - Azadeh Ghaee
- Faculty of New Sciences and Technologies, Department of Life Science Engineering, University of Tehran, Tehran 1439957131, Iran
| | - Mehdi Farokhi
- National Cell Bank of Iran, Pasteur Institute of Iran, Tehran 1316943551, Iran
| | - Mahshid Chekini
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
| | - Eugenia Kumacheva
- Department of Chemistry, University of Toronto, 80 Saint George Street, Toronto, Ontario M5S 3H6, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, 4 Taddle Creek Road, Toronto, Ontario M5S 3G9, Canada
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| |
Collapse
|
21
|
Mizuno Y, Taguchi T. A hydrophobic gelatin fiber sheet promotes secretion of endogenous vascular endothelial growth factor and stimulates angiogenesis. RSC Adv 2020; 10:24800-24807. [PMID: 35517459 PMCID: PMC9055140 DOI: 10.1039/d0ra03593a] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 06/20/2020] [Indexed: 01/24/2023] Open
Abstract
In tissue engineering and regenerative medicine, the formation of vascular beds is an effective method to supply oxygen and nutrients to implanted cells or tissues to improve their survival and promote normal cellular functions. Various types of angiogenic materials have been developed by incorporating growth factors, such as vascular endothelial growth factor, in biocompatible materials. However, these exogenous growth factors suffer from instability and inactivation under physiological conditions. In this study, we designed a novel angiogenic electrospun fiber sheet (C16-FS) composed of Alaska pollock-derived gelatin (ApGltn) modified with hexadecyl (C16) groups to induce localized and sustained angiogenesis without growth factors. C16-FS was thermally crosslinked to enhance its stability. We demonstrated that C16-FS swells in phosphate-buffered saline for over 24 h and resists degradation. Laser doppler perfusion imaging showed that C16-FS induced increased blood perfusion when implanted subcutaneously in rats compared with unmodified ApGltn-fiber sheets (Org-FS) and the sham control. Furthermore, angiogenesis was sustained for up to 7 days following implantation. Immunohistochemical studies revealed elevated nuclear factor-κB and CD31 levels around the C16-FS implantation site compared with the Org-FS implantation site and the control incision site. These results demonstrate that C16-FS is a promising angiogenic material to promote the formation of vascular beds for cell and tissue transplantation without the need for growth factors. In vivo long-term growth factor-free angiogenesis by LPS-mimicking C16-modified gelatin based electrospun fiber sheet.![]()
Collapse
Affiliation(s)
- Yosuke Mizuno
- Graduate School of Science and Technology
- University of Tsukuba
- Tsukuba
- Japan
- Polymers and Biomaterials Field
| | - Tetsushi Taguchi
- Graduate School of Science and Technology
- University of Tsukuba
- Tsukuba
- Japan
- Polymers and Biomaterials Field
| |
Collapse
|
22
|
Fan C, Shi J, Zhuang Y, Zhang L, Huang L, Yang W, Chen B, Chen Y, Xiao Z, Shen H, Zhao Y, Dai J. Myocardial-Infarction-Responsive Smart Hydrogels Targeting Matrix Metalloproteinase for On-Demand Growth Factor Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2019; 31:e1902900. [PMID: 31408234 DOI: 10.1002/adma.201902900] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/22/2019] [Indexed: 06/10/2023]
Abstract
Although in situ restoration of blood supply to the infarction region and attenuating pre-existing extracellular matrix degradation remain potential therapeutic approaches for myocardial infarction (MI), local delivery of therapeutics has been limited by low accumulation (inefficacy) and unnecessary diffusion (toxicity). Here, a dual functional MI-responsive hydrogel is fabricated for on-demand drug delivery to promote angiogenesis and inhibit cardiac remodeling by targeting upregulated matrix metalloproteinase-2/9 (MMP-2/9) after MI. A glutathione (GSH)-modified collagen hydrogel (collagen-GSH) is prepared by conjugating collagen amine groups with GSH sulfhydryl groups and the recombinant protein GST-TIMP-bFGF (bFGF: basic fibroblast growth factor) by fusing bFGF with glutathione-S-transferase (GST) and MMP-2/9 cleavable peptide PLGLAG (TIMP). Specific binding between GST and GSH significantly improves the amount of GST-TIMP-bFGF loaded in collagen-GSH hydrogel. The TIMP peptide enclosed between GST and bFGF responds to MMPs for on-demand release during MI. Additionally, the TIMP peptide is a competitive substrate of MMPs that inhibits the excessive degradation of cardiac matrix by MMPs after MI. GST-TIMP-bFGF/collagen-GSH hydrogels promote the recovery of MI rats by enhancing vascularization and ameliorating myocardium remodeling. The results suggest that on-demand growth factor delivery by synchronously controlling binding and responsive release to promote angiogenesis and attenuate cardiac remodeling might be promising for the treatment of ischemic heart disease.
Collapse
Affiliation(s)
- Caixia Fan
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jiajia Shi
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, 230026, China
| | - Yan Zhuang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Lulu Zhang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Lei Huang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Wen Yang
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Bing Chen
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yanyan Chen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - He Shen
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jianwu Dai
- Key Laboratory for Nano-Bio Interface Research, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
- State Key Laboratory of Molecular Development Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| |
Collapse
|
23
|
Mizuno Y, Taguchi T. Growth factor-free, angiogenic hydrogel based on hydrophobically modified Alaska pollock gelatin. J Tissue Eng Regen Med 2019; 13:2291-2299. [PMID: 31503405 DOI: 10.1002/term.2957] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/11/2019] [Accepted: 09/02/2019] [Indexed: 12/11/2022]
Abstract
Angiogenesis is important for supplying oxygen and nutrients to implanted cells and organs and thereby promoting their survival. However, exogenously administered growth factors such as vascular endothelial growth factor (VEGF) have a short half-life and are unstable under physiological conditions. In the present study, we developed an angiogenesis-inducing hydrogel by modifying Alaska pollock-derived gelatin with a dodecyl group (C12-ApGltn), and demonstrated that it is biodegradable and highly fluid at room temperature (25°C). C12-ApGltn dissolved in phosphate-buffered saline at 20 w/v% formed a self-assembling hydrogel with thixotropic properties that stimulated VEGF secretion by macrophage-like RAW264 cells. Moreover, C12-ApGltn stimulated nuclear factor-κB and VEGF expression when subcutaneously injected into mice and increased the cluster of differentiation 31-positive area compared with injection of unmodified ApGltn and phosphate-buffered saline control in the absence of any growth factors. Hematoxylin and eosin staining confirmed vascular capillaries around the C12-ApGltn injection site. These results demonstrate that C12-ApGltn hydrogel is a promising angiogenic material for clinical applications that can stimulate endogenous VEGF expression without requiring additional growth factors.
