1
|
Wu Z, Chang L, Li C, Xu P, Liu L, Tong A, Li Y, Tong C, Liu B. Prodigiosin Loaded SN-PB@PG NPs-Based Multimodal Therapy for the Healing of Bacterial Infected Chronic Wounds. Adv Healthc Mater 2025; 14:e2405100. [PMID: 40123202 DOI: 10.1002/adhm.202405100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Healing of infected chronic wounds faces dual challenges: persistent inflammation and impaired angiogenesis. To address these, SN-PB@PG nanocomplexes were prepared by hybridisation of nitroprusside (SNP) with Prussian blue (SN-PB NPs) and loaded with prodigiosin (PG). Under near-infrared (NIR) irradiation, SN-PB NPs generated mild hyperthermia, facilitating the release of nitric oxide (NO) and PG to combat bacterial biofilms and multidrug-resistant pathogens. The in vivo assay using diabetic infected wounds demonstrated that SN-PB@PG NPs with NIR reduced the wound area to 10.6% by the 11th day, which is superior to that of control group (29.6%). In the flap transplantation experiments, the data showed SN-PB@PG NPs with NIR group only have a necrobiosis of 3.8% of flaps on the 8th day, which is superior to 31.3% of the control group. Additionally, the release of NO promoted vascular regeneration by up-regulating vascular endothelial growth factor (VEGF) and platelet endothelial cell adhesion molecule-1 (CD31), and regenerated sarcomeric tissue by down-regulating MMP-9. The results indicated that the combination of SN-PB@PG NPs with gas and photothermal therapy exerted a combined antibacterial and wound healing effect. Compared to traditional clinical methods such as surgical debridement and hyperbaric oxygen therapy, this new strategy efficiently addresses issues of infection and healing, which is convenient for clinical application.
Collapse
Affiliation(s)
- Zhou Wu
- College of Biology, Hunan University, Changsha, 410082, China
| | - Li Chang
- Institute of Bast Fiber Crop Chinese Academy of Agriculture Sciences, Changsha, 410205, China
| | - Changling Li
- College of Biological and Food Engineering, Huaihua University, Huaihua, 418000, China
| | - Pengxiang Xu
- College of Biology, Hunan University, Changsha, 410082, China
| | - Liuying Liu
- College of Biology, Hunan University, Changsha, 410082, China
| | - Aidi Tong
- College of Biology, Hunan University, Changsha, 410082, China
| | - Yilong Li
- Zonsen PepLib Biotech Inc., Shifeng District, Zhuzhou, 412000, China
| | - Chunyi Tong
- College of Biology, Hunan University, Changsha, 410082, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, 410082, China
| |
Collapse
|
2
|
Zeng Z, Liu T, Zeng P, Xie Y, Li L, Tan J, Wang H, Liu S, Bian Q, Xiao H, Liang S, Chen J, Chen Y, Lu L. Enhancing vascular implants with heparin-polylysine-copper nanozyme coating for synergistic anticoagulation and antirestenotic activity. Int J Biol Macromol 2025; 309:143048. [PMID: 40216105 DOI: 10.1016/j.ijbiomac.2025.143048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 03/31/2025] [Accepted: 04/08/2025] [Indexed: 04/18/2025]
Abstract
Re-establishing blood flow through vascular implants, such as stents, faces significant challenges, including late stent thrombosis (LST) and in-stent restenosis (ISR). Strategies to overcome these issues focus on enhancing stent surfaces with anticoagulant, pro-endothelialization, and anti-neointimal hyperplasia (NIH) properties. However, achieving all of these functionalities typically requires complex surface modifications. In this study, we developed nanozyme particles by assembling heparin, polylysine (PLL) and copper ions, which catalytically release nitric oxide (NO) in situ. A functional coating was then formed on pre-deposited polydopamine (PDA) transition layer. Our nanozyme coating not only exhibits robust anticoagulant activity but also enables sustained, in situ release of NO, a critical gas molecule for maintaining vascular health and patency. In vitro results showed that the coating significantly inhibited platelet aggregation, remarkably prolonged activated partial thromboplastin time (APTT), and selectively promoted endothelial cell growth over smooth muscle cells. Ex vivo blood circulation models confirmed its superior anti-thrombotic efficacy, while in vivo experiments further validated its ability to facilitate endothelial regeneration and suppress NIH. This technology offers significant potential for improving the safety and outcomes of cardiovascular implants.
Collapse
Affiliation(s)
- Zheng Zeng
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China; Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Tao Liu
- South China Research Center for Acupuncture and Moxibustion, Medical College of Acu-Moxi and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Peiying Zeng
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Yinhong Xie
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Li Li
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Jianying Tan
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Huanran Wang
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Sainan Liu
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Qihao Bian
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China
| | - Hongkai Xiao
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Siyu Liang
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China
| | - Junying Chen
- Key Laboratory of Advanced Technology of Materials, Ministry of Education, Southwest Jiaotong University, Chengdu 610031, China.
| | - Yin Chen
- The Fourth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 511436, China.
| | - Lei Lu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou 325027, China.
