1
|
Gachi MZ, Solouk A, Shafieian M, Daemi H. Chemical structure of antibiotics determines their release rate from drug-loaded poly(vinyl alcohol)/sodium sulfated alginate nanofibrous wound dressings. Int J Biol Macromol 2025; 307:141669. [PMID: 40032114 DOI: 10.1016/j.ijbiomac.2025.141669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 02/16/2025] [Accepted: 02/28/2025] [Indexed: 03/05/2025]
Abstract
Antibiotics are widely used for treatment of infected wounds; however, their application through a local and controlled release system may cause more effectiveness and fewer side-effects. In this study, we fabricated drug-loaded poly(vinyl alcohol)/sodium sulfated alginate (PVA/SSA) nanofibrous mats incorporating cationic antibiotic drugs, i.e., salts of gentamicin, tetracycline, ciprofloxacin and minocycline, and examined their physicochemical and biological properties. The results of FTIR spectroscopy showed that cationic drugs have different interactions with carboxylate and sulfate functional groups of SSA depending on their chemical structure. Furthermore, the results of viscometry and conductivity analyses of the solutions revealed that the solutions with drugs with more electrical charge or/and higher functional groups resulted in a lower viscosity and conductivity compared to other drugs, due to the ability to form more hydrogen bonds. The release profiles of drug-loaded nanofibrous mats showed a burst release and then, a sustained release for 5 days, where the burst release of tetracycline (30.0 ± 0.3 %) from crosslinked mats was noticeably lower than other drugs. Biological assays confirmed the cytocompatibility, antibacterial activity and non-hemolytic behavior of all drug-loaded mats. Finally, ciprofloxacin-loaded nanofibrous mat was used as wound dressing for full-thickness wounds on rats and its efficacy was confirmed.
Collapse
Affiliation(s)
- Maryam Zare Gachi
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Atefeh Solouk
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Mehdi Shafieian
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Biomaterials, ZFZ Chemical Company, Tehran, Iran.
| |
Collapse
|
2
|
Czechowicz P, Więch-Walów A, Sławski J, Collawn JF, Bartoszewski R. Old drugs, new challenges: reassigning drugs for cancer therapies. Cell Mol Biol Lett 2025; 30:27. [PMID: 40038587 PMCID: PMC11881393 DOI: 10.1186/s11658-025-00710-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/24/2025] [Indexed: 03/06/2025] Open
Abstract
The "War on Cancer" began with the National Cancer Act of 1971 and despite more than 50 years of effort and numerous successes, there still remains much more work to be done. The major challenge remains the complexity and intrinsic polygenicity of neoplastic diseases. Furthermore, the safety of the antitumor therapies still remains a concern given their often off-target effects. Although the amount of money invested in research and development required to introduce a novel FDA-approved drug has continuously increased, the likelihood for a new cancer drug's approval remains limited. One interesting alternative approach, however, is the idea of repurposing of old drugs, which is both faster and less costly than developing new drugs. Repurposed drugs have the potential to address the shortage of new drugs with the added benefit that the safety concerns are already established. That being said, their interactions with other new drugs in combination therapies, however, should be tested. In this review, we discuss the history of repurposed drugs, some successes and failures, as well as the multiple challenges and obstacles that need to be addressed in order to enhance repurposed drugs' potential for new cancer therapies.
Collapse
Affiliation(s)
- Paulina Czechowicz
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Anna Więch-Walów
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - Jakub Sławski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland
| | - James F Collawn
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, USA
| | - Rafal Bartoszewski
- Department of Biophysics, Faculty of Biotechnology, University of Wroclaw, F. Joliot-Curie 14a Street, 50-383, Wroclaw, Poland.
| |
Collapse
|
3
|
Mssillou I, Amrati FEZ, Saghrouchni H, El Abdali Y, Lefrioui Y, Batiha GES, Giesy JP, Aboul-Soud MAM, Hassani R, Khalid A, Bousta D. Recent advances in the use of essential oils and their nanoformulations for wound treatment. Burns 2025; 51:107260. [PMID: 39522139 DOI: 10.1016/j.burns.2024.08.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 08/14/2024] [Accepted: 08/31/2024] [Indexed: 11/16/2024]
Abstract
Despite progress in medical and surgical treatments of wounds, bioactive compounds still offer an effective and safe approach to accelerate wound healing (WH). In this review, recent results of studies on WH by essential oils (EOs) and their terpenoids are reported. Mechanisms of action of these substances and their possible use in drug delivery systems (DDSs) for WH are discussed. EOs of 38 species from 16 families have been evaluated for their potential to treat wounds. Lamiaceae was the most representative family with 10 species, followed by Myrtaceae and Asteraceae. EOs improve WH by acting as anti-inflammatory, antioxidant, and antimicrobial agents. Some other EOs were involved by increasing expression of transforming growth factor (TGF), inhibition of several factors, including plasminogen activator inhibitor-1 (PAI-1), substitution of type III collagen by type I collagen, and up-regulation of insulin-like growth factor-1 (IGF-1), fibroblast growth factor 2 (FGF-2), and vascular endothelial growth factor (VEGF). These mechanisms improved repair of cells and increased proliferation. Alternatively, DDSs based on nanomaterials (NMs) used to carry EOs for WH are mainly based on nanoparticles (NPs), microparticles (MPs) and scaffolds. There is much evidence that EOs can promote WH. Advancement of nanotechnology in recent years has contributed to improving use of EO with DDSs in WH management. However, some limitations need to be addressed to achieve the translation of this technology into clinical applications for wound treatment.
Collapse
Affiliation(s)
- Ibrahim Mssillou
- Laboratory of Natural Substances, Pharmacology, Environment, Modeling, Health & Quality of Life (SNAMOPEQ), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco.
| | - Fatima Ez-Zahra Amrati
- Laboratory of Cell Biology and Molecular Genetics (LBCGM), Department of Biology, Faculty of Sciences, Ibn Zohr University, Agadir, Souss Massa, Morocco
| | - Hamza Saghrouchni
- Department of Biotechnology, Institute of Natural and Applied Sciences, Çukurova University, 01330 Balcalı/Sarıçam, Adana, Turkey
| | - Youness El Abdali
- Laboratory of Biotechnology, Health, Agrofood and Environment (LBEAS), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
| | - Youssra Lefrioui
- Laboratory of Biotechnology, Health, Agrofood and Environment (LBEAS), Faculty of Sciences Dhar El Mahraz, Sidi Mohamed Ben Abdellah University, Fez 30000, Morocco
| | - Gaber El-Saber Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour 22511 Albeheira, Egypt
| | - John P Giesy
- Department of Veterinary Biomedical Sciences and Toxicology Centre, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK S7N 5B4, Canada; Department of Integrative Biology and Center for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA; Department of Environmental Sciences, Baylor University, Waco 76706, USA
| | - Mourad A M Aboul-Soud
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Rym Hassani
- Environment and Nature Research Centre, Jazan University, P. O. Box 114, Jazan 45142, Saudi Arabia
| | - Asaad Khalid
- Health Research Center, Jazan University, P.O. Box, 114, Jazan 45142, Saudi Arabia.
| | - Dalila Bousta
- National Agency of Medicinal and Aromatic Plants, 34025 Taounate, Morocco
| |
Collapse
|
4
|
Rodrigues FAP, Oliveira CS, Sá SC, Tavaria FK, Lee SJ, Oliveira AL, Costa JB. Molecules in Motion: Unravelling the Dynamics of Vascularization Control in Tissue Engineering. Macromol Biosci 2024; 24:e2400139. [PMID: 39422632 DOI: 10.1002/mabi.202400139] [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: 03/25/2024] [Revised: 08/14/2024] [Indexed: 10/19/2024]
Abstract
Significant progress has been made in tissue engineering (TE), aiming at providing personalized solutions and overcoming the current limitations of traditional tissue and organ transplantation. 3D bioprinting has emerged as a transformative technology in the field, able to mimic key properties of the natural architecture of the native tissues. However, most successes in the area are still limited to avascular or thin tissues due to the difficulties in controlling the vascularization of the engineered tissues. To address this issue, several molecules, biomaterials, and cells with pro- and anti-angiogenic potential have been intensively investigated. Furthermore, different bioreactors capable to provide a dynamic environment for in vitro vascularization control have been also explored. The present review summarizes the main molecules and TE strategies used to promote and inhibit vascularization in TE, as well as the techniques used to deliver them. Additionally, it also discusses the current challenges in 3D bioprinting and in tissue maturation to control in vitro/in vivo vascularization. Currently, this field of investigation is of utmost importance and may open doors for the design and development of more precise and controlled vascularization strategies in TE.
Collapse
Affiliation(s)
- Francisco A P Rodrigues
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Cláudia S Oliveira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Simone C Sá
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Freni K Tavaria
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Ana L Oliveira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - João B Costa
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| |
Collapse
|
5
|
K Karunakar K, Cheriyan BV, R K, M G, B A. "Therapeutic advancements in nanomedicine: The multifaceted roles of silver nanoparticles". BIOTECHNOLOGY NOTES (AMSTERDAM, NETHERLANDS) 2024; 5:64-79. [PMID: 39416696 PMCID: PMC11446369 DOI: 10.1016/j.biotno.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 05/31/2024] [Accepted: 05/31/2024] [Indexed: 10/19/2024]
Abstract
Nanotechnology has the advantages of enhanced bioactivity, reduced toxicity, target specificity, and sustained release and NPs can penetrate cell membranes. The small size of silver nanoparticles, AgNPs, large surface area, and unique physicochemical properties contribute to cell lysis and increased permeability of cell membranes used in the field of biomedicine. Functional precursors integrate with phytochemicals to create distinctive therapeutic properties and the stability of the nanoparticles can be enhanced by Surface coatings and encapsulation methods, The current study explores the various synthesis methods and characterization techniques of silver nanoparticles (AgNPs) and highlights their intrinsic activity in therapeutic applications, Anti-cancer activity noted at a concentration range of 5-50 μg/ml and angiogenesis is mitigated at a dosage range of 10-50 μg/ml, Diabetes is controlled within the same concentration. Wound healing is improved at concentrations of 10-50 μg/ml and with a typical range of 10-08 μg/ml for bacteria with antimicrobial capabilities. Advancement of silver nanoparticles with a focus on the future use of AgNPs-coated wound dressings and medical devices to decrease the risk of infection. Chemotherapeutic drugs can be administered by AgNPs, which reduces adverse effects and an improvement in treatment outcomes. AgNPs have been found to improve cell proliferation and differentiation, making them beneficial for tissue engineering and regenerative medicine. Our study highlights emerging patterns and developments in the field of medicine, inferring potential future paths.
Collapse
Affiliation(s)
- Karthik K Karunakar
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Binoy Varghese Cheriyan
- Department of Pharmaceutical Chemistry, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Krithikeshvaran R
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Gnanisha M
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| | - Abinavi B
- Department of Pharmacy Practice, Saveetha College of Pharmacy, Saveetha Institute of Medical and Technical Sciences, Chennai, 602105, TN, India
| |
Collapse
|
6
|
Senrung A, Tripathi T, Aggarwal N, Janjua D, Yadav J, Chaudhary A, Chhokar A, Joshi U, Bharti AC. Phytochemicals Showing Antiangiogenic Effect in Pre-clinical Models and their Potential as an Alternative to Existing Therapeutics. Curr Top Med Chem 2024; 24:259-300. [PMID: 37867279 DOI: 10.2174/0115680266264349231016094456] [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: 05/27/2023] [Revised: 07/25/2023] [Accepted: 08/10/2023] [Indexed: 10/24/2023]
Abstract
Angiogenesis, the formation of new blood vessels from a pre-existing vascular network, is an important hallmark of several pathological conditions, such as tumor growth and metastasis, proliferative retinopathies, including proliferative diabetic retinopathy and retinopathy of prematurity, age-related macular degeneration, rheumatoid arthritis, psoriasis, and endometriosis. Putting a halt to pathology-driven angiogenesis is considered an important therapeutic strategy to slow down or reduce the severity of pathological disorders. Considering the attrition rate of synthetic antiangiogenic compounds from the lab to reaching the market due to severe side effects, several compounds of natural origin are being explored for their antiangiogenic properties. Employing pre-clinical models for the evaluation of novel antiangiogenic compounds is a promising strategy for rapid screening of antiangiogenic compounds. These studies use a spectrum of angiogenic model systems that include HUVEC two-dimensional culture, nude mice, chick chorioallantoic membrane, transgenic zebrafish, and dorsal aorta from rats and chicks, depending upon available resources. The present article emphasizes the antiangiogenic activity of the phytochemicals shown to exhibit antiangiogenic behavior in these well-defined existing angiogenic models and highlights key molecular targets. Different models help to get a quick understanding of the efficacy and therapeutics mechanism of emerging lead molecules. The inherent variability in assays and corresponding different phytochemicals tested in each study prevent their immediate utilization in clinical studies. This review will discuss phytochemicals discovered using suitable preclinical antiangiogenic models, along with a special mention of leads that have entered clinical evaluation.