Collapse
Affiliation(s)
- Yosuke Mizuno
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Tetsushi Taguchi
- Graduate School of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan.,Biomaterials Field, Research Center for Functional Materials, National Institute for Materials Science, Tsukuba, Japan
| |
Collapse
|
24
|
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
|
25
|
Malakar AK, Choudhury D, Halder B, Paul P, Uddin A, Chakraborty S. A review on coronary artery disease, its risk factors, and therapeutics. J Cell Physiol 2019; 234:16812-16823. [PMID: 30790284 DOI: 10.1002/jcp.28350] [Citation(s) in RCA: 575] [Impact Index Per Article: 95.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 01/22/2019] [Accepted: 01/24/2019] [Indexed: 12/19/2022]
Abstract
Coronary artery disease (CAD) is one of the major cardiovascular diseases affecting the global human population. This disease has been proved to be the major cause of death in both the developed and developing countries. Lifestyle, environmental factors, and genetic factors pose as risk factors for the development of cardiovascular disease. The prevalence of risk factors among healthy individuals elucidates the probable occurrence of CAD in near future. Genome-wide association studies have suggested the association of chromosome 9p21.3 in the premature onset of CAD. The risk factors of CAD include diabetes mellitus, hypertension, smoking, hyperlipidemia, obesity, homocystinuria, and psychosocial stress. The eradication and management of CAD has been established through extensive studies and trials. Antiplatelet agents, nitrates, β-blockers, calcium antagonists, and ranolazine are some of the few therapeutic agents used for the relief of symptomatic angina associated with CAD.
Collapse
Affiliation(s)
- Arup Kr Malakar
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | | | - Binata Halder
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | - Prosenjit Paul
- Department of Biotechnology, Assam University, Silchar, Assam, India
| | - Arif Uddin
- Department of Zoology, Moinul Hoque Choudhury Memorial Science College, Hailakandi, Assam, India
| | | |
Collapse
|
26
|
Wang C, Yu Y, Chen H, Zhang S, Wang J, Liu C. Construction of cytokine reservoirs based on sulfated chitosan hydrogels for the capturing of VEGF in situ. J Mater Chem B 2019; 7:1882-1892. [PMID: 32255051 DOI: 10.1039/c8tb02895h] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Nutrients and oxygen are delivered mainly by blood vessels to nourish the cells and tissues in the body. Thus, biomaterials are processed by loading cytokines, such as vascular endothelial growth factors (VEGF), to facilitate angiogenesis in order to accelerate tissue regeneration. Nevertheless, the unpredictable biosecurity of exogenous cytokines is still a controversial issue for its clinical application. In this study, we constructed a kind of cytokine reservoir utilizing the binding affinity between heparin-like sulfate polysaccharide and endogenous growth factors. Two types of sulfated chitosan hydrogels, namely 6-O-sulfated chitosan (6-O-SCS) and 2-N,6-O-sulfated chitosan (2-N,6-O-SCS) hydrogels, were formed on the surface of the gelatin sponge matrix. The microstructure of the SCS-coated scaffolds is porous and interconnected, which is beneficial for cellular infiltration. Besides, human umbilical vein endothelial cells (HUVECs) can adhere and proliferate well on the surface of the scaffolds. Notably, sulfated chitosan-coated scaffolds exhibit an ability to capture VEGF in vitro & vivo, especially for the 2-N,6-O-SCS-coated scaffold. It is also verified by mice models that sulfated chitosan-coated scaffolds result in a concentrated VEGF microenvironment in specific domains as cytokine reservoirs and induce mass microvessels after implantation into subcutaneous tissues. As such, the sulfated chitosan-coated scaffolds served as VEGF reservoirs to accelerate angiogenesis and wound healing. This beneficial strategy may be applicable to in situ tissue regeneration by capturing more cytokines and promoting healing.
Collapse
Affiliation(s)
- Chenmin Wang
- The State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, People's Republic of China
| | | | | | | | | | | |
Collapse
|
27
|
Bozic M, Betriu A, Bermudez-Lopez M, Ortiz A, Fernandez E, Valdivielso JM, on behalf of the NEFRONA investigators. Association of FGF-2 Concentrations with Atheroma Progression in Chronic Kidney Disease Patients. Clin J Am Soc Nephrol 2018; 13:577-584. [PMID: 29519952 PMCID: PMC5969461 DOI: 10.2215/cjn.07980717] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 01/03/2018] [Indexed: 12/21/2022]
Abstract
BACKGROUND AND OBJECTIVES Atherosclerosis is highly prevalent in CKD. The rate of progression of atherosclerosis is associated with cardiovascular events. Fibroblast growth factor 2 (FGF-2) is a member of the FGF family with potentially both protective and deleterious effects in the development of atherosclerosis. The role of circulating FGF-2 levels in the progression of atherosclerosis in CKD is unknown. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS We used a multicenter, prospective, observational cohorts study of 481 patients with CKD. We determined the presence of atheroma plaque in ten arterial territories by carotid and femoral ultrasounds. Progression of atheromatosis was defined as an increase in the number of territories with plaque after 24 months. Plasma levels of FGF-2 were measured by multiplex analysis. A multivariable logistic regression analysis was performed to determine whether plasma FGF-2 levels were associated with atheromatosis progression. RESULTS Average age of the population was 61 years. The percentage of patients in each CKD stage was 51% in stage 3, 41% in stages 4-5, and 8% in dialysis. A total of 335 patients (70%) showed plaque at baseline. Atheromatosis progressed in 289 patients (67%). FGF-2 levels were similar between patients with or without plaque at baseline (79 versus 88 pg/ml), but lower in patients with atheromatosis progression after 2 years (78 versus 98 pg/ml; P<0.01). In adjusted analyses, higher plasma FGF-2 was associated with lower risk of atheromatosis progression (odds ratio [OR], 0.86; 95% confidence interval [95% CI], 0.76 to 0.96; per 50 pg/ml increment). Analysis of FGF-2 in tertiles showed that atheroma progression was observed for 102 participants in the lowest tertile of FGF-2 (reference group), 86 participants in the middle tertile of FGF-2 (adjusted OR, 0.70; 95% CI, 0.40 to 1.20), and 74 participants in the lowest tertile of FGF-2 (adjusted OR, 0.48; 95% CI, 0.28 to 0.82). CONCLUSIONS Low FGF-2 levels are independently associated with atheromatosis progression in CKD.