Collapse
Affiliation(s)
- Anna Senrung
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Neuropharmacology and Drug Delivery Laboratory, Daulat Ram College, University of Delhi, Delhi, India
| | - Tanya Tripathi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Nikita Aggarwal
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Divya Janjua
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Joni Yadav
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Apoorva Chaudhary
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Arun Chhokar
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
- Deshbandhu College, University of Delhi, Delhi, India
| | - Udit Joshi
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| | - Alok Chandra Bharti
- Department of Zoology, Molecular Oncology Laboratory, University of Delhi (North Campus), Delhi, 110007, India
| |
Collapse
|
7
|
Davodabadi F, Sajjadi SF, Sarhadi M, Mirghasemi S, Nadali Hezaveh M, Khosravi S, Kamali Andani M, Cordani M, Basiri M, Ghavami S. Cancer chemotherapy resistance: Mechanisms and recent breakthrough in targeted drug delivery. Eur J Pharmacol 2023; 958:176013. [PMID: 37633322 DOI: 10.1016/j.ejphar.2023.176013] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 08/28/2023]
Abstract
Conventional chemotherapy, one of the most widely used cancer treatment methods, has serious side effects, and usually results in cancer treatment failure. Drug resistance is one of the primary reasons for this failure. The most significant drawbacks of systemic chemotherapy are rapid clearance from the circulation, the drug's low concentration in the tumor site, and considerable adverse effects outside the tumor. Several ways have been developed to boost neoplasm treatment efficacy and overcome medication resistance. In recent years, targeted drug delivery has become an essential therapeutic application. As more mechanisms of tumor treatment resistance are discovered, nanoparticles (NPs) are designed to target these pathways. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation. Nano-drugs have been increasingly employed in medicine, incorporating therapeutic applications for more precise and effective tumor diagnosis, therapy, and targeting. Many benefits of NP-based drug delivery systems in cancer treatment have been proven, including good pharmacokinetics, tumor cell-specific targeting, decreased side effects, and lessened drug resistance. As more mechanisms of tumor treatment resistance are discovered, NPs are designed to target these pathways. At the moment, this innovative technology has the potential to bring fresh insights into cancer therapy. Therefore, understanding the limitations and challenges of this technology is critical for nanocarrier evaluation.
Collapse
Affiliation(s)
- Fatemeh Davodabadi
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Seyedeh Fatemeh Sajjadi
- School of Biological Science, Institute for Research in Fundamental Sciences (IPM), Tehran, Iran.
| | - Mohammad Sarhadi
- Cellular and Molecular Research Center, Research Institute of Cellular and Molecular Sciences in Infectious Diseases, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Shaghayegh Mirghasemi
- Department of Chemistry, Science and Research Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Nadali Hezaveh
- Department of Chemical Engineering, Isfahan University of Technology, Isfahan, 84156-83111, Iran.
| | - Samin Khosravi
- Department of Cellular and Molecular Biology, Faculty of Biological Sciences, North Tehran Branch, Islamic Azad University, Tehran, Iran.
| | - Mahdieh Kamali Andani
- Department of Biology, Faculty of Basic Science, Payame Noor University, Tehran, Iran.
| | - Marco Cordani
- Department of Biochemistry and Molecular Biology, Faculty of Biological Sciences, Complutense University of Madrid, Madrid, Spain; Instituto de Investigaciones Sanitarias San Carlos (IdISSC), Madrid, Spain.
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| | - Saeid Ghavami
- Academy of Silesia, Faculty of Medicine, Rolna 43, 40-555. Katowice, Poland; Research Institute of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P5, Canada; Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 3P5, Canada.
| |
Collapse
|
8
|
Malektaj H, Nour S, Imani R, Siadati MH. Angiogenesis induction as a key step in cardiac tissue Regeneration: From angiogenic agents to biomaterials. Int J Pharm 2023; 643:123233. [PMID: 37460050 DOI: 10.1016/j.ijpharm.2023.123233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. After myocardial infarction, the vascular supply of the heart is damaged or blocked, leading to the formation of scar tissue, followed by several cardiac dysfunctions or even death. In this regard, induction of angiogenesis is considered as a vital process for supplying nutrients and oxygen to the cells in cardiac tissue engineering. The current review aims to summarize different approaches of angiogenesis induction for effective cardiac tissue repair. Accordingly, a comprehensive classification of induction of pro-angiogenic signaling pathways through using engineered biomaterials, drugs, angiogenic factors, as well as combinatorial approaches is introduced as a potential platform for cardiac regeneration application. The angiogenic induction for cardiac repair can enhance patient treatment outcomes and generate economic prospects for the biomedical industry. The development and commercialization of angiogenesis methods often involves collaboration between academic institutions, research organizations, and biomedical companies.
Collapse
Affiliation(s)
- Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg 9220, Denmark
| | - Shirin Nour
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, VIC 3010, Australia; Department of Chemical Engineering, The University of Melbourne, VIC 3010, Australia
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Mohammad H Siadati
- Materials Science and Engineering Faculty, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
9
|
Hoffmann E, Gerwing M, Krähling T, Hansen U, Kronenberg K, Masthoff M, Geyer C, Höltke C, Wachsmuth L, Schinner R, Hoerr V, Heindel W, Karst U, Eisenblätter M, Maus B, Helfen A, Faber C, Wildgruber M. Vascular response patterns to targeted therapies in murine breast cancer models with divergent degrees of malignancy. Breast Cancer Res 2023; 25:56. [PMID: 37221619 DOI: 10.1186/s13058-023-01658-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 05/14/2023] [Indexed: 05/25/2023] Open
Abstract
BACKGROUND Response assessment of targeted cancer therapies is becoming increasingly challenging, as it is not adequately assessable with conventional morphological and volumetric analyses of tumor lesions. The tumor microenvironment is particularly constituted by tumor vasculature which is altered by various targeted therapies. The aim of this study was to noninvasively assess changes in tumor perfusion and vessel permeability after targeted therapy in murine models of breast cancer with divergent degrees of malignancy. METHODS Low malignant 67NR or highly malignant 4T1 tumor-bearing mice were treated with either the multi-kinase inhibitor sorafenib or immune checkpoint inhibitors (ICI, combination of anti-PD1 and anti-CTLA4). Dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) with i.v. injection of albumin-binding gadofosveset was conducted on a 9.4 T small animal MRI. Ex vivo validation of MRI results was achieved by transmission electron microscopy, immunohistochemistry and laser ablation-inductively coupled plasma-mass spectrometry. RESULTS Therapy-induced changes in tumor vasculature differed between low and highly malignant tumors. Sorafenib treatment led to decreased tumor perfusion and endothelial permeability in low malignant 67NR tumors. In contrast, highly malignant 4T1 tumors demonstrated characteristics of a transient window of vascular normalization with an increase in tumor perfusion and permeability early after therapy initiation, followed by decreased perfusion and permeability parameters. In the low malignant 67NR model, ICI treatment also mediated vessel-stabilizing effects with decreased tumor perfusion and permeability, while ICI-treated 4T1 tumors exhibited increasing tumor perfusion with excessive vascular leakage. CONCLUSION DCE-MRI enables noninvasive assessment of early changes in tumor vasculature after targeted therapies, revealing different response patterns between tumors with divergent degrees of malignancy. DCE-derived tumor perfusion and permeability parameters may serve as vascular biomarkers that allow for repetitive examination of response to antiangiogenic treatment or immunotherapy.
Collapse
Grants
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
- 446302350, 194468054, 431460824 Deutsche Forschungsgemeinschaft
Collapse
Affiliation(s)
- Emily Hoffmann
- Clinic of Radiology, University of Münster, Münster, Germany.
| | - Mirjam Gerwing
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Tobias Krähling
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Uwe Hansen
- Institute for Musculoskeletal Medicine, University of Münster, Münster, Germany
| | - Katharina Kronenberg
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Max Masthoff
- Clinic of Radiology, University of Münster, Münster, Germany
| | | | - Carsten Höltke
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Lydia Wachsmuth
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Regina Schinner
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| | - Verena Hoerr
- Clinic of Radiology, University of Münster, Münster, Germany
- Heart Center Bonn, Department of Internal Medicine II, University Hospital Bonn, Bonn, Germany
| | - Walter Heindel
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Uwe Karst
- Institute of Inorganic and Analytical Chemistry, University of Münster, Münster, Germany
| | - Michel Eisenblätter
- Clinic of Radiology, University of Münster, Münster, Germany
- Department of Diagnostic and Interventional Radiology, Medical Faculty OWL, University of Bielefeld, Bielefeld, Germany
| | - Bastian Maus
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Anne Helfen
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Cornelius Faber
- Clinic of Radiology, University of Münster, Münster, Germany
| | - Moritz Wildgruber
- Clinic of Radiology, University of Münster, Münster, Germany
- Department of Radiology, University Hospital, LMU Munich, Munich, Germany
| |
Collapse
|
10
|
Singh P, Shukla P, Narula AK, Deswal D. Polysaccharides and lipoproteins as reactants for the synthesis of pharmaceutically important scaffolds: A review. Int J Biol Macromol 2023; 242:124884. [PMID: 37207747 DOI: 10.1016/j.ijbiomac.2023.124884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/17/2023] [Accepted: 05/12/2023] [Indexed: 05/21/2023]
Abstract
The growing number of diseases in the past decade has once again highlighted the need for extensive research on the development of novel drugs. There has been a major expansion in the number of people suffering from malignant diseases and types of life-threatening microbial infections. The high mortality rates caused by such infections, their associated toxicity, and a growing number of microbes with acquired resistance necessitate the need to further explore and develop the synthesis of pharmaceutically important scaffolds. Chemical entities derived from biological macromolecules like carbohydrates and lipids have been explored and observed to be effective agents in the treatment of microbial infections and diseases. These biological macromolecules offer a variety of chemical properties that have been exploited for the synthesis of pharmaceutically relevant scaffolds. All biological macromolecules are long chains of similar atomic groups which are connected by covalent bonds. By altering the attached groups, the physical and chemical properties can be altered and molded as per the clinical applications and needs, this ring them potential candidates for drug synthesis. The present review establishes the role and significance of biological macromolecules by articulating various reactions and pathways reported in the literature.
Collapse
Affiliation(s)
- Parinita Singh
- Centre of Excellence in Pharmaceutical Sciences (CEPS), Guru Gobind Singh Indraprastha University (GGSIPU), New Delhi, India
| | - Pratibha Shukla
- Centre of Excellence in Pharmaceutical Sciences (CEPS), Guru Gobind Singh Indraprastha University (GGSIPU), New Delhi, India
| | - A K Narula
- Centre of Excellence in Pharmaceutical Sciences (CEPS), Guru Gobind Singh Indraprastha University (GGSIPU), New Delhi, India
| | - Deepa Deswal
- Centre of Excellence in Pharmaceutical Sciences (CEPS), Guru Gobind Singh Indraprastha University (GGSIPU), New Delhi, India.
| |
Collapse
|
11
|
Liu T, Lu Y, Zhan R, Qian W, Luo G. Nanomaterials and nanomaterials-based drug delivery to promote cutaneous wound healing. Adv Drug Deliv Rev 2023; 193:114670. [PMID: 36538990 DOI: 10.1016/j.addr.2022.114670] [Citation(s) in RCA: 56] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 11/24/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Various factors could damage the structure and integrity of skin to cause wounds. Nonhealing or chronic wounds seriously affect the well-being of patients and bring heavy burdens to the society. The past few decades have witnessed application of numerous nanomaterials to promote wound healing. Owing to the unique physicochemical characteristics at nanoscale, nanomaterials-based therapy has been regarded as a potential approach to promote wound healing. In this review, we first overview the wound categories, wound healing process and critical influencing factors. Then applications of nanomaterials with intrinsic therapeutic effect and nanomaterials-based drug delivery systems to promote wound healing are addressed in detail. Finally, current limitations and future perspectives of nanomaterials in wound healing are discussed.
Collapse
Affiliation(s)
- Tengfei Liu
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yifei Lu
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Rixing Zhan
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Wei Qian
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Gaoxing Luo
- Institute of Burn Research, Southwest Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
12
|
Development of L-Lysine-Loaded PLGA Microparticles as a Controlled Release System for Angiogenesis Enhancement. Pharmaceutics 2023; 15:pharmaceutics15020479. [PMID: 36839801 PMCID: PMC9961840 DOI: 10.3390/pharmaceutics15020479] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/04/2023] Open
Abstract
Vascularization is a highly conserved and considerably complex and precise process that is finely driven by endogenous regulatory processes at the tissue and systemic levels. However, it can reveal itself to be slow and inadequate for tissue repair and regeneration consequent to severe lesions/damages. Several biomaterial-based strategies were developed to support and enhance vasculogenesis by supplying pro-angiogenic agents. Several approaches were adopted to develop effective drug delivery systems for the controlled release of a huge variety of compounds. In this work, a microparticulate system was chosen to be loaded with the essential amino acid L-lysine, a molecule that has recently gained interest due to its involvement in pro-angiogenic, pro-regenerative, and anti-inflammatory mechanisms. Poly (lactic-co-glycolic acid), the most widely used FDA-approved biodegradable synthetic polymer for the development of drug delivery systems, was chosen due to its versatility and ability to promote neovascularization and wound healing. This study dealt with the development and the effectiveness evaluation of a PLGA-based microparticulate system for the controlled release of L-lysine. Therefore, in order to maximize L-lysine encapsulation efficiency and tune its release kinetics, the microparticle synthesis protocol was optimized by varying some processing parameters. All developed formulations were characterized from a morphological and physicochemical point of view. The optimized formulation was further characterized via the evaluation of its preliminary biological efficacy in vitro. The cellular and molecular studies revealed that the L-lysine-loaded PLGA microparticles were non-toxic, biocompatible, and supported cell proliferation and angiogenesis well by stimulating the expression of pro-angiogenic genes such as metalloproteinase-9, focal adhesion kinases, and different growth factors. Thus, this work showed the potential of delivering L-lysine encapsulated in PLGA microparticles as a cost-effective promoter system for angiogenesis enhancement and rapid healing.
Collapse
|
13
|
Bhushan NP, Stack T, Scott EA, Shull KR, Mathew B, Bijukumar D. In vitro assessment of varying peptide surface density on the suppression of angiogenesis by micelles displaying αvβ3 blocking peptides. J Biomed Mater Res B Appl Biomater 2023; 111:343-353. [PMID: 36054456 PMCID: PMC9771939 DOI: 10.1002/jbm.b.35154] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/18/2022] [Accepted: 08/17/2022] [Indexed: 12/24/2022]
Abstract
Ligand targeted therapy (LTT) is a precision medicine strategy that can selectively target diseased cells while minimizing off-target effects on healthy cells. Integrin-targeted LTT has been developed recently for angiogenesis-related diseases. However, the clinical success is based on the optimal design of the nanoparticles for inducing receptor clustering within the cell membrane. The current study focused on determining the surface density of Ser-Asp-Val containing anti-integrin heptapeptide on poly (ethylene glycol)-b-poly(propylene sulfide) micelles (MC) required for anti-angiogenic effects on HUVECs. Varying peptide density on PEG-b-PPS/Pep-PA MCs (Pep-PA-Peptide-palmitoleic acid) was used in comparison to a random peptide (SGV) and cRGD (cyclic-Arginine-Glycine-Aspartic acid) construct at 5%-density on MCs. Immunocytochemistry using CD51/CD31 antibody was performed to study the integrin blocking by MCs. In addition, the expression of VWF and PECAM-1, cell migration and tube formation was evaluated in the presence of PEG-b-PPS/Pep-PA MCs. The results show PEG-b-PPS/SDV-PA MCs with 5%-peptide density to achieve significantly higher αvβ3 blocking compared to random peptide as well as cRGD. In addition, αvβ3 blocking via MCs further reduced the expression of vWF and PECAM-1 angiogenesis protein expression in HUVECs. Although a significant level of integrin blocking was observed for 1%-peptide density on MCs, the cell migration and tube formation were not significantly affected. In conclusion, the results of this study demonstrate that the peptide surface density on PEG-b-PPS/Pep-PA MCs has a significant impact in integrin blocking as well as inhibiting angiogenesis during LTT. The outcomes of this study provides insight into the design of ligand targeted nanocarriers for various disease conditions.
Collapse
Affiliation(s)
- Neha Phani Bhushan
- Department of Biomedical SciencesUniversity of Illinois College of Medicine at RockfordRockfordIllinoisUSA
| | - Trevor Stack
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIllinoisUSA
| | - Evan A. Scott
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIllinoisUSA
| | - Kenneth R. Shull
- Department of Biomedical EngineeringNorthwestern UniversityEvanstonIllinoisUSA
| | - Benjamin Mathew
- Department of Ophthalmology and Visual SciencesUniversity of IllinoisChicagoIllinoisUSA
| | - Divya Bijukumar
- Department of Biomedical SciencesUniversity of Illinois College of Medicine at RockfordRockfordIllinoisUSA
| |
Collapse
|
14
|
Ren S, Guo S, Yang L, Wang C. Effect of composite biodegradable biomaterials on wound healing in diabetes. Front Bioeng Biotechnol 2022; 10:1060026. [PMID: 36507270 PMCID: PMC9732485 DOI: 10.3389/fbioe.2022.1060026] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/14/2022] [Indexed: 11/27/2022] Open
Abstract
The repair of diabetic wounds has always been a job that doctors could not tackle quickly in plastic surgery. To solve this problem, it has become an important direction to use biocompatible biodegradable biomaterials as scaffolds or dressing loaded with a variety of active substances or cells, to construct a wound repair system integrating materials, cells, and growth factors. In terms of wound healing, composite biodegradable biomaterials show strong biocompatibility and the ability to promote wound healing. This review describes the multifaceted integration of biomaterials with drugs, stem cells, and active agents. In wounds, stem cells and their secreted exosomes regulate immune responses and inflammation. They promote angiogenesis, accelerate skin cell proliferation and re-epithelialization, and regulate collagen remodeling that inhibits scar hyperplasia. In the process of continuous combination with new materials, a series of materials that can be well matched with active ingredients such as cells or drugs are derived for precise delivery and controlled release of drugs. The ultimate goal of material development is clinical transformation. At present, the types of materials for clinical application are still relatively single, and the bottleneck is that the functions of emerging materials have not yet reached a stable and effective degree. The development of biomaterials that can be further translated into clinical practice will become the focus of research.
Collapse
Affiliation(s)
- Sihang Ren
- NHC Key Laboratory of Reproductive Health and Medical Genetics (Liaoning Research Institute of Family Planning), The Affiliated Reproductive Hospital of China Medical University, Shenyang, China
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
- The First Clinical College of China Medical UniversityChina Medical University, Shenyang, China
- Department of Plastic Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Shuaichen Guo
- The First Clinical College of China Medical UniversityChina Medical University, Shenyang, China
| | - Liqun Yang
- NHC Key Laboratory of Reproductive Health and Medical Genetics (Liaoning Research Institute of Family Planning), The Affiliated Reproductive Hospital of China Medical University, Shenyang, China
| | - Chenchao Wang
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
15
|
Tuli HS, Kumar A, Sak K, Aggarwal D, Gupta DS, Kaur G, Vashishth K, Dhama K, Kaur J, Saini AK, Varol M, Capanoglu E, Haque S. Gut Microbiota-Assisted Synthesis, Cellular Interactions and Synergistic Perspectives of Equol as a Potent Anticancer Isoflavone. Pharmaceuticals (Basel) 2022; 15:1418. [PMID: 36422548 PMCID: PMC9697248 DOI: 10.3390/ph15111418] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 10/30/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
It is well known that, historically, plants have been an important resource of anticancer agents, providing several clinically approved drugs. Numerous preclinical studies have shown a strong anticancer potential of structurally different phytochemicals, including polyphenolic constituents of plants, flavonoids. In this review article, suppressing effects of equol in different carcinogenesis models are unraveled, highlighting the mechanisms involved in these anticancer activities. Among flavonoids, daidzein is a well-known isoflavone occurring in soybeans and soy products. In a certain part of population, this soy isoflavone is decomposed to equol under the action of gut microflora. Somewhat surprisingly, this degradation product has been shown to be more bioactive than its precursor daidzein, revealing a strong and multifaceted anticancer potential. In this way, it is important to bear in mind that the metabolic conversion of plant flavonoids might lead to products that are even more efficient than the parent compounds themselves, definitely deserving further studies.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Ambala 133207, India
| | - Ajay Kumar
- Punjab Biotechnology Incubator (PBTI), Phase VIII, Mohali 160071, India
| | | | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Ambala 133207, India
| | - Dhruv Sanjay Gupta
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s, NMIMS, Mumbai 400056, India
| | - Ginpreet Kaur
- Department of Pharmacology, Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s, NMIMS, Mumbai 400056, India
| | - Kanupriya Vashishth
- Advance Cardiac Centre Department of Cardiology, Post Graduate Institute of Medical Education and Research (PGIMER) Chandigarh 160012, India
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122, India
| | - Jagjit Kaur
- Graduate School of Biomedical Engineering, ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia
| | - Adesh K. Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar (Deemed to be University), Ambala 133207, India
| | - Mehmet Varol
- Department of Molecular Biology and Genetics, Faculty of Science, Kotekli Campus, Mugla Sitki Kocman University, Mugla 48000, Turkey
| | - Esra Capanoglu
- Department of Food Engineering, Faculty of Chemical and Metallurgical Engineering, Istanbul Technical University, Istanbul 34469, Turkey
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia
| |
Collapse
|
16
|
Damanik FR, Rothuizen CT, Lalai R, Khoenkhoen S, van Blitterswijk C, Rotmans JI, Moroni L. Long-Term Controlled Growth Factor Release Using Layer-by-Layer Assembly for the Development of In Vivo Tissue-Engineered Blood Vessels. ACS APPLIED MATERIALS & INTERFACES 2022; 14:28591-28603. [PMID: 35696386 PMCID: PMC9247980 DOI: 10.1021/acsami.2c05988] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The development of a well-designed tissue-engineered blood vessel (TEBV) still remains a challenge. In recent years, approaches in which the host response to implanted biomaterials is used to generate vascular constructs within the patient's body have gained increasing interest. The delivery of growth factors to these in situ-engineered vascular grafts might enhance myofibroblast recruitment and the secretion of essential extracellular matrix proteins, thereby optimizing their functional properties. Layer-by-layer (LbL) coating has emerged as an innovative technology for the controlled delivery of growth factors in tissue engineering applications. In this study, we combined the use of surface-etched polymeric rods with LbL coatings to control the delivery of TGF-β1, PDGF-BB, and IGF-1 and steer the foreign body response toward the formation of a functional vascular graft. Results showed that the regenerated tissue is composed of elastin, glycosaminoglycans, and circumferentially oriented collagen fibers, without calcification or systemic spill of the released growth factors. Functional controlled delivery was observed, whereas myofibroblast-rich tissue capsules were formed with enhanced collagen and elastin syntheses using TGF-β1 and TGF-β1/PDGF-BB releasing rods, when compared to control rods that were solely surface-engineered by chloroform etching. By combining our optimized LbL method and surface-engineered rods in an in vivo bioreactor approach, we could regulate the fate and ECM composition of in situ-engineered vascular grafts to create a successful in vivo vascular tissue-engineered replacement.