Collapse
Affiliation(s)
- Milica Bozic
- Vascular and Renal Translational Research Group, Institut de Recerca Biomedica de Lleida, Lleida, Spain; and
| | - Angels Betriu
- Vascular and Renal Translational Research Group, Institut de Recerca Biomedica de Lleida, Lleida, Spain; and
| | - Marcelino Bermudez-Lopez
- Vascular and Renal Translational Research Group, Institut de Recerca Biomedica de Lleida, Lleida, Spain; and
| | - Alberto Ortiz
- Instituto de Investigacion Sanitaria Fundación Jiménez Díaz, Autonomous University of Madrid, Red de Investigación Renal del Instituto de Salud Carlos III, Madrid, Spain
| | - Elvira Fernandez
- Vascular and Renal Translational Research Group, Institut de Recerca Biomedica de Lleida, Lleida, Spain; and
| | - Jose M. Valdivielso
- Vascular and Renal Translational Research Group, Institut de Recerca Biomedica de Lleida, Lleida, Spain; and
| | - on behalf of the NEFRONA investigators
- Vascular and Renal Translational Research Group, Institut de Recerca Biomedica de Lleida, Lleida, Spain; and
- Instituto de Investigacion Sanitaria Fundación Jiménez Díaz, Autonomous University of Madrid, Red de Investigación Renal del Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
28
|
Naqvi SM, Gansau J, Buckley CT. Priming and cryopreservation of microencapsulated marrow stromal cells as a strategy for intervertebral disc regeneration. ACTA ACUST UNITED AC 2018; 13:034106. [PMID: 29380742 DOI: 10.1088/1748-605x/aaab7f] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
A challenge in using stromal cells for intervertebral disc (IVD) regeneration is their limited differentiation capacity in vivo without exogenous growth factor (GF) supplementation. Priming of stromal cells prior to transplantation may offer a feasible strategy to overcome this limitation. Furthermore, the ability to cryopreserve cells could help alleviate logistical issues associated with storage and transport. With these critical translational challenges in mind, we aimed to develop a strategy involving priming and subsequent cryopreservation of microencapsulated bone marrow stromal cells (BMSCs). In phase one, we utilised the electrohydrodynamic atomisation process to fabricate BMSC-encapsulated microcapsules that were primed with TGF-β3 for 14 d after which they were cultured for a further 21 d under basal or GF supplemented media conditions. Results showed that priming induced differentiation of BMSC microcapsules such that they synthesised significant amounts of sGAG (61.9 ± 2.0 μg and 55.3 ± 6.1 μg for low and high cell densities) and collagen (24.4 ± 1.9 μg and 55.3 ± 4.6 μg for low and high cell densities) in continued culture without GF supplementation compared to Unprimed microcapsules. Phase two of this work assessed the extracellular matrix forming capacity of Primed BMSC microcapsules over 21 d after cryopreservation. Notably, primed and cryopreserved BMSCs successfully retained the ability to synthesise both sGAG (24.8 ± 2.7 μg and 75.1 ± 11.6 μg for low and high cell densities) and collagen (26.4 ± 7.8 μg and 93.1 ± 10.2 μg for low and high cell densities) post-cryopreservation. These findings demonstrate the significant potential of priming and cryopreservation approaches for IVD repair and could possibly open new horizons for pre-designed, 'off-the-shelf' injectable therapeutics.
Collapse
Affiliation(s)
- Syeda M Naqvi
- Trinity Centre for Bioengineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Ireland. School of Engineering, Trinity College Dublin, Ireland
| | | | | |
Collapse
|
29
|
Schumacher M, Reither L, Thomas J, Kampschulte M, Gbureck U, Lode A, Gelinsky M. Calcium phosphate bone cement/mesoporous bioactive glass composites for controlled growth factor delivery. Biomater Sci 2018; 5:578-588. [PMID: 28154869 DOI: 10.1039/c6bm00903d] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Calcium phosphate (CaP) bone cements are widely used for the treatment of bone defects and have been proposed to serve as a delivery platform for therapeutic drugs, proteins and growth factors into the defect region. However, they lack sufficient porosity to allow immediate bone ingrowth and thus foster rapid integration into the bone tissue. In this study we investigated a composite prepared from a hydroxyapatite forming bone cement and mesoporous bioactive glass (MBG) granules as a potential carrier for biologically active proteins. The mechanical properties of the composite were not compromised by up to 10 wt% MBG granule addition, which can be attributed to the strong interface between the cement matrix and MBG particles, however this modification induced a significant increase in porosity within 3 weeks ageing in an aqueous liquid. The release profiles of two proteins, lysozyme and the vascular endothelial growth factor (VEGF), could be controlled when they were loaded onto MBG granules that were subsequently embedded into the cement when compared to direct loading into the cement precursor. Both proteins were also demonstrated to maintain their biologic activity during embedding and release from the composite. These findings suggest the CaP bone cement/MBG composite developed in this study as a potential delivery platform for growth factors or other bioactive substances.
Collapse
Affiliation(s)
- M Schumacher
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Hospital, Technische Universität Dresden, Dresden, Germany.
| | - L Reither
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Hospital, Technische Universität Dresden, Dresden, Germany.
| | - J Thomas
- Leibniz Institute for Solid State and Materials Research (IFW), Dresden, Germany
| | - M Kampschulte
- Department of Diagnostic and Interventional Radiology, University Hospital Giessen, Germany and Laboratory of Experimental Radiology, Justus Liebig University Giessen, Germany
| | - U Gbureck
- Department of Functional Materials in Medicine and Dentistry, University of Würzburg, Germany
| | - A Lode
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Hospital, Technische Universität Dresden, Dresden, Germany.
| | - M Gelinsky
- Centre for Translational Bone, Joint and Soft Tissue Research, Medical Faculty and University Hospital, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
30
|
Yang Z, Wan J, Pan W, Zou J. Expression of vascular endothelial growth factor in cardiac repair: Signaling mechanisms mediating vascular protective effects. Int J Biol Macromol 2018; 113:179-185. [PMID: 29462681 DOI: 10.1016/j.ijbiomac.2018.02.111] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 02/16/2018] [Accepted: 02/16/2018] [Indexed: 01/19/2023]
Abstract
The present study was aimed to investigate the vascular endothelial growth factor expression pattern in acute myocardial infarction induced rats. Serum level of vascular endothelial growth factor and its mRNA expression in myocardium were determined. Protein expression of vascular endothelial growth factor and endothelial nitric oxide synthase were measured. Serum level of vascular endothelial growth factor was increased 105.3, 260, 378.2 and 271.3% following the onset of acute myocardial infarction at 3, 6, 9 and 12days respectively. The mRNA and protein expression of vascular endothelial growth factor was substantially increased following the onset of acute myocardial infarction. Protein expression of endothelial nitric oxide synthase was increased up to 1.02 fold. Taking all these data together, it is concluded that the vascular endothelial growth factor was increased in serum and tissue and attained peak at 9th day following the onset of acute myocardial infarction. Increased vascular endothelial growth factor level in serum and tissue could increase endothelial cell proliferation and angiogenesis, and endothelial nitric oxide synthase could inhibit apoptosis and protect cardiomyocytes. In conclusion, the increased vascular endothelial growth factor expression could play an essential role in cardiac repair following the onset of acute myocardial infarction.