Collapse
Affiliation(s)
- Febriyani
F. R. Damanik
- Tissue
Regeneration Department, MIRA Institute for Biomedical Technology
and Technical Medicine, University of Twente, Drienerlolaan 5, Zuidhorst 145, 7522 NB Enschede, The Netherlands
- Faculty
of Science, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Carolien T. Rothuizen
- Department
of Internal Medicine, Leiden University
Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Reshma Lalai
- Department
of Internal Medicine, Leiden University
Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Sandhia Khoenkhoen
- Faculty
of Science, Radboud University, Heyendaalseweg 135, 6525 AJ Nijmegen, The Netherlands
| | - Clemens van Blitterswijk
- Tissue
Regeneration Department, MIRA Institute for Biomedical Technology
and Technical Medicine, University of Twente, Drienerlolaan 5, Zuidhorst 145, 7522 NB Enschede, The Netherlands
- Complex
Tissue Regeneration Department, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| | - Joris I. Rotmans
- Department
of Internal Medicine, Leiden University
Medical Center, Albinusdreef 2, 2333 ZA Leiden, The Netherlands
| | - Lorenzo Moroni
- Tissue
Regeneration Department, MIRA Institute for Biomedical Technology
and Technical Medicine, University of Twente, Drienerlolaan 5, Zuidhorst 145, 7522 NB Enschede, The Netherlands
- Complex
Tissue Regeneration Department, MERLN Institute for Technology Inspired
Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, The Netherlands
| |
Collapse
|
17
|
Martinez B, Peplow PV. MicroRNAs as diagnostic and prognostic biomarkers of age-related macular degeneration: advances and limitations. Neural Regen Res 2021; 16:440-447. [PMID: 32985463 PMCID: PMC7996036 DOI: 10.4103/1673-5374.293131] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/11/2020] [Accepted: 04/02/2020] [Indexed: 01/10/2023] Open
Abstract
A main cause of vision loss in the elderly is age-related macular degeneration (AMD). Among the cellular, biochemical, and molecular changes linked to this disease, inflammation and angiogenesis appear as being crucial in AMD pathogenesis and progression. There are two forms of the disease: dry AMD, accounting for 80-90% of cases, and wet AMD. The disease usually begins as dry AMD associated with retinal pigment epithelium and photoreceptor degeneration, whereas wet AMD is associated with choroidal neovascularization resulting in severe vision impairment. The new vessels are largely malformed, leading to blood and fluid leakage within the disrupted tissue, which provokes inflammation and scar formation and results in retinal damage and detachment. MicroRNAs are dysregulated in AMD and may facilitate the early detection of the disease and monitoring disease progression. Two recent reviews of microRNAs in AMD had indicated weaknesses or limitations in four earlier investigations. Studies in the last three years have shown considerable progress in overcoming some of these concerns and identifying specific microRNAs as biomarkers for AMD. Further large-scale studies are warranted using appropriate statistical methods to take into account gender and age disparity in the study populations and confounding factors such as smoking status.
Collapse
Affiliation(s)
- Bridget Martinez
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA
- Department of Medicine, St. George's University School of Medicine, Grenada
| | - Philip V. Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Zabroski IO, Nugent MA. Lipid Raft Association Stabilizes VEGF Receptor 2 in Endothelial Cells. Int J Mol Sci 2021; 22:ijms22020798. [PMID: 33466887 PMCID: PMC7830256 DOI: 10.3390/ijms22020798] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/04/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The binding of vascular endothelial growth factor A (VEGF) to VEGF receptor-2 (VEGFR-2) stimulates angiogenic signaling. Lipid rafts are cholesterol-dense regions of the plasma membrane that serve as an organizational platform for biomolecules. Although VEGFR2 has been shown to colocalize with lipid rafts to regulate its activation, the effect of lipid rafts on non-activated VEGFR2 has not been explored. Here, we characterized the involvement of lipid rafts in modulating the stability of non-activated VEGFR2 in endothelial cells using raft disrupting agents: methyl-β-cyclodextrin, sphingomyelinase and simvastatin. Disrupting lipid rafts selectively decreased the levels of non-activated VEGFR2 as a result of increased lysosomal degradation. The decreased expression of VEGFR2 translated to reduced VEGF-activation of the extracellular signal-regulated protein kinases (ERK). Overall, our results indicate that lipid rafts stabilize VEGFR2 and its associated signal transduction activities required for angiogenesis. Thus, modulation of lipid rafts may provide a means to regulate the sensitivity of endothelial cells to VEGF stimulation. Indeed, the ability of simvastatin to down regulate VEGFR2 and inhibit VEGF activity suggest a potential mechanism underlying the observation that this drug improves outcomes in the treatment of certain cancers.
Collapse
|
19
|
Martinez B, Peplow PV. MicroRNAs in laser-induced choroidal neovascularization in mice and rats: their expression and potential therapeutic targets. Neural Regen Res 2021; 16:621-627. [PMID: 33063711 PMCID: PMC8067925 DOI: 10.4103/1673-5374.295271] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Choroidal neovascularization characterizes wet age-related macular degeneration. Choroidal neovascularization formation involves a primarily angiogenic process that is combined with both inflammation and proteolysis. A primary cause of choroidal neovascularization pathogenesis is alterations in pro- and anti-angiogenic factors derived from the retinal pigment epithelium, with vascular endothelium growth factor being mainly responsible for both clinical and experimental choroidal neovascularization. MicroRNAs (miRNAs) which are short, non-coding, endogenous RNA molecules have a major role in regulating various pathological processes, including inflammation and angiogenesis. A review of recent studies with the mouse laser-induced choroidal neovascularization model has shown alterations in miRNA expression in choroidal neovascularization tissues and could be potential therapeutic targets for wet age-related macular degeneration. Upregulation of miR-505 (days 1 and 3 post-laser), miR-155 (day 14) occurred in retina; miR-342-5p (days 3 and 7), miR-126-3p (day 14) in choroid; miR-23a, miR-24, miR-27a (day 7) in retina/choroid; miR-505 (days 1 and 3) in retinal pigment epithelium/choroid; downregulation of miR-155 (days 1 and 3), miR-29a, miR-29b, miR-29c (day 5), miR-93 (day 14), miR-126 (day 14) occurred in retinal pigment epithelium/choroid. Therapies using miRNA mimics or inhibitors were found to decrease choroidal neovascularization lesions. Choroidal neovascularization development was reduced by overexpression of miR-155, miR-188-5p, miR-(5,B,7), miR-126-3p, miR-342-5p, miR-93, miR-126, miR-195a-3p, miR-24, miR-21, miR-31, miR-150, and miR-184, or suppression of miR-505, miR-126-3p, miR-155, and miR-23/27. Further studies are warranted to determine miRNA expression in mouse laser-induced choroidal neovascularization models in order to validate and extend the reported findings. Important experimental variables need to be standardized; these include the strain and age of animals, gender, number and position of laser burns to the eye, laser parameters to induce choroidal neovascularization lesions including wavelength, power, spot size, and duration.
Collapse
Affiliation(s)
- Bridget Martinez
- Physical Chemistry and Applied Spectroscopy, Chemistry Division, Los Alamos National Laboratory, Los Alamos, NM, USA; Department of Medicine, St. Georges University School of Medicine, Grenada
| | - Philip V Peplow
- Department of Anatomy, University of Otago, Dunedin, New Zealand
| |
Collapse
|
20
|
Kumar AS, Kamalasanan K. Drug delivery to optimize angiogenesis imbalance in keloid: A review. J Control Release 2020; 329:1066-1076. [PMID: 33091533 DOI: 10.1016/j.jconrel.2020.10.035] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 10/16/2020] [Accepted: 10/17/2020] [Indexed: 12/12/2022]
Abstract
The wound healing process involves three continuous stages. Where, any imbalance can lead to the formation of unwanted keloids, hypertrophic scar, or tumors. Keloids are any unpleasant, non-compliant comorbidity affecting a major section of people around the globe who acquire it either genetically or by pathological means as a result of a skin injury. Angiogenesis is unavoidable in the healing process after an injury or disruption of skin to promote tissue regeneration. Uncontrolled angiogenesis during the healing process can initiate the unwanted response in the wound that facilitate keloid. Angiogenic therapy is adapted to accelerate healing after an injury. Else ways, there exists a risk of keloid formation due to excessive angiogenesis during the wound healing process. There are numerous strategies to treat keloid. Anti-angiogenic factors are provided to patients post-surgery to prevent the keloid formation; however, they come into the picture after the formation of keloid. The available strategies to treat keloids are steroidal injections, surgical excision of the keloid, radiotherapy, pressure therapy, the use of cryosurgery, and many more. The available treatments are not promising in reducing the recurrent rate of keloids as there are chances of high re-occurrences with similar/larger lesions on the removed keloid site. In this review, we are discussing the importance of controlled angiogenesis with the help of controlled drug delivery strategies enabling the wound healing process without the induction of keloid.
Collapse
Affiliation(s)
- Aishwari S Kumar
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, AIMS Ponekkara PO, Kochi, Kerala, 682041, India
| | - Kaladhar Kamalasanan
- Department of Pharmaceutics, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, AIMS Ponekkara PO, Kochi, Kerala, 682041, India.
| |
Collapse
|
21
|
Sarkar B, Siddiqui Z, Kim KK, Nguyen PK, Reyes X, McGill TJ, Kumar VA. Implantable anti-angiogenic scaffolds for treatment of neovascular ocular pathologies. Drug Deliv Transl Res 2020; 10:1191-1202. [PMID: 32232681 PMCID: PMC7483832 DOI: 10.1007/s13346-020-00753-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The retinal physiology can accrue oxidative damage and inflammatory insults due to age and metabolic irregularities. Two notable diseases that involve retinal and choroidal neovascularization are proliferative diabetic retinopathy and wet age-related macular degeneration. Currently, these diseases are mainly treated with anti-VEGF drugs (VEGF = vascular endothelial growth factor), generally on a monthly dosage scheme. We discuss recent developments for the treatment of these diseases, including bioactive tissue-engineered materials, which may reduce frequency of dosage and propose a path forward for improving patient outcomes. Graphical abstract Development of materials for long-term intravitreal delivery for management of posterior segment diseases.
Collapse
Affiliation(s)
- Biplab Sarkar
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Zain Siddiqui
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Ka Kyung Kim
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Peter K Nguyen
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Xavier Reyes
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA
| | - Trevor J McGill
- Casey Eye Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Vivek A Kumar
- Department of Biomedical Engineering, New Jersey Institute of Technology, 138 Warren St. LSEB 316, Newark, NJ, 07102, USA.
- Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
- Department of Restorative Dentistry, Rutgers School of Dental Medicine, Newark, NJ, USA.
| |
Collapse
|
22
|
Xing F, Xiang Z, Rommens PM, Ritz U. 3D Bioprinting for Vascularized Tissue-Engineered Bone Fabrication. MATERIALS (BASEL, SWITZERLAND) 2020; 13:E2278. [PMID: 32429135 PMCID: PMC7287611 DOI: 10.3390/ma13102278] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 03/26/2020] [Accepted: 04/08/2020] [Indexed: 02/05/2023]
Abstract
Vascularization in bone tissues is essential for the distribution of nutrients and oxygen, as well as the removal of waste products. Fabrication of tissue-engineered bone constructs with functional vascular networks has great potential for biomimicking nature bone tissue in vitro and enhancing bone regeneration in vivo. Over the past decades, many approaches have been applied to fabricate biomimetic vascularized tissue-engineered bone constructs. However, traditional tissue-engineered methods based on seeding cells into scaffolds are unable to control the spatial architecture and the encapsulated cell distribution precisely, which posed a significant challenge in constructing complex vascularized bone tissues with precise biomimetic properties. In recent years, as a pioneering technology, three-dimensional (3D) bioprinting technology has been applied to fabricate multiscale, biomimetic, multi-cellular tissues with a highly complex tissue microenvironment through layer-by-layer printing. This review discussed the application of 3D bioprinting technology in the vascularized tissue-engineered bone fabrication, where the current status and unique challenges were critically reviewed. Furthermore, the mechanisms of vascular formation, the process of 3D bioprinting, and the current development of bioink properties were also discussed.
Collapse
Affiliation(s)
- Fei Xing
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany; (F.X.); (P.M.R.)
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China;
- Trauma Medical Center of West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China
| | - Zhou Xiang
- Department of Orthopaedics, West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China;
- Trauma Medical Center of West China Hospital, Sichuan University, No. 37 Guoxue Lane, Chengdu 610041, China
| | - Pol Maria Rommens
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany; (F.X.); (P.M.R.)
| | - Ulrike Ritz
- Department of Orthopaedics and Traumatology, Biomatics Group, University Medical Center of the Johannes Gutenberg University, Mainz 55131, Germany; (F.X.); (P.M.R.)
| |
Collapse
|
23
|
Lee JB, Kim DH, Yoon JK, Park DB, Kim HS, Shin YM, Baek W, Kang ML, Kim HJ, Sung HJ. Microchannel network hydrogel induced ischemic blood perfusion connection. Nat Commun 2020; 11:615. [PMID: 32001693 PMCID: PMC6992688 DOI: 10.1038/s41467-020-14480-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 01/12/2020] [Indexed: 12/13/2022] Open
Abstract
Angiogenesis induction into damaged sites has long been an unresolved issue. Local treatment with pro-angiogenic molecules has been the most common approach. However, this approach has critical side effects including inflammatory coupling, tumorous vascular activation, and off-target circulation. Here, the concept that a structure can guide desirable biological function is applied to physically engineer three-dimensional channel networks in implant sites, without any therapeutic treatment. Microchannel networks are generated in a gelatin hydrogel to overcome the diffusion limit of nutrients and oxygen three-dimensionally. Hydrogel implantation in mouse and porcine models of hindlimb ischemia rescues severely damaged tissues by the ingrowth of neighboring host vessels with microchannel perfusion. This effect is guided by microchannel size-specific regenerative macrophage polarization with the consequent functional recovery of endothelial cells. Multiple-site implantation reveals hypoxia and neighboring vessels as major causative factors of the beneficial function. This technique may contribute to the development of therapeutics for hypoxia/inflammatory-related diseases.
Collapse
Affiliation(s)
- Jung Bok Lee
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dae-Hyun Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Dan Bi Park
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hye-Seon Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Young Min Shin
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Wooyeol Baek
- Department of Plastic & Reconstructive Surgery, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Mi-Lan Kang
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- TMD LAB Co. Ltd., 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
| | - Hyun Jung Kim
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hak-Joon Sung
- Department of Medical Engineering, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Republic of Korea.
| |
Collapse
|
24
|
Associations of microRNAs, Angiogenesis-Regulating Factors and CFH Y402H Polymorphism-An Attempt to Search for Systemic Biomarkers in Age-Related Macular Degeneration. Int J Mol Sci 2019; 20:ijms20225750. [PMID: 31731799 PMCID: PMC6887747 DOI: 10.3390/ijms20225750] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 11/08/2019] [Accepted: 11/08/2019] [Indexed: 01/13/2023] Open
Abstract
Age-related macular degeneration (AMD) remains the leading cause of blindness in elderly people, but the pathophysiology of this disease is still largely unknown. We investigated the systemic expression of angiogenesis-regulating growth factors and selected miRNAs known to regulate angiogenesis in AMD patients. We also focused on possible correlations of their expression with the presence of CFH Y402H or ARMS A69S risk variants. A total of 354 AMD patients and 121 controls were enrolled in this study. The levels of angiogenesis-regulating factors were analyzed in plasma samples using Luminex technology. The expression of selected miRNAs was analyzed in peripheral blood plasma using real-time qPCR. The genetic analysis was performed with an Illumina NextSeq500 system. AMD was an independent factor associated with lower levels of angiogenin (β = −0.29, p < 0.001), endostatin (β = −0.18, p < 0.001), FGF-basic (β = −0.18, p < 0.001), PlGF (β = −0.24, p < 0.001), miRNA-21-3p (β = −0.13, p = 0.01) and miRNA-155-5p (β = −0.16, p = 0.002); and with higher levels of FGF-acidic (β = 0.11, p = 0.03), miRNA-23a-3p (β = 0.17, p < 0.001), miRNA-126-5p (β = 0.13, p = 0.009), miRNA-16-5p (β = 0.40, p < 0.001), miRNA-17-3p (β = 0.13, p = 0.01), miRNA-17-5p (β = 0.17, p < 0.001), miRNA-223-3p (β = 0.15, p = 0.004), and miRNA-93 (β = 0.11, p = 0.04). The expression of analyzed miRNA molecules significantly correlated with the levels of tested angiogenesis-regulating factors and clinical parameters in AMD patients, whereas such correlations were not observed in controls. We also found an association between the CFH Y402H polymorphism and miRNA profiles, whereby TT homozygotes showed evidently higher expression of miRNA-16-5p than CC homozygotes or TC heterozygotes (p = 0.0007). Our results suggest that the balance between systemic pro- and anti-angiogenic factors and miRNAs is vital in multifactorial AMD pathogenesis.
Collapse
|
25
|
Sun B, Zhang M, Shen J, He Z, Fatehi P, Ni Y. Applications of Cellulose-based Materials in Sustained Drug Delivery Systems. Curr Med Chem 2019; 26:2485-2501. [DOI: 10.2174/0929867324666170705143308] [Citation(s) in RCA: 71] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 03/08/2017] [Accepted: 03/25/2017] [Indexed: 11/22/2022]
Abstract
Bio-compatible, bio-degradable, and bio-available excipients are of critical interest
for drug delivery systems. Cellulose and its derivative-based excipients have been
well studied due to their green/natural and unique encapsulation/binding properties. They
are often used in controlled/sustained drug delivery systems. In these applications, cellulose
and its derivatives function generally can modify the solubility/gelling behavior of
drugs, resulting in different mechanisms for controlling the release profiles of drugs. In
this paper, the current knowledge in the structure and chemistry of conventional cellulose
derivatives, and their applications in drug delivery systems are briefly reviewed. The development
of innovative cellulose-based materials, including micro-cellulose (MC) and
nano-cellulose (NC) in the applications of sustained drug delivery, is also discussed.
Collapse
Affiliation(s)
- Bo Sun
- Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology), Ministry of Education, Tianjin 300457, China
| | - Min Zhang
- Key Laboratory of Food Nutrition and Safety (Tianjin University of Science and Technology), Ministry of Education, Tianjin 300457, China
| | - Jing Shen
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick, E3B 5A3, Canada
| | - Zhibin He
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick, E3B 5A3, Canada
| | - Pedram Fatehi
- Department of Chemical Engineering, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario P7B 5E1, Canada
| | - Yonghao Ni
- Department of Chemical Engineering, University of New Brunswick, Fredericton, New Brunswick, E3B 5A3, Canada
| |
Collapse
|
26
|
Gaspar D, Peixoto R, De Pieri A, Striegl B, Zeugolis DI, Raghunath M. Local pharmacological induction of angiogenesis: Drugs for cells and cells as drugs. Adv Drug Deliv Rev 2019; 146:126-154. [PMID: 31226398 DOI: 10.1016/j.addr.2019.06.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 05/12/2019] [Accepted: 06/16/2019] [Indexed: 12/12/2022]
Abstract
The past decades have seen significant advances in pro-angiogenic strategies based on delivery of molecules and cells for conditions such as coronary artery disease, critical limb ischemia and stroke. Currently, three major strategies are evolving. Firstly, various pharmacological agents (growth factors, interleukins, small molecules, DNA/RNA) are locally applied at the ischemic region. Secondly, preparations of living cells with considerable bandwidth of tissue origin, differentiation state and preconditioning are delivered locally, rarely systemically. Thirdly, based on the notion, that cellular effects can be attributed mostly to factors secreted in situ, the cellular secretome (conditioned media, exosomes) has come into the spotlight. We review these three strategies to achieve (neo)angiogenesis in ischemic tissue with focus on the angiogenic mechanisms they tackle, such as transcription cascades, specific signalling steps and cellular gases. We also include cancer-therapy relevant lymphangiogenesis, and shall seek to explain why there are often conflicting data between in vitro and in vivo. The lion's share of data encompassing all three approaches comes from experimental animal work and we shall highlight common technical obstacles in the delivery of therapeutic molecules, cells, and secretome. This plethora of preclinical data contrasts with a dearth of clinical studies. A lack of adequate delivery vehicles and standardised assessment of clinical outcomes might play a role here, as well as regulatory, IP, and manufacturing constraints of candidate compounds; in addition, completed clinical trials have yet to reveal a successful and efficacious strategy. As the biology of angiogenesis is understood well enough for clinical purposes, it will be a matter of time to achieve success for well-stratified patients, and most probably with a combination of compounds.
Collapse
Affiliation(s)
- Diana Gaspar
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Rita Peixoto
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Andrea De Pieri
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Proxy Biomedical Ltd., Coilleach, Spiddal, Galway, Ireland
| | - Britta Striegl
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland
| | - Dimitrios I Zeugolis
- Regenerative, Modular & Developmental Engineering Laboratory (REMODEL), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland; Science Foundation Ireland (SFI) Centre for Research in Medical Devices (CÚRAM), Biomedical Sciences Building, National University of Ireland Galway (NUI Galway), Galway, Ireland
| | - Michael Raghunath
- Competence Centre Tissue Engineering for Drug Development (TEDD), Centre for Cell Biology & Tissue Engineering, Institute for Chemistry and Biotechnology, Zurich University of Applied Sciences, Zurich, Switzerland.
| |
Collapse
|
27
|
Saberianpour S, Heidarzadeh M, Geranmayeh MH, Hosseinkhani H, Rahbarghazi R, Nouri M. Tissue engineering strategies for the induction of angiogenesis using biomaterials. J Biol Eng 2018; 12:36. [PMID: 30603044 PMCID: PMC6307144 DOI: 10.1186/s13036-018-0133-4] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023] Open
Abstract
Angiogenesis is touted as a fundamental procedure in the regeneration and restoration of different tissues. The induction of de novo blood vessels seems to be vital to yield a successful cell transplantation rate loaded on various scaffolds. Scaffolds are natural or artificial substances that are considered as one of the means for delivering, aligning, maintaining cell connection in a favor of angiogenesis. In addition to the potential role of distinct scaffold type on vascularization, the application of some strategies such as genetic manipulation, and conjugation of pro-angiogenic factors could intensify angiogenesis potential. In the current review, we focused on the status of numerous scaffolds applicable in the field of vascular biology. Also, different strategies and priming approaches useful for the induction of pro-angiogenic signaling pathways were highlighted.
Collapse
Affiliation(s)
- Shirin Saberianpour
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 2Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Morteza Heidarzadeh
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
| | - Mohammad Hossein Geranmayeh
- 3Neuroscience Research Center, Imam Reza Medical Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Reza Rahbarghazi
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 5Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Nouri
- 2Department of Molecular Medicine, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
- 1Stem Cell Research Center, Tabriz University of Medical Sciences, Imam Reza St., Golgasht St, Tabriz, 5166614756 Iran
- 5Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
28
|
De Rosa L, Di Stasi R, D'Andrea LD. Pro-angiogenic peptides in biomedicine. Arch Biochem Biophys 2018; 660:72-86. [DOI: 10.1016/j.abb.2018.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Revised: 10/11/2018] [Accepted: 10/13/2018] [Indexed: 12/12/2022]
|
29
|
Nguyen EH, Murphy WL. Customizable biomaterials as tools for advanced anti-angiogenic drug discovery. Biomaterials 2018; 181:53-66. [PMID: 30077137 DOI: 10.1016/j.biomaterials.2018.07.041] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 07/17/2018] [Accepted: 07/25/2018] [Indexed: 12/12/2022]
Abstract
The inhibition of angiogenesis is a critical element of cancer therapy, as cancer vasculature contributes to tumor expansion. While numerous drugs have proven to be effective at disrupting cancer vasculature, patient survival has not significantly improved as a result of anti-angiogenic drug treatment. Emerging evidence suggests that this is due to a combination of unintended side effects resulting from the application of anti-angiogenic compounds, including angiogenic rebound after treatment and the activation of metastasis in the tumor. There is currently a need to better understand the far-reaching effects of anti-angiogenic drug treatments in the context of cancer. Numerous innovations and discoveries in biomaterials design and tissue engineering techniques are providing investigators with tools to develop physiologically relevant vascular models and gain insights into the holistic impact of drug treatments on tumors. This review examines recent advances in the design of pro-angiogenic biomaterials, specifically in controlling integrin-mediated cell adhesion, growth factor signaling, mechanical properties and oxygen tension, as well as the implementation of pro-angiogenic materials into sophisticated co-culture models of cancer vasculature.
Collapse
Affiliation(s)
- Eric H Nguyen
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin, Madison, WI, USA; Human Models for Analysis of Pathways (Human MAPs) Center, University of Wisconsin, Madison, WI, USA; Department of Orthopedics and Rehabilitation, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA.
| |
Collapse
|
30
|
Saghazadeh S, Rinoldi C, Schot M, Kashaf SS, Sharifi F, Jalilian E, Nuutila K, Giatsidis G, Mostafalu P, Derakhshandeh H, Yue K, Swieszkowski W, Memic A, Tamayol A, Khademhosseini A. Drug delivery systems and materials for wound healing applications. Adv Drug Deliv Rev 2018; 127:138-166. [PMID: 29626550 PMCID: PMC6003879 DOI: 10.1016/j.addr.2018.04.008] [Citation(s) in RCA: 453] [Impact Index Per Article: 64.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Revised: 04/01/2018] [Accepted: 04/03/2018] [Indexed: 01/22/2023]
Abstract
Chronic, non-healing wounds place a significant burden on patients and healthcare systems, resulting in impaired mobility, limb amputation, or even death. Chronic wounds result from a disruption in the highly orchestrated cascade of events involved in wound closure. Significant advances in our understanding of the pathophysiology of chronic wounds have resulted in the development of drugs designed to target different aspects of the impaired processes. However, the hostility of the wound environment rich in degradative enzymes and its elevated pH, combined with differences in the time scales of different physiological processes involved in tissue regeneration require the use of effective drug delivery systems. In this review, we will first discuss the pathophysiology of chronic wounds and then the materials used for engineering drug delivery systems. Different passive and active drug delivery systems used in wound care will be reviewed. In addition, the architecture of the delivery platform and its ability to modulate drug delivery are discussed. Emerging technologies and the opportunities for engineering more effective wound care devices are also highlighted.
Collapse
Affiliation(s)
- Saghi Saghazadeh
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
| | - Chiara Rinoldi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
- Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology. Warsaw 02-507, Poland
| | - Maik Schot
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
- MIRA Institute of Biomedical Technology and Technical Medicine, Department of Developmental BioEngineering, University of Twente, Enschede, The Netherlands
| | - Sara Saheb Kashaf
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
- The University of Chicago Medical Scientist Training Program, Pritzker School of Medicine, University of Chicago, Chicago, Illinois, USA
| | - Fatemeh Sharifi
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
- School of Mechanical Engineering, Sharif University of Technology, Tehran, Iran
| | - Elmira Jalilian
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
| | - Kristo Nuutila
- Division of Plastic Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Giorgio Giatsidis
- Division of Plastic Surgery, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Pooria Mostafalu
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
| | - Hossein Derakhshandeh
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE, 68508, USA
| | - Kan Yue
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
| | - Wojciech Swieszkowski
- Materials Design Division, Faculty of Materials Science and Engineering, Warsaw University of Technology. Warsaw 02-507, Poland
| | - Adnan Memic
- Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Ali Tamayol
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
- Department of Mechanical and Materials Engineering, University of Nebraska, Lincoln, NE, 68508, USA
| | - Ali Khademhosseini
- Biomaterials Innovation Research Center, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School. Boston, MA 02139, USA
- Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology. Cambridge, MA 02139, USA
- Center of Nanotechnology, Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia
- Department of Chemical and Biomolecular Engineering, Department of Bioengineering, Department of Radiology, California NanoSystems Institute (CNSI), University of California, Los Angeles, CA, 90095, USA
| |
Collapse
|
31
|
Huang G, Li F, Zhao X, Ma Y, Li Y, Lin M, Jin G, Lu TJ, Genin GM, Xu F. Functional and Biomimetic Materials for Engineering of the Three-Dimensional Cell Microenvironment. Chem Rev 2017; 117:12764-12850. [PMID: 28991456 PMCID: PMC6494624 DOI: 10.1021/acs.chemrev.7b00094] [Citation(s) in RCA: 514] [Impact Index Per Article: 64.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The cell microenvironment has emerged as a key determinant of cell behavior and function in development, physiology, and pathophysiology. The extracellular matrix (ECM) within the cell microenvironment serves not only as a structural foundation for cells but also as a source of three-dimensional (3D) biochemical and biophysical cues that trigger and regulate cell behaviors. Increasing evidence suggests that the 3D character of the microenvironment is required for development of many critical cell responses observed in vivo, fueling a surge in the development of functional and biomimetic materials for engineering the 3D cell microenvironment. Progress in the design of such materials has improved control of cell behaviors in 3D and advanced the fields of tissue regeneration, in vitro tissue models, large-scale cell differentiation, immunotherapy, and gene therapy. However, the field is still in its infancy, and discoveries about the nature of cell-microenvironment interactions continue to overturn much early progress in the field. Key challenges continue to be dissecting the roles of chemistry, structure, mechanics, and electrophysiology in the cell microenvironment, and understanding and harnessing the roles of periodicity and drift in these factors. This review encapsulates where recent advances appear to leave the ever-shifting state of the art, and it highlights areas in which substantial potential and uncertainty remain.
Collapse
Affiliation(s)
- Guoyou Huang
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Chemistry, School of Science,
Xi’an Jiaotong University, Xi’an 710049, People’s Republic
of China
| | - Xin Zhao
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Interdisciplinary Division of Biomedical
Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong,
People’s Republic of China
| | - Yufei Ma
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Yuhui Li
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Min Lin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Guorui Jin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| | - Tian Jian Lu
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- MOE Key Laboratory for Multifunctional Materials
and Structures, Xi’an Jiaotong University, Xi’an 710049,
People’s Republic of China
| | - Guy M. Genin
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
- Department of Mechanical Engineering &
Materials Science, Washington University in St. Louis, St. Louis 63130, MO,
USA
- NSF Science and Technology Center for
Engineering MechanoBiology, Washington University in St. Louis, St. Louis 63130,
MO, USA
| | - Feng Xu
- MOE Key Laboratory of Biomedical Information
Engineering, School of Life Science and Technology, Xi’an Jiaotong
University, Xi’an 710049, People’s Republic of China
- Bioinspired Engineering and Biomechanics Center
(BEBC), Xi’an Jiaotong University, Xi’an 710049, People’s
Republic of China
| |
Collapse
|
32
|
Gupta MK, Lee Y, Boire TC, Lee JB, Kim WS, Sung HJ. Recent strategies to design vascular theranostic nanoparticles. Nanotheranostics 2017; 1:166-177. [PMID: 29071185 PMCID: PMC5646719 DOI: 10.7150/ntno.18531] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 03/11/2017] [Indexed: 01/08/2023] Open
Abstract
Vascular disease is a leading cause of death and disability worldwide. Current surgical intervention and treatment options for vascular diseases have exhibited limited long-term success, emphasizing the need to develop advanced treatment paradigms for early detection and more effective treatment of dysfunctional cells in a specific blood vessel lesion. Advances in targeted nanoparticles mediating cargo delivery enables more robust prevention, screening, diagnosis, and treatment of vascular disorders. In particular, nanotheranostics integrates diagnostic imaging and therapeutic function into a single agent, and is an emerging platform towards more effective and localized vascular treatment. This review article highlights recent advances and current challenges associated with the utilization of targeted nanoparticles for real-time diagnosis and treatment of vascular diseases. Given recent developments, nanotheranostics offers great potential to serve as an effective platform for targeted, localized, and personalized vascular treatment.
Collapse
Affiliation(s)
- Mukesh K. Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, US
| | - Yunki Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, US
| | - Timothy C. Boire
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, US
| | - Jung-Bok Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, US
| | - Won Shik Kim
- Department of Otorhinolaryngology, Yonsei University, College of Medicine, South Korea
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, US
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, US
- Severance Biomedical Science Institute, College of Medicine, Yonsei University, Seoul, South Korea
| |
Collapse
|
33
|
Hayashi K, Iwai H, Kamei T, Iwamoto K, Shimanouchi T, Fujita S, Nakamura H, Umakoshi H. Tailor-made drug carrier: Comparison of formation-dependent physicochemical properties within self-assembled aggregates for an optimal drug carrier. Colloids Surf B Biointerfaces 2017; 152:269-276. [DOI: 10.1016/j.colsurfb.2017.01.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/30/2016] [Accepted: 01/07/2017] [Indexed: 10/20/2022]
|
34
|
Eshun D, Saraf R, Bae S, Jeganathan J, Mahmood F, Dilmen S, Ke Q, Lee D, Kang PM, Matyal R. Neuropeptide Y 3-36 incorporated into PVAX nanoparticle improves functional blood flow in a murine model of hind limb ischemia. J Appl Physiol (1985) 2017; 122:1388-1397. [PMID: 28302707 DOI: 10.1152/japplphysiol.00467.2016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 02/21/2017] [Accepted: 03/12/2017] [Indexed: 02/07/2023] Open
Abstract
We generated a novel nanoparticle called PVAX, which has intrinsic antiapoptotic and anti-inflammatory properties. This nanoparticle was loaded with neuropeptide Y3-36 (NPY3-36), an angiogenic neurohormone that plays a central role in angiogenesis. Subsequently, we investigated whether PVAX-NPY3-36 could act as a therapeutic agent and induce angiogenesis and vascular remodeling in a murine model of hind limb ischemia. Adult C57BL/J6 mice (n = 40) were assigned to treatment groups: control, ischemia PBS, ischemia PVAX, ischemia NPY3-36, and Ischemia PVAX-NPY3-36 Ischemia was induced by ligation of the femoral artery in all groups except control and given relevant treatments (PBS, PVAX, NPY3-36, and PVAX-NPY3-36). Blood flow was quantified using laser Doppler imaging. On days 3 and 14 posttreatment, mice were euthanized to harvest gastrocnemius muscle for immunohistochemistry and immunoblotting. Blood flow was significantly improved in the PVAX-NPY3-36 group after 14 days. Western blot showed an increase in angiogenic factors VEGF-R2 and PDGF-β (P = 0.0035 and P = 0.031, respectively) and antiapoptotic marker Bcl-2 in the PVAX-NPY3-36 group compared with ischemia PBS group (P = 0.023). Proapoptotic marker Smad5 was significantly decreased in the PVAX-NPY3-36 group as compared with the ischemia PBS group (P = 0.028). Furthermore, Y2 receptors were visualized in endothelial cells of newly formed arteries in the PVAX-NPY3-36 group. In conclusion, we were able to show that PVAX-NPY3-36 can induce angiogenesis and arteriogenesis as well as improve functional blood flow in a murine model of hind limb ischemia.NEW & NOTEWORTHY Our research project proposes a novel method for drug delivery. Our patented PVAX nanoparticle can detect areas of ischemia and oxidative stress. Although there have been studies about delivering angiogenic molecules to areas of ischemic injury, there are drawbacks of nonspecific delivery as well as short half-lives. Our study is unique because it can specifically deliver NPY3-36 to ischemic tissue and appears to extend the amount of time therapy is available, despite NPY3-36's short half-life.