Collapse
Affiliation(s)
- Zefu Yang
- Department of Cardiovascular Medicine, People's Hospital of Nanhai District, Foshan City, Guangdong Province 528200, China.
| | - Jianping Wan
- Department of Cardiovascular Medicine, People's Hospital of Nanhai District, Foshan City, Guangdong Province 528200, China
| | - Wei Pan
- Department of Cardiovascular Medicine, People's Hospital of Nanhai District, Foshan City, Guangdong Province 528200, China
| | - Jun Zou
- Department of Cardiovascular Medicine, People's Hospital of Nanhai District, Foshan City, Guangdong Province 528200, China
| |
Collapse
|
31
|
Nethi SK, Barui AK, Bollu VS, Rao BR, Patra CR. Pro-angiogenic Properties of Terbium Hydroxide Nanorods: Molecular Mechanisms and Therapeutic Applications in Wound Healing. ACS Biomater Sci Eng 2017; 3:3635-3645. [DOI: 10.1021/acsbiomaterials.7b00457] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Susheel Kumar Nethi
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
- Training and Development
Complex, Academy of Scientific and Innovative Research (AcSIR), CSIR
Campus, CSIR Road, Taramani, Chennai 600113, India
| | - Ayan Kumar Barui
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
- Training and Development
Complex, Academy of Scientific and Innovative Research (AcSIR), CSIR
Campus, CSIR Road, Taramani, Chennai 600113, India
| | - Vishnu Sravan Bollu
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
- Training and Development
Complex, Academy of Scientific and Innovative Research (AcSIR), CSIR
Campus, CSIR Road, Taramani, Chennai 600113, India
| | - Bonda Rama Rao
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
- Training and Development
Complex, Academy of Scientific and Innovative Research (AcSIR), CSIR
Campus, CSIR Road, Taramani, Chennai 600113, India
| | - Chitta Ranjan Patra
- Chemical Biology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, Telangana State, India
- Training and Development
Complex, Academy of Scientific and Innovative Research (AcSIR), CSIR
Campus, CSIR Road, Taramani, Chennai 600113, India
| |
Collapse
|
32
|
Anderson EM, Silva EA, Hao Y, Martinick KD, Lewin SA, Stafford AG, Doherty EG, Wang L, Doherty EJ, Grossman PM, Mooney DJ. VEGF and IGF Delivered from Alginate Hydrogels Promote Stable Perfusion Recovery in Ischemic Hind Limbs of Aged Mice and Young Rabbits. J Vasc Res 2017; 54:288-298. [PMID: 28930755 PMCID: PMC5642984 DOI: 10.1159/000479869] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 07/28/2017] [Indexed: 12/16/2022] Open
Abstract
Biomaterial-based delivery of angiogenic growth factors restores perfusion more effectively than bolus delivery methods in rodent models of peripheral vascular disease, but the same success has not yet been demonstrated in clinically relevant studies of aged or large animals. These studies explore, in clinically relevant models, a therapeutic angiogenesis strategy for the treatment of peripheral vascular disease that overcomes the challenges encountered in previous clinical trials. Alginate hydrogels providing sustained release of vascular endothelial growth factor (VEGF) and insulin-like growth factor-1 (IGF) were injected into ischemic hind limbs in middle-aged and old mice, and also in young rabbits, as a test of the scalability of this local growth factor treatment. Spontaneous perfusion recovery diminished with increasing age, and only the combination of VEGF and IGF delivery from gels significantly rescued perfusion in middle-aged (13 months) and old (20 months) mice. In rabbits, the delivery of VEGF alone or in combination with IGF from alginate hydrogels, at a dose 2 orders of magnitude lower than the typical doses used in past rabbit studies, enhanced perfusion recovery when given immediately after surgery, or as a treatment for chronic ischemia. Capillary density measurements and angiographic analysis demonstrated the benefit of gel delivery. These data together suggest that alginate hydrogels providing local delivery of low doses of VEGF and IGF constitute a safe and effective treatment for hind-limb ischemia in clinically relevant animal models, thereby supporting the potential clinical translation of this concept.
Collapse
Affiliation(s)
- Erin M Anderson
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Eduardo A Silva
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
- Department of Biomedical Engineering, University of California, Davis, CA 95616
| | - Yibai Hao
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Kathleen D Martinick
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Sarah A Lewin
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Alexander G Stafford
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Elisabeth G Doherty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Lin Wang
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
| | - Edward J Doherty
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| | - Paul M Grossman
- Division of Cardiology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109
| | - Dave J Mooney
- School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115
| |
Collapse
|
33
|
Zhu H, Li X, Yuan M, Wan W, Hu M, Wang X, Jiang X. Intramyocardial delivery of bFGF with a biodegradable and thermosensitive hydrogel improves angiogenesis and cardio-protection in infarcted myocardium. Exp Ther Med 2017; 14:3609-3615. [PMID: 29042955 PMCID: PMC5639332 DOI: 10.3892/etm.2017.5015] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Accepted: 03/17/2017] [Indexed: 11/30/2022] Open
Abstract
Basic fibroblast growth factor (bFGF), a known angiogenic factor, may provide a potential strategy for the treatment of myocardial infarction (MI), but it is limited by a relatively short half-life. Dex-PCL-HEMA/PNIPAAm hydrogel provides a reservoir for the controlled release of growth factors. The aim of the current study was to evaluate the effects of bFGF incorporated into a Dex-PCL-HEMA/PNIPAAm hydrogel on angiogenesis and cardiac health in a rat model of acute MI, induced by coronary artery ligation. Phosphate-buffered solution (PBS group), Dex-PCL-HEMA/PNIPAAm hydrogel (Gel group), bFGF in phosphate-buffered solution (bFGF group) or bFGF in hydrogel (Gel + bFGF group) was injected into a peri-infarcted area of cardiac tissue immediately following MI. On day 30 post-surgery, cardiac function was assessed by echocardiography, apoptosis index by terminal deoxynucleotidyl transferase dUTP nick-end labeling assessment and vascular development by immunohistochemical staining. The findings demonstrated that injection of bFGF along with hydrogel induced angiogenesis, reduced collagen content, MI area and cell apoptosis and improved cardiac function compared with the injection of either bFGF or hydrogel alone. bFGF incorporated with Dex-PCL-HEMA/PNIPAAm hydrogel injection induces angiogenesis, attenuates cardiac remodeling and improves cardiac function following MI.
Collapse
Affiliation(s)
- Hongling Zhu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoyan Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Mingjie Yuan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Weiguo Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Miaoyang Hu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xiaoding Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| | - Xuejun Jiang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China.,Cardiovascular Research Institute, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, P.R. China
| |
Collapse
|
34
|
Guan Y, Cai B, Wu X, Peng S, Gan L, Huang D, Liu G, Dong L, Xiao L, Liu J, Zhang B, Cai WJ, Schaper J, Schaper W. microRNA-352 regulates collateral vessel growth induced by elevated fluid shear stress in the rat hind limb. Sci Rep 2017; 7:6643. [PMID: 28751690 PMCID: PMC5532297 DOI: 10.1038/s41598-017-06910-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Accepted: 06/20/2017] [Indexed: 12/17/2022] Open
Abstract
Although collateral vessel growth is distinctly enhanced by elevated fluid shear stress (FSS), the underlying regulatory mechanism of this process remains incompletely understood. Recent studies have shown that microRNAs (miRNAs) play a pivotal role in vascular development, homeostasis and a variety of diseases. Therefore, this study was designed to identify miRNAs involved in elevated FSS-induced collateral vessel growth in rat hind limbs. A side-to-side arteriovenous (AV) shunt was created between the distal stump of one of the bilaterally occluded femoral arteries and the accompanying vein. The miRNA array profile showed 94 differentially expressed miRNAs in FSS-stressed collaterals including miRNA-352 which was down-regulated. Infusion of antagomir-352 increased the number and proliferation of collateral vessels and promoted collateral flow restoration in a model of rat hind limb ligation. In cell culture studies, the miR-352 inhibitor increased endothelial proliferation, migration and tube formation. In addition, antagomir-352 up-regulated the expression of insulin-like growth factor II receptor (IGF2R), which may play a part in the complex pathway leading to arterial growth. We conclude that enhanced collateral vessel growth is controlled by miRNAs, among which miR-352 is a novel candidate that negatively regulates arteriogenesis, meriting additional studies to unravel the pathways leading to improved collateral circulation.
Collapse
Affiliation(s)
- Yinglu Guan
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.,Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, 77204, Houston, TX, United States
| | - Baizhen Cai
- Department of Intensive Care Unit, the 3rd Xiangya Hospital, Central South University Changsha, 410013, Hunan, P.R. China
| | - Xiaoqiong Wu
- Department of Anatomy & Neurobiology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Song Peng
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.,Department of Radiology, the 3rd Xiangya Hospital, Central South University Changsha, 410078, Hunan, P.R. China
| | - Liaoying Gan
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Da Huang
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Guangmin Liu
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Liping Dong
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Lin Xiao
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Junwen Liu
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China
| | - Bin Zhang
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.
| | - Wei-Jun Cai
- Department of Histology & Embryology, School of Basic Medicine, Central South University Changsha, 410013, Hunan, P.R. China.
| | - Jutta Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany.
| | - Wolfgang Schaper
- Max-Planck-Institute for Heart and Lung Research, Arteriogenesis Research Group, Bad Nauheim, D-61231, Germany
| |
Collapse
|
35
|
Kalayi Nia S, Ziaee S, Boroumand MA, Sotudeh Anvari M, Pourgholi L, Jalali A. The impact of vascular endothelial growth factor +405 C/G polymorphism on long-term outcome and severity of coronary artery disease. J Clin Lab Anal 2017; 31:e22066. [PMID: 27704620 PMCID: PMC6816936 DOI: 10.1002/jcla.22066] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/16/2016] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The association between genetic variations of vascular endothelial growth factor (VEGF) gene and the risk for atherosclerosis has been hypothesized. We aimed to assess the relationship between rs2010963 (+405 C/G) polymorphism and presence, severity, and outcome of coronary artery disease (CAD) in an Iranian cohort. METHODS Genotyping of VEGF rs2010963 polymorphism was performed on 520 individuals, comprising 347 patients with documented coronary artery disease based on angiography report and 173 individuals with normal coronary arteries, using the TaqMan real-time PCR method. In final, 484 subjects were followed up over a 5-year period for cardiovascular-related outcomes. RESULTS C allele of VEGF rs2010963 polymorphism was related to increase risk for CAD and also slightly to 5-year cardiovascular mortality. The 5-year survival in C and G allele subgroups were 92.3% and 94.3% in CAD group and 95.7% and 98.0% in non-CAD group, respectively. CONCLUSIONS Vascular endothelial growth factor rs2010963 polymorphism may be associated with the presence of CAD and its long-term survival, but not with its severity. To the best of our knowledge, this is the first report of genetic association between rs2010963 SNP and CAD-related death. It can be thus suggested that rs2010963 VEGF gene can be considered as a genetic risk predictor for CAD and its outcomes.
Collapse
Affiliation(s)
- Samira Kalayi Nia
- Department of Molecular PathologyTehran Heart CenterTehran University of Medical SciencesTehranIran
| | - Shayan Ziaee
- Department of Molecular PathologyTehran Heart CenterTehran University of Medical SciencesTehranIran
| | - Mohammad Ali Boroumand
- Department of Pathology and Laboratory MedicineTehran Heart CenterTehran University of Medical SciencesTehranIran
| | - Maryam Sotudeh Anvari
- Department of Pathology and Laboratory MedicineTehran Heart CenterTehran University of Medical SciencesTehranIran
| | - Leyla Pourgholi
- Department of Molecular PathologyTehran Heart CenterTehran University of Medical SciencesTehranIran
| | - Arash Jalali
- Department of BiostatisticsTehran Heart CenterTehran University of Medical SciencesTehranIran
| |
Collapse
|
36
|
Nethi SK, Nanda HS, Steele TWJ, Patra CR. Functionalized nanoceria exhibit improved angiogenic properties. J Mater Chem B 2017; 5:9371-9383. [DOI: 10.1039/c7tb01957b] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Overall schematic representation of the synthesis, characterization and proangiogenic activity of functionalized nanoceria.
Collapse
Affiliation(s)
- Susheel Kumar Nethi
- Chemical Biology Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| | - Himansu Sekhar Nanda
- School of Materials Science and Engineering
- Nanyang Technological University (NTU)
- Singapore 639798
- Singapore
| | - Terry W. J. Steele
- School of Materials Science and Engineering
- Nanyang Technological University (NTU)
- Singapore 639798
- Singapore
| | - Chitta Ranjan Patra
- Chemical Biology Division
- CSIR-Indian Institute of Chemical Technology
- Hyderabad – 500007
- India
- Academy of Scientific and Innovative Research (AcSIR)
| |
Collapse
|
37
|
Lee H, Chung HJ, Park TG. Perspectives On: Local and Sustained Delivery of Angiogenic Growth Factors. J BIOACT COMPAT POL 2016. [DOI: 10.1177/0883911506073363] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
This review emphasizes the role of angiogenesis in tissue engineering, introduces various angiogenic growth factors, and highlights current status of delivery systems for angiogenic growth factors using natural and synthetic biomaterials. A short overview of angiogenic growth factors is presented, followed by the introduction of emerging strategies for designing smart delivery carriers.