Collapse
Affiliation(s)
- Derek Eshun
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Rabya Saraf
- Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Soochan Bae
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Jelliffe Jeganathan
- Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; and
| | - Feroze Mahmood
- Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; and
| | - Serkan Dilmen
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Dongwon Lee
- Department of Polymer⋅Nano Science and Technology, Chonbuk National University, Jeonju, South Korea
| | - Peter M Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, Massachusetts
| | - Robina Matyal
- Department of Anesthesia, Critical Care & Pain Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts; and
| |
Collapse
|
35
|
Wang J, Qin B, Chen X, Wagner WR, Villanueva FS. Ultrasound Molecular Imaging of Angiogenesis Using Vascular Endothelial Growth Factor-Conjugated Microbubbles. Mol Pharm 2017; 14:781-790. [PMID: 28165246 DOI: 10.1021/acs.molpharmaceut.6b01033] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Imaging of angiogenesis receptors could provide a sensitive and clinically useful method for detecting neovascularization such as occurs in malignant tumors, and responses to antiangiogenic therapies for such tumors. We tested the hypothesis that microbubbles (MB) tagged with human VEGF121 (MBVEGF) bind to the kinase insert domain receptor (KDR) in vitro and angiogenic endothelium in vivo, and that this specific binding can be imaged on a clinical ultrasound system. In this work, targeted adhesion of MBVEGF was evaluated in vitro using a parallel plate flow system containing adsorbed recombinant human KDR. There was more adhesion of MBVEGF to KDR-coated plates when the amount of VEGF121 on each MB or KDR density on the plate was increased. MBVEGF adhesion to KDR-coated plates decreased with increasing wall shear rate. On intravital microscopic imaging of bFGF-stimulated rat cremaster muscle, there was greater microvascular adhesion of MBVEGF compared to that of isotype IgG-conjugated control MB (MBCTL). To determine if MBVEGF could be used to ultrasonically image angiogenesis, ultrasound imaging was performed in mice bearing squamous cell carcinoma after intravenous injection of MBVEGF. Ultrasound videointensity enhancement in tumor was significantly higher for MBVEGF (17.3 ± 9.7 dB) compared to MBCTL (3.8 ± 4.4 dB, n = 6, p < 0.05). This work demonstrates the feasibility of targeted ultrasound imaging of an angiogenic marker using MBVEGF. This approach offers a noninvasive bedside method for detecting tumor angiogenesis and could be extended to other applications such as molecular monitoring of therapeutic angiogenesis or antiangiogenic therapies in cardiovascular disease or cancer.
Collapse
Affiliation(s)
- Jianjun Wang
- Center of Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center and the University of Pittsburgh , Pittsburgh, Pennsylvania 15213, United States
| | - Bin Qin
- Center of Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center and the University of Pittsburgh , Pittsburgh, Pennsylvania 15213, United States
| | - Xucai Chen
- Center of Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center and the University of Pittsburgh , Pittsburgh, Pennsylvania 15213, United States
| | - William R Wagner
- McGowan Center for Regenerative Medicine, University of Pittsburgh , Pittsburgh, Pennsylvania 15260, United States
| | - Flordeliza S Villanueva
- Center of Ultrasound Molecular Imaging and Therapeutics, University of Pittsburgh Medical Center and the University of Pittsburgh , Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
36
|
Lai PX, Chen CW, Wei SC, Lin TY, Jian HJ, Lai IPJ, Mao JY, Hsu PH, Lin HJ, Tzou WS, Chen SY, Harroun SG, Lai JY, Huang CC. Ultrastrong trapping of VEGF by graphene oxide: Anti-angiogenesis application. Biomaterials 2016; 109:12-22. [DOI: 10.1016/j.biomaterials.2016.09.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 08/31/2016] [Accepted: 09/07/2016] [Indexed: 01/13/2023]
|
37
|
Wang Z, Samanipour R, Kim K. Organ-on-a-Chip Platforms for Drug Screening and Tissue Engineering. BIOSYSTEMS & BIOROBOTICS 2016. [DOI: 10.1007/978-3-319-21813-7_10] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
38
|
Li H, Wan H, Xia T, Chen M, Zhang Y, Luo X, Li X. Therapeutic angiogenesis in ischemic muscles after local injection of fragmented fibers with loaded traditional Chinese medicine. NANOSCALE 2015; 7:13075-13087. [PMID: 26176198 DOI: 10.1039/c5nr02005k] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Therapeutic angiogenesis remains the most effective method to re-establish a proper blood flow in ischemic tissues. There is a great clinical need to identify an injectable format to achieve a well accumulation following local administration and a sustained delivery of biological factors at the ischemic sites. In the current study, fragmented nanofibers with loaded traditional Chinese medicines, astragaloside IV (AT), the main active ingredient of astragalus, and ferulic acid (FA), the main ingredient of angelica, were proposed to promote the microvessel formation after intramuscular injection into ischemic hindlimbs. Fragmented fibers with average lengths of 5 (FF-5), 20 (FF-20) and 80 μm (FF-80) were constructed by the cryocutting of aligned electrospun fibers. Their dispersion in sodium alginate solution (0.2%) indicated good injectability. After injection into the quadriceps muscles of the hindlimbs, FF-20 and FF-80 fiber fragments showed higher tissue retentions than FF-5, and around 90% of the injected doses were determined after 7 days. On a hindlimb ischemia model established by ligating the femoral arteries, intramuscular injection of the mixtures of FA-loaded and AT-loaded FF-20 fiber fragments substantially reduced the muscle degeneration with minimal fibrosis formation, significantly enhanced the neovessel formation and hindlimb perfusion in the ischemic tissues, and efficiently promoted the limb salvage with few limb losses. Along with the easy manipulation and lower invasiveness for in vivo administration, fragmented fibers should become potential drug carriers for disease treatment, wound recovery and tissue repair after local injection.
Collapse
Affiliation(s)
- Huiyan Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education of China, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
39
|
Norton KA, Popel AS. An agent-based model of cancer stem cell initiated avascular tumour growth and metastasis: the effect of seeding frequency and location. J R Soc Interface 2015; 11:20140640. [PMID: 25185580 DOI: 10.1098/rsif.2014.0640] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
It is very important to understand the onset and growth pattern of breast primary tumours as well as their metastatic dissemination. In most cases, it is the metastatic disease that ultimately kills the patient. There is increasing evidence that cancer stem cells are closely linked to the progression of the metastatic tumour. Here, we investigate stem cell seeding to an avascular tumour site using an agent-based stochastic model of breast cancer metastatic seeding. The model includes several important cellular features such as stem cell symmetric and asymmetric division, migration, cellular quiescence, senescence, apoptosis and cell division cycles. It also includes external features such as stem cell seeding frequency and location. Using this model, we find that cell seeding rate and location are important features for tumour growth. We also define conditions in which the tumour growth exhibits decremented and exponential growth patterns. Overall, we find that seeding, senescence and division limit affect not only the number of stem cells, but also their spatial and temporal distribution.
Collapse
Affiliation(s)
- Kerri-Ann Norton
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA Department of Oncology and Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD 21205, USA
| |
Collapse
|
40
|
Lorden ER, Levinson HM, Leong KW. Integration of drug, protein, and gene delivery systems with regenerative medicine. Drug Deliv Transl Res 2015; 5:168-86. [PMID: 25787742 PMCID: PMC4382089 DOI: 10.1007/s13346-013-0165-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Regenerative medicine has the potential to drastically change the field of health care from reactive to preventative and restorative. Exciting advances in stem cell biology and cellular reprogramming have fueled the progress of this field. Biochemical cues in the form of small molecule drugs, growth factors, zinc finger protein transcription factors and nucleases, transcription activator-like effector nucleases, monoclonal antibodies, plasmid DNA, aptamers, or RNA interference agents can play an important role to influence stem cell differentiation and the outcome of tissue regeneration. Many of these biochemical factors are fragile and must act intracellularly at the molecular level. They require an effective delivery system, which can take the form of a scaffold (e.g., hydrogels and electrospun fibers), carrier (viral and nonviral), nano- and microparticle, or genetically modified cell. In this review, we will discuss the history and current technologies of drug, protein, and gene delivery in the context of regenerative medicine. Next, we will present case examples of how delivery technologies are being applied to promote angiogenesis in nonhealing wounds or prevent angiogenesis in age related macular degeneration. Finally, we will conclude with a brief discussion of the regulatory pathway from bench to bedside for the clinical translation of these novel therapeutics.
Collapse
Affiliation(s)
| | - Howard M. Levinson
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Duke University Medical Center, Durham, NC, USA
| | - Kam W. Leong
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| |
Collapse
|
41
|
Lee E, Lee SJ, Koskimaki JE, Han Z, Pandey NB, Popel AS. Inhibition of breast cancer growth and metastasis by a biomimetic peptide. Sci Rep 2014; 4:7139. [PMID: 25409905 PMCID: PMC4238022 DOI: 10.1038/srep07139] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 11/05/2014] [Indexed: 12/21/2022] Open
Abstract
Metastasis is the main cause of mortality in cancer patients. Though there are many anti-cancer drugs targeting primary tumor growth, anti-metastatic agents are rarely developed. Angiogenesis and lymphangiogenesis are crucial for cancer progression, particularly, lymphangiogenesis is pivotal for metastasis in breast cancer. Here we report that a novel collagen IV derived biomimetic peptide inhibits breast cancer growth and metastasis by blocking angiogenesis and lymphangiogenesis. The peptide inhibits blood and lymphatic endothelial cell viability, migration, adhesion, and tube formation by targeting IGF1R and Met signals. The peptide blocks MDA-MB-231 tumor growth by inhibiting tumor angiogenesis in vivo. Moreover, the peptide inhibits lymphangiogenesis in primary tumors. MDA-MB-231 tumor conditioned media (TCM) was employed to accelerate spontaneous metastasis in tumor xenografts, and the anti-metastatic activity of the peptide was tested in this model. The peptide prevents metastasis to the lungs and lymph nodes by inhibiting TCM-induced lymphangiogenesis and angiogenesis in the pre-metastatic organs. In summary, a novel biomimetic peptide inhibits breast cancer growth and metastasis by blocking angiogenesis and lymphangiogenesis in the pre-metastatic organs as well as primary tumors.