Collapse
Affiliation(s)
- Hyukjin Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Hyun Jung Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea
| | - Tae Gwan Park
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon 305-701, Korea,
| |
Collapse
|
38
|
Schwarz ER, Meven DA, Sulemanjee NZ, Kersting PH, Tussing T, Skobel EC, Hanrath P, Uretsky BF. Monocyte Chemoattractant Protein 1-Induced Monocyte Infiltration Produces Angiogenesis but Not Arteriogenesis in Chronically Infarcted Myocardium. J Cardiovasc Pharmacol Ther 2016; 9:279-89. [PMID: 15678247 DOI: 10.1177/107424840400900408] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background: Monocyte chemoattractant protein 1 (MCP-1) stimulates the invasion of monocytes into ischemic tissue with concomitant adhesion to endothelial cells. Monocyte stimulation has been shown to be involved in the induction of arteriogenesis, which is the development of functional arterioles resulting in improvement of perfusion. However, angiogenesis (newly developed capillaries contribute to improved tissue perfusion) in several models has not resulted in any improvement in blood flow. Objective: The effects of MCP-1 on potential angiogenesis and arteriogenesis as well as changes in left ventricular function were tested in a chronic infarct model in rat hearts. Methods: Anesthetized rats were subjected to open-chest ligation of the left coronary artery with subsequent myocardial infarction. After 6 weeks, animals were randomized to receive either MCP-1 (3 µL in 0.15 mL NaCl, group 1, n = 9) or saline (0.15 mL, group 2, n = 9), which was injected into the myocardium at the border zones of the infarcts. For assessment of left ventricular dimensions and global cardiac function, transthoracic two-dimensional echocardiography was performed at baseline, 6 weeks after myocardial infarction, and 4 weeks after MCP-1 or saline injection, by use of a 12-MHz pediatric transducer. For light microscopic analysis, myocardial tissue was stained with Elastica-van-Giesson and von Willebrand factor for blood vessels and endothelial cells, respectively. In a subset of animals, hearts were excised 24 hours after MCP-1 administration (n = 4) or saline administration (n = 4) for assessment of monocyte infiltration by immunohistologic staining of the CD31 antigen. Results: Left ventricular dimensions and ejection fraction changed after coronary occlusion (from 60.4% ± 2.85% to 24.8% ± 5.01% ejection fraction in group 1, and from 58.4% ±2.06% to 26.3% ± 4.3% ejection fraction in group 2 at 6 weeks, P < .005) without any further change 4 weeks after treatment (ejection fraction in group 1, 26.3% ± 2.7%, ejection fraction in group 2, 25.0% ± 5.18%). The MCP-1 group resulted in 390.6 ± 10.36 endothelial cells compared with 285.2 ± 13.56 in group 2 ( P < .005) at the injection site. Monocyte infiltration was observed at the MCP-1 injection site with an increase in capillary growth (angiogenesis). However, there was no difference in the number of arteriolar structures between animals treated with MCP-1 and saline animals (group 1, 19.0 ± 1.52 vs group 2,16.4 ± 0.68, P > .05). Conclusion: A single intramyocardial injection of MCP-1 into the infarct border zone resulted in neo-angiogenesis and monocyte infiltration but not arteriogenesis in the rat heart. There was no functional change of chronically infarcted myocardium in the present model.
Collapse
Affiliation(s)
- Ernst R Schwarz
- Department of Cardiology, Rheinisch-Westfaelisch Technische Hochschule University Hospital Aachen, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Ye L, Haider HK, Jiang SJ, Sim EKW. Therapeutic Angiogenesis Using Vascular Endothelial Growth Factor. Asian Cardiovasc Thorac Ann 2016; 12:173-81. [PMID: 15213090 DOI: 10.1177/021849230401200221] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Therapeutic angiogenesis using vascular endothelial growth factor can reduce tissue ischemia by simulating the natural process of angiogenesis. Vascular endothelial growth factor not only stimulates endothelial cells to proliferate and migrate, but also mobilizes endothelial progenitor cells and achieves vascular protection. Besides direct administration of angiogenic proteins, plasmids and viral vectors carrying angiogenic genes have been used. Animal experiments have shown promise with evidence of neovascularization and improved perfusion in the target myocardium. Initial phase I and II clinical trials results are encouraging and reflect the potential success of therapeutic angiogenesis as a clinical modality for the treatment of ischemic heart disease. This review discusses the role of vascular endothelial growth factor in therapeutic angiogenesis, along with the problems and considerations of this approach as a treatment strategy.
Collapse
Affiliation(s)
- Lei Ye
- Department of Cardiothoracic and Vascular Surgery, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
40
|
Zhao H, Huang H, Lin S. Chemical approaches to angiogenesis in development and regeneration. Methods Cell Biol 2016; 134:369-76. [PMID: 27312498 DOI: 10.1016/bs.mcb.2016.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Vascular endothelial cells are essential building blocks of angiogenesis, which is required for normal embryonic development and tissue regeneration. In this chapter, we describe how to use transgenic zebrafish embryos expressing vascular-specific green fluorescent protein to evaluate differentiation, growth, and morphogenesis of endothelial cells. When combined with instrument automation and computational analysis, this method allows high-throughput screening for biologically active small chemical molecules that are effective in promoting angiogenesis. These molecules can be validated in mammalian endothelial cell differentiation and proliferation assays. These studies provide new reagents and therapeutic candidates for regenerative medicine studies.
Collapse
Affiliation(s)
- H Zhao
- University of California Los Angeles, Los Angeles, CA, United States; Jinan University, Guangzhou, China
| | - H Huang
- University of California Los Angeles, Los Angeles, CA, United States
| | - S Lin
- University of California Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
41
|
Kirby GTS, White LJ, Steck R, Berner A, Bogoevski K, Qutachi O, Jones B, Saifzadeh S, Hutmacher DW, Shakesheff KM, Woodruff MA. Microparticles for Sustained Growth Factor Delivery in the Regeneration of Critically-Sized Segmental Tibial Bone Defects. MATERIALS 2016; 9:ma9040259. [PMID: 28773384 PMCID: PMC5502923 DOI: 10.3390/ma9040259] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/18/2016] [Accepted: 03/18/2016] [Indexed: 11/16/2022]
Abstract
This study trialled the controlled delivery of growth factors within a biodegradable scaffold in a large segmental bone defect model. We hypothesised that co-delivery of vascular endothelial growth factor (VEGF) and platelet derived growth factor (PDGF) followed by bone morphogenetic protein-2 (BMP-2) could be more effective in stimulating bone repair than the delivery of BMP-2 alone. Poly(lactic-co-glycolic acid) (PLGA ) based microparticles were used as a delivery system to achieve a controlled release of growth factors within a medical-grade Polycaprolactone (PCL) scaffold. The scaffolds were assessed in a well-established preclinical ovine tibial segmental defect measuring 3 cm. After six months, mechanical properties and bone tissue regeneration were assessed. Mineralised bone bridging of the defect was enhanced in growth factor treated groups. The inclusion of VEGF and PDGF (with BMP-2) had no significant effect on the amount of bone regeneration at the six-month time point in comparison to BMP-2 alone. However, regions treated with VEGF and PDGF showed increased vascularity. This study demonstrates an effective method for the controlled delivery of therapeutic growth factors in vivo, using microparticles.
Collapse
Affiliation(s)
- Giles T S Kirby
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
- School of Pharmacy, University Park, The University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Lisa J White
- School of Pharmacy, University Park, The University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Roland Steck
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
| | - Arne Berner
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
- Department of Trauma Surgery, University of Regensburg, Regensburg 93164, Germany.
| | - Kristofor Bogoevski
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
| | - Omar Qutachi
- School of Pharmacy, University Park, The University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Brendan Jones
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
| | - Siamak Saifzadeh
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
| | - Dietmar W Hutmacher
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
| | - Kevin M Shakesheff
- School of Pharmacy, University Park, The University of Nottingham, University Park, Nottingham NG7 2RD, UK.
| | - Maria A Woodruff
- Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisban, QLD 4006, Australia.
| |
Collapse
|
42
|
Guan Y, Cai B, Liu Z, Ye F, Deng P, Cai WJ, Schaper J, Schaper W. The Formation of Aberrant Collateral Vessels during Coronary Arteriogenesis in Dog Heart. Cells Tissues Organs 2016; 201:118-29. [PMID: 26796132 DOI: 10.1159/000442381] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2015] [Indexed: 11/19/2022] Open
Abstract
We previously reported excessive growth of collateral vessels in the dog heart during arteriogenesis induced by implantation of an ameroid constrictor around the circumflex branch of the left coronary artery. In the present study, using histology and immunocofocal microscopy, we further investigated how these aberrant collateral vessels form. By comparison with mature collateral vessels the following findings were made: perivascular space was very narrow where damage of the perivascular myocardium occurred; the neointima was very thick, resulting in a very small lumen; elastica van Gieson staining revealed the absence of the internal elastic lamina and of elastic fibers in the adventitia, but abundant collagen in the adventitia as well as in the neointima; smooth muscle cells of the neointima expressed less α-SM actin and little desmin; expression of the fibroblast growth factors aFGF, bFGF and platelet-derived growth factor (PDGF)-AB was observed mainly in the endothelial cells and abluminal region, but transforming growth factor-β1 was only present in the adventitia and damaged myocardium; angiogenesis in the neointima was observed in some collateral vessels expressing high levels of eNOS, and cell proliferation was mainly present in the abluminal region, but apoptosis was in the deep neointima. In conclusion, these data for the first time reveal that the formation of the aberrant collateral vessels in the dog heart involves active extracellular proteolysis and a special expression profile of growth factors, eNOS, cell proliferation and apoptosis. The finding of a narrow perivascular space and perivascular myocardial damage suggests that anatomical constraint is most likely the cause for exacerbated inward remodeling in aberrant collateral vessels in dog heart.
Collapse
Affiliation(s)
- Yinglu Guan
- Department of Histology and Embryology, School of Basic Medicine, Central South University Changsha, Hunan, PR China
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Namdee K, Sobczynski DJ, Onyskiw PJ, Eniola-Adefeso O. Differential Impact of Plasma Proteins on the Adhesion Efficiency of Vascular-Targeted Carriers (VTCs) in Blood of Common Laboratory Animals. Bioconjug Chem 2015; 26:2419-28. [PMID: 26505780 PMCID: PMC4866610 DOI: 10.1021/acs.bioconjchem.5b00474] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Vascular-targeted carrier (VTC) interaction with human plasma is known to reduce targeted adhesion efficiency in vitro. However, the role of plasma proteins on the adhesion efficiency of VTCs in laboratory animals remains unknown. Here, in vitro blood flow assays are used to explore the effects of plasma from mouse, rabbit, and porcine on VTC adhesion. Porcine blood exhibited a strong negative plasma effect on VTC adhesion while no significant plasma effect was found with rabbit and mouse blood. A brush density poly(ethylene glycol) (PEG) on VTCs was effective at improving adhesion of microsized, but not nanosized, VTCs in porcine blood. Overall, the results suggest that porcine models, as opposed to mouse, can serve as better models in preclinical research for predicting the in vivo functionality of VTCs for use in humans. These considerations hold great importance for the design of various pharmaceutical products and development of reliable drug delivery systems.
Collapse
Affiliation(s)
| | | | - Peter J. Onyskiw
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109
| | - Omolola Eniola-Adefeso
- Department of Chemical Engineering, University of Michigan, 2800 Plymouth Road, Ann Arbor, MI, 48109
| |
Collapse
|
44
|
Novel biodegradable polymers for local growth factor delivery. Eur J Pharm Biopharm 2015; 97:318-28. [DOI: 10.1016/j.ejpb.2015.06.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 06/06/2015] [Accepted: 06/09/2015] [Indexed: 01/09/2023]
|
45
|
Yang Y, Shi C, Hou X, Zhao Y, Chen B, Tan B, Deng Z, Li Q, Liu J, Xiao Z, Miao Q, Dai J. Modified VEGF targets the ischemic myocardium and promotes functional recovery after myocardial infarction. J Control Release 2015; 213:27-35. [PMID: 26144351 DOI: 10.1016/j.jconrel.2015.06.036] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2015] [Revised: 06/25/2015] [Accepted: 06/29/2015] [Indexed: 02/07/2023]
Abstract
Vascular endothelial growth factor (VEGF) promotes angiogenesis and improves cardiac function after myocardial infarction (MI). However, the non-targeted delivery of VEGF decreases its therapeutic efficacy due to an insufficient local concentration in the ischemic myocardium. In this study, we used a specific peptide to modify VEGF and determined that this modified VEGF (IMT-VEGF) localized to the ischemic myocardium through intravenous injection by interacting with cardiac troponin I (cTnI). When IMT-VEGF was used to mediate cardiac repair in a rat model of ischemia-reperfusion (I-R) injury, we observed a decreased scar size, enhanced angiogenesis and improved cardiac function. Moreover, an alternative treatment using the repeated administration of a low-dose IMT-VEGF also promoted angiogenesis and functional recovery. The therapeutic effects of IMT-VEGF were further confirmed in a pig model of MI as the result of the conserved properties of its interacting protein, cTnI. These results suggest a promising therapeutic strategy for MI based on the targeted delivery of IMT-VEGF.
Collapse
Affiliation(s)
- Yun Yang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Graduate School, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China
| | - Chunying Shi
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute for Translational Medicine, College of Medicine, Qingdao University, 308 Ningxia Road, Qingdao, 266021, China
| | - Xianglin Hou
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Bo Tan
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou, 215123, China
| | - Zongwu Deng
- Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou, 215123, China
| | - Qingguo Li
- Department of Cardiothoracic Surgery, the affiliated Drum Tower Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, China
| | - Jianzhou Liu
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Peking Union Medical College, 1 Shuaifuyuan, Beijing, 100730, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China
| | - Qi Miao
- Department of Cardiac Surgery, Peking Union Medical College Hospital, Peking Union Medical College, 1 Shuaifuyuan, Beijing, 100730, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 3 Nanyitiao, Zhongguancun, Beijing, 100190, China; Institute of Combined Injury, State Key Laboratory of Trauma, Burns and Combined Injury, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Road, Chongqing, 400038, China.
| |
Collapse
|
46
|
Effect of variation in hemorheology between human and animal blood on the binding efficacy of vascular-targeted carriers. Sci Rep 2015; 5:11631. [PMID: 26113000 PMCID: PMC4481524 DOI: 10.1038/srep11631] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Accepted: 06/01/2015] [Indexed: 11/10/2022] Open
Abstract
Animal models are extensively used to evaluate the in vivo functionality of novel drug delivery systems (DDS). However, many variations likely exist in vivo between the animals and human physiological environment that significantly alter results obtained with animal models relative to human system. To date, it is not clear if the variation in hemorheology and hemodynamics between common animal and human models affect the functionality of DDS. This study investigates the role of hemorheology of humans and various animal models in dictating the binding efficiency of model vascular-targeted carriers (VTCs) to the wall in physiological blood flows. Specifically, the adhesion of sLeA-coated nano- and micro-spheres to inflamed endothelial cells monolayers were conducted via a parallel plate flow chamber assay with steady and disturbed red blood cells (RBCs)-in-buffer and whole blood flows of common animal models. Our results suggest that the ratio of carrier size to RBC size dictate particle binding in blood flow. Additionally, the presence of white blood cells affects the trend of particle adhesion depending on the animal species. Overall, this work sheds light on some deviation in VTC vascular wall interaction results obtained with in vivo animal experimentation from expected outcome and efficiency in vivo in human.