Collapse
Affiliation(s)
- Esak Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Seung Jae Lee
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Jacob E Koskimaki
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Zheyi Han
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Niranjan B Pandey
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Aleksander S Popel
- 1] Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States [2] Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| |
Collapse
|
42
|
Norton KA, Han Z, Popel AS, Pandey NB. Antiangiogenic cancer drug sunitinib exhibits unexpected proangiogenic effects on endothelial cells. Onco Targets Ther 2014; 7:1571-82. [PMID: 25228815 PMCID: PMC4164292 DOI: 10.2147/ott.s65055] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Angiogenesis, the formation of new blood vessels, is an essential step for cancer progression, but antiangiogenic therapies have shown limited success. Therefore, a better understanding of the effects of antiangiogenic treatments on endothelial cells is necessary. In this study, we evaluate the changes in cell surface vascular endothelial growth factor receptor (VEGFR) expression on endothelial cells in culture treated with the antiangiogenic tyrosine kinase inhibitor drug sunitinib, using quantitative flow cytometry. We find that proangiogenic VEGFR2 cell surface receptor numbers are increased with sunitinib treatment. This proangiogenic effect might account for the limited effects of sunitinib as a cancer therapy. We also find that this increase is inhibited by brefeldin A, an inhibitor of protein transport from the endoplasmic reticulum to the Golgi apparatus. The complex dynamics of cell surface VEGFRs may be important for successful treatment of cancer with antiangiogenic therapeutics.
Collapse
Affiliation(s)
- Kerri-Ann Norton
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Zheyi Han
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Aleksander S Popel
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA ; Department of Oncology and the Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Niranjan B Pandey
- Department of Biomedical Engineering, School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
43
|
Bhise NS, Ribas J, Manoharan V, Zhang YS, Polini A, Massa S, Dokmeci MR, Khademhosseini A. Organ-on-a-chip platforms for studying drug delivery systems. J Control Release 2014; 190:82-93. [PMID: 24818770 DOI: 10.1016/j.jconrel.2014.05.004] [Citation(s) in RCA: 254] [Impact Index Per Article: 23.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2014] [Revised: 04/24/2014] [Accepted: 05/02/2014] [Indexed: 01/03/2023]
Abstract
Novel microfluidic tools allow new ways to manufacture and test drug delivery systems. Organ-on-a-chip systems - microscale recapitulations of complex organ functions - promise to improve the drug development pipeline. This review highlights the importance of integrating microfluidic networks with 3D tissue engineered models to create organ-on-a-chip platforms, able to meet the demand of creating robust preclinical screening models. Specific examples are cited to demonstrate the use of these systems for studying the performance of drug delivery vectors and thereby reduce the discrepancies between their performance at preclinical and clinical trials. We also highlight the future directions that need to be pursued by the research community for these proof-of-concept studies to achieve the goal of accelerating clinical translation of drug delivery nanoparticles.
Collapse
Affiliation(s)
- Nupura S Bhise
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - João Ribas
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA; Doctoral Programme in Experimental Biology and Biomedicine, Center for Neuroscience and Cell Biology, Institute for Interdisciplinary Research, University of Coimbra, 3030-789 Coimbra, Portugal; Biocant - Biotechnology Innovation Center, 3060-197 Cantanhede, Portugal
| | - Vijayan Manoharan
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Yu Shrike Zhang
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Alessandro Polini
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Solange Massa
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Mehmet R Dokmeci
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA
| | - Ali Khademhosseini
- Division of Biomedical Engineering, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Cambridge, 02139, USA; Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, 02139, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston 02115, USA; Department of Physics, King Abdulaziz University, Jeddah 21569, Saudi Arabia.
| |
Collapse
|
44
|
Sun Z, Huang P, Tong G, Lin J, Jin A, Rong P, Zhu L, Nie L, Niu G, Cao F, Chen X. VEGF-loaded graphene oxide as theranostics for multi-modality imaging-monitored targeting therapeutic angiogenesis of ischemic muscle. NANOSCALE 2013; 5:6857-66. [PMID: 23770832 PMCID: PMC4607062 DOI: 10.1039/c3nr01573d] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Herein we report the design and synthesis of multifunctional VEGF-loaded IR800-conjugated graphene oxide (GO-IR800-VEGF) for multi-modality imaging-monitored therapeutic angiogenesis of ischemic muscle. The as-prepared GO-IR800-VEGF positively targets VEGF receptors, maintains an elevated level of VEGF in ischemic tissues for a prolonged time, and finally leads to remarkable therapeutic angiogenesis of ischemic muscle. Although more efforts are required to further understand the in vivo behaviors and the long-term toxicology of GO, our work demonstrates the success of using GO for efficient VEGF delivery in vivo by intravenous administration and suggests the great promise of using graphene oxide in theranostic applications for treating ischemic disease.
Collapse
Affiliation(s)
- Zhongchan Sun
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China 710032
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Peng Huang
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
- Research Institute of Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Guang Tong
- Department of Cardiovascular Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, China
| | - Jing Lin
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
- Research Institute of Micro/Nano Science and Technology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Albert Jin
- Laboratory of Cellular Imaging and Macromolecular Biophysics, National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Pengfei Rong
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Lei Zhu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
- Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361005 China
| | - Liming Nie
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| | - Feng Cao
- Department of Cardiology, Xijing Hospital, The Fourth Military Medical University, Xi'an, China 710032
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, MD 20892-2281, USA
| |
Collapse
|
45
|
Kim E, Stamatelos S, Cebulla J, Bhujwalla ZM, Popel AS, Pathak AP. Multiscale imaging and computational modeling of blood flow in the tumor vasculature. Ann Biomed Eng 2012; 40:2425-41. [PMID: 22565817 DOI: 10.1007/s10439-012-0585-5] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 04/27/2012] [Indexed: 12/30/2022]
Abstract
The evolution in our understanding of tumor angiogenesis has been the result of pioneering imaging and computational modeling studies spanning the endothelial cell, microvasculature and tissue levels. Many of these primary data on the tumor vasculature are in the form of images from pre-clinical tumor models that provide a wealth of qualitative and quantitative information in many dimensions and across different spatial scales. However, until recently, the visualization of changes in the tumor vasculature across spatial scales remained a challenge due to a lack of techniques for integrating micro- and macroscopic imaging data. Furthermore, the paucity of three-dimensional (3-D) tumor vascular data in conjunction with the challenges in obtaining such data from patients presents a serious hurdle for the development and validation of predictive, multiscale computational models of tumor angiogenesis. In this review, we discuss the development of multiscale models of tumor angiogenesis, new imaging techniques capable of reproducing the 3-D tumor vascular architecture with high fidelity, and the emergence of "image-based models" of tumor blood flow and molecular transport. Collectively, these developments are helping us gain a fundamental understanding of the cellular and molecular regulation of tumor angiogenesis that will benefit the development of new cancer therapies. Eventually, we expect this exciting integration of multiscale imaging and mathematical modeling to have widespread application beyond the tumor vasculature to other diseases involving a pathological vasculature, such as stroke and spinal cord injury.
Collapse
Affiliation(s)
- Eugene Kim
- Department of Biomedical Engineering, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | |
Collapse
|
46
|
Ohr M, Kaiser PK. Intravitreal aflibercept injection for neovascular (wet) age-related macular degeneration. Expert Opin Pharmacother 2012; 13:585-91. [DOI: 10.1517/14656566.2012.658368] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
47
|
Gene delivery nanoparticles specific for human microvasculature and macrovasculature. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2012; 8:1200-7. [PMID: 22306159 DOI: 10.1016/j.nano.2012.01.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2011] [Revised: 12/22/2011] [Accepted: 01/16/2012] [Indexed: 12/31/2022]
Abstract
UNLABELLED Endothelial cell dysfunction is a critical component of ocular diseases such as age-related macular degeneration and diabetic retinopathy. An important limitation in endothelial cell research is the difficulty in achieving efficient transfection of these cells. A new polymer library was here synthesized and utilized to find polymeric nanoparticles that can transfect macrovascular (human umbilical vein, HUVECs) and microvascular (human retinal, HRECs) endothelial cells. Nanoparticles were synthesized that can achieve transfection efficiency of up to 85% for HRECs and 65% for HUVECs. These nanoparticle systems enable high levels of expression while avoiding problems associated with viral gene delivery. The polymeric nanoparticles also show cell-specific behavior, with a high correlation between microvascular and macrovascular transfection (R(2) = 0.81) but low correlation between retinal endothelial and retinal epithelial transfection (R(2) = 0.21). These polymeric nanoparticles can be used in vitro as experimental tools and potentially in vivo to target and treat vascular-specific diseases. FROM THE CLINICAL EDITOR Polymeric nanoparticles were synthesized with the goal of transfecting endothelial cells, which are commonly considered difficult targets. The authors report excellent transfection efficiency of up to 85% for human retinal and 65% for human umbilical vein endothelial cells. These NPs can be used in vitro as experimental tools and potentially in vivo to target and treat vascular-specific diseases.
Collapse
|
48
|
Tzeng SY, Yang PH, Grayson WL, Green JJ. Synthetic poly(ester amine) and poly(amido amine) nanoparticles for efficient DNA and siRNA delivery to human endothelial cells. Int J Nanomedicine 2011; 6:3309-22. [PMID: 22228998 PMCID: PMC3252678 DOI: 10.2147/ijn.s27269] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Biodegradable poly(ester amine) (PEA)-based and poly(amido amine) (PAA)-based nanoparticles were developed for efficient in vitro siRNA delivery to human umbilical vein endothelial cells (HUVECs). They were screened, characterized, and compared with traditionally studied DNA-containing particles. Several of the polymeric nanoparticles tested were found to be effective for delivering functional siRNA to green fluorescent protein (GFP) + HUVECs, achieving 60%–75% GFP knockdown while maintaining high viability. While PEAs have been used previously to form polyplexes or nanoparticles for DNA delivery, highly effective siRNA delivery in hard-to-transfect human cell types has not been previously reported. PEAs and linear nondendrimeric PAAs were also found to be effective for DNA delivery to HUVECs using GFP-encoding plasmid DNA (up to 50%–60% transfection efficiency). PEAs and PAAs can be separated into groups that form polymeric nanoparticles effective for siRNA delivery, for DNA delivery, or for both.
Collapse
Affiliation(s)
- Stephany Y Tzeng
- Department of Biomedical Engineering and the Institute for NanoBioTechnology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, USA
| | | | | | | |
Collapse
|
49
|
Lee E, Rosca EV, Pandey NB, Popel AS. Small peptides derived from somatotropin domain-containing proteins inhibit blood and lymphatic endothelial cell proliferation, migration, adhesion and tube formation. Int J Biochem Cell Biol 2011; 43:1812-21. [PMID: 21920451 DOI: 10.1016/j.biocel.2011.08.020] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Revised: 07/18/2011] [Accepted: 08/30/2011] [Indexed: 11/27/2022]
Abstract
Angiogenesis is thoroughly balanced and regulated in health; however, it is dysregulated in many diseases including cancer, age-related macular degeneration, cardiovascular diseases such as coronary and peripheral artery diseases and stroke, abnormal embryonic development, and abnormal wound healing. In addition to angiogenesis, lymphangiogenesis is pivotal for maintaining the immune system, homeostasis of body fluids and lymphoid organs; dysregulated lymphangiogenesis may cause inflammatory diseases and lymph node mediated tumor metastasis. Anti-angiogenic or anti-lymphangiogenic small peptides may play an important role as therapeutic agents normalizing angiogenesis or lymphangiogenesis in disease conditions. Several novel endogenous peptides derived from proteins containing a conserved somatotropin domain have been previously identified with the help of our bioinformatics-based methodology. These somatotropin peptides were screened for inhibition of angiogenesis and lymphangiogenesis using in vitro proliferation, migration, adhesion and tube formation assays with blood and lymphatic endothelial cells. We found that the peptides have the potential for inhibiting both angiogenesis and lymphangiogenesis. Focusing the study on the inhibition of lymphangiogenesis, we found that a peptide derived from the somatotropin conserved domain of transmembrane protein 45A human was the most potent lymphangiogenesis inhibitor, blocking lymphatic endothelial cell migration, adhesion, and tube formation.
Collapse
Affiliation(s)
- Esak Lee
- Department of Chemical and Biomolecular Engineering, School of Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | | | | | | |
Collapse
|