Collapse
|
47
|
McDonald AI, Iruela-Arispe ML. Healing arterial ulcers: Endothelial lining regeneration upon vascular denudation injury. Vascul Pharmacol 2015; 72:9-15. [PMID: 26093336 DOI: 10.1016/j.vph.2015.06.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2015] [Accepted: 06/16/2015] [Indexed: 12/20/2022]
Abstract
Thrombosis and restenosis are the most prevalent late complications of coronary artery stenting. Current standards of clinical care focus on prevention of smooth muscle cell proliferation by the use of drug-eluting stents able to release anti-proliferative drugs. Unfortunately, these drugs also block endothelial cell proliferation and, in this manner, prevent recovery of endothelial cell coverage. Continued lack of endothelial repair leaves the root cause of thrombosis and restenosis unchanged, creating a vicious cycle where drug-mediated prevention of restenosis simultaneously implies promotion of thrombosis. In this issue of Vascular Pharmacology, Hussner and colleagues provide in vitro evidence and a mechanistic basis for the use of atorvastatin in stents as a way to bypass this roadblock. Here we review the pathological mechanisms and therapeutic approaches to restore flow in occluded arteries. We argue that rational design of drug eluting stents should focus on specific inhibition of smooth muscle cell proliferation with concurrent stimulation of endothelial regeneration. We comment on the current poor understanding of the cellular and molecular regulation of endothelial cell proliferation in the context of a functional artery, and on the pitfalls of extrapolating from the well-studied process of neovascularization by sprouting vessel formation.
Collapse
Affiliation(s)
- Austin I McDonald
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA
| | - M Luisa Iruela-Arispe
- Molecular Biology Institute, University of California, Los Angeles, CA 90095, USA; Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, CA 90095, USA..
| |
Collapse
|
48
|
Nethi SK, Veeriah V, Barui AK, Rajendran S, Mattapally S, Misra S, Chatterjee S, Patra CR. Investigation of molecular mechanisms and regulatory pathways of pro-angiogenic nanorods. NANOSCALE 2015; 7:9760-9770. [PMID: 25963768 PMCID: PMC4724197 DOI: 10.1039/c5nr01327e] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Angiogenesis, a process involving the growth of new blood vessels from the pre-existing vasculature, plays a crucial role in various pathophysiological conditions. We have previously demonstrated that europium hydroxide [Eu(III)(OH)3] nanorods (EHNs) exhibit pro-angiogenic properties through the generation of reactive oxygen species (ROS) and mitogen activated protein kinase (MAPK) activation. Considering the enormous implication of angiogenesis in cardiovascular diseases (CVDs) and cancer, it is essential to understand in-depth molecular mechanisms and signaling pathways in order to develop the most efficient and effective alternative treatment strategy for CVDs. However, the exact underlying mechanism and cascade signaling pathways behind the pro-angiogenic properties exhibited by EHNs still remain unclear. Herein, we report for the first time that the hydrogen peroxide (H2O2), a redox signaling molecule, generated by these EHNs activates the endothelial nitric oxide synthase (eNOS) that promotes the nitric oxide (NO) production in a PI3K (phosphoinositide 3-kinase)/Akt dependent manner, eventually triggering angiogenesis. We intensely believe that the investigation and understanding of the in-depth molecular mechanism and signaling pathways of EHNs induced angiogenesis will help us in developing an effective alternative treatment strategy for cardiovascular related and ischemic diseases where angiogenesis plays an important role.
Collapse
Affiliation(s)
- Susheel Kumar Nethi
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), 2 Rafi Marg, New Delhi, India
| | - Vimal Veeriah
- Vascular Biology Lab, Life Sciences Division, AU-KBC Research Centre, Anna University, Chennai, Tamil Nadu, India
| | - Ayan Kumar Barui
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), 2 Rafi Marg, New Delhi, India
| | - Saranya Rajendran
- Vascular Biology Lab, Life Sciences Division, AU-KBC Research Centre, Anna University, Chennai, Tamil Nadu, India
| | - Saidulu Mattapally
- Medicinal Chemistry and Pharmacology Division, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
| | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Suvro Chatterjee
- Vascular Biology Lab, Life Sciences Division, AU-KBC Research Centre, Anna University, Chennai, Tamil Nadu, India
- Department of Biotechnology, Anna University, Chennai, India
| | - Chitta Ranjan Patra
- Biomaterials Group, CSIR-Indian Institute of Chemical Technology, Uppal Road, Tarnaka, Hyderabad 500007, India
- Academy of Scientific and Innovative Research (AcSIR), 2 Rafi Marg, New Delhi, India
| |
Collapse
|
49
|
Yu Y, Chen J, Chen R, Cao L, Tang W, Lin D, Wang J, Liu C. Enhancement of VEGF-Mediated Angiogenesis by 2-N,6-O-Sulfated Chitosan-Coated Hierarchical PLGA Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2015; 7:9982-9990. [PMID: 25905780 DOI: 10.1021/acsami.5b02324] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Rapid and controlled vascularization within scaffolds remains one of the key limitations in tissue engineering applications. This study describes the fabrication and characterization of 2-N,6-O-sulfated chitosan (26SCS)-coated hierarchical scaffold composed of poly(lactic-co-glycolic acid) (PLGA) microspheres, as a desirable vehicle for vascular endothelial growth factor (VEGF) delivery and consequent angiogenic boosting in vitro. Owing to the hierarchical porous structure and high affinity between VEGF and 26SCS, the 26SCS-modified PLGA (S-PLGA) scaffold possesses excellent entrapment and sustained release of VEGF. Using human umbilical vein endothelial cells (HUVECs) as a cell model, the VEGF-loaded S-PLGA scaffold shows desirable cell viability and attachment. The bioactivity of released VEGF is validated by intracellular nitric oxide secretion and capillary tube formation, demonstrating the improved capacity of VEGF-mediated pro-angiogenesis ascribed to 26SCS incorporation. Such a strategy will afford an effective method to prepare a scaffold with promoted angiogenesis.
Collapse
Affiliation(s)
- Yuanman Yu
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Jie Chen
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Rui Chen
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Lingyan Cao
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Wei Tang
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Dan Lin
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Jing Wang
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Changsheng Liu
- †The State Key Laboratory of Bioreactor Engineering, ‡Key Laboratory for Ultrafine Materials of Ministry of Education, §Engineering Research Center for Biomedical Materials of Ministry of Education, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| |
Collapse
|
50
|
Chung HJ, Kim JT, Kim HJ, Kyung HW, Katila P, Lee JH, Yang TH, Yang YI, Lee SJ. Epicardial delivery of VEGF and cardiac stem cells guided by 3-dimensional PLLA mat enhancing cardiac regeneration and angiogenesis in acute myocardial infarction. J Control Release 2015; 205:218-30. [DOI: 10.1016/j.jconrel.2015.02.013] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 01/25/2015] [Accepted: 02/04/2015] [Indexed: 02/01/2023]
|