| |
Collapse
|
3
|
de Almeida HV, Bomediano MP, Catori DM, Silva EHC, de Oliveira MG. Integrating 3D printing of biomaterials with nitric oxide release. Biomater Sci 2025; 13:858-874. [PMID: 39804041 DOI: 10.1039/d4bm01304b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2025]
Abstract
The pivotal roles played by nitric oxide (NO) in tissue repair, inflammation, and immune response have spurred the development of a wide range of NO-releasing biomaterials. More recently, 3D printing techniques have significantly broadened the potential applications of polymeric biomaterials in biomedicine. In this context, the development of NO-releasing biomaterials that can be fabricated through 3D printing techniques has emerged as a promising strategy for harnessing the benefits of localized NO release from implantable devices, tissue regeneration scaffolds, or bandages for topical applications. Although 3D printing techniques allow for the creation of polymeric constructs with versatile designs and high geometric precision, integrating NO-releasing functional groups or molecules into these constructs poses several challenges. NO donors, such as S-nitrosothiols (RSNOs) or diazeniumdiolates (NONOates), may release NO thermally, complicating their incorporation into resins that require heating for extrusion-based 3D printing. Conversely, NO released photochemically from RSNOs effectively inhibits radical propagation, thus hindering photoinduced 3D printing processes. This review outlines the primary strategies employed to overcome these challenges in developing NO-releasing biomaterials via 3D printing, and explores future prospects in this rapidly evolving field.
Collapse
Affiliation(s)
- Herllan V de Almeida
- Institute of Chemistry, University of Campinas, UNICAMP, Campinas 13083-970, São Paulo, Brazil.
| | - Mateus P Bomediano
- Institute of Chemistry, University of Campinas, UNICAMP, Campinas 13083-970, São Paulo, Brazil.
| | - Daniele M Catori
- School of Chemical, Materials and Biomedical Engineering, College of Engineering, University of Georgia, Athens 30602, Georgia, USA
| | - Elizaura H C Silva
- Institute of Chemistry, University of Campinas, UNICAMP, Campinas 13083-970, São Paulo, Brazil.
| | - Marcelo G de Oliveira
- Institute of Chemistry, University of Campinas, UNICAMP, Campinas 13083-970, São Paulo, Brazil.
| |
Collapse
|
4
|
Zhang RR, Ran XY, Yu KK, Zhao Y, Zhang LN, Lv XF, Zhang H, Yu XQ, Li K. Rational Design of NIR-II Fluorescence/Photoacoustic Nanosensor Tailored for Mechanisms of Diabetes-Related Breast Cancer. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2415891. [PMID: 39757524 DOI: 10.1002/adma.202415891] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/12/2024] [Indexed: 01/07/2025]
Abstract
Breast cancer (BC) is the second most common cause of cancer induced death worldwide. Current statistics has disclosed that the diabetic BC patients have significantly worse survival rate compared with nondiabetic BC patients. However, the specific mechanism is still being explored. Herein, a novel NIR-II nanosensor DNPS for nitric oxide (NO) with fluorescence/photoacoustic (FL/PA) imaging capability is developed to explore the mechanism by which diabetes promoting breast cancer progression. In diabetic BC model, DNPS exhibits great advantages of low intrinsic background, high sensitivity, and deep tissue penetration and successfully confirmed the expression level of NO is higher than BC model, indicating that diabetes causes elevated nitric oxide levels in the tumor microenvironment. RNA-seq analysis results show that hyperglycemia caused by diabetes leads to weakened immune response and initiates the transcription and translation of the inducible nitric oxide synthase (iNOS) gene to produce NO. Besides, the increased expression of carcinogens related to Nitric oxide synthase 2 (Nos2), such as Spp1, Mmp11, and Kitl, causes breast cancer to develop more rapidly. Here, NIR-II imaging probe is applied first to study diabetes-related breast cancer and certain reference value is provided for subsequent research on the mechanism of diabetes promoting the progression of breast cancer.
Collapse
Affiliation(s)
- Rui-Rui Zhang
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Xiao-Yun Ran
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Kang-Kang Yu
- Key Laboratory of Bio-Resources and Eco-Environment, Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Yu Zhao
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Li-Na Zhang
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Xiao-Fang Lv
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| | - Hong Zhang
- Huaxi MR Research Center (HMRRC), Department of Radiology, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Xiao-Qi Yu
- Department of Chemistry, Xihua University, Chengdu, 610039, P. R. China
| | - Kun Li
- Key Laboratory of Green Chemistry and Technology, Ministry of Education, College of Chemistry, Sichuan University, Chengdu, 610064, China
| |
Collapse
|
5
|
Zhou K, Huang C, Li J, Dai W, Lu Z, Jia F, Jin Q, Shang M, Ji J, Fu G. Multifunctional NO supramolecular nanomedicine for thrombus risk reduction and intimal hyperplasia inhibition. J Mater Chem B 2025; 13:1811-1822. [PMID: 39744991 DOI: 10.1039/d4tb02271h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Cardiovascular diseases (CVDs) are the foremost cause of mortality worldwide, with incidence and mortality rates persistently climbing despite extensive research efforts. Innovative therapeutic approaches are still needed to extend patients' lives and preserve their health. In the present study, novel supramolecular nanomedicine with both nitric oxide (NO) and antioxidant releasing ability was developed to enhance therapeutic efficacy against vascular injuries. Utilizing α-cyclodextrin (α-CD) as a host molecule, the nanomedicine aims to achieve site-specific delivery via targeting peptide rendered selective accumulation. By scavenging ROS and amplifying NO's regulatory effects, the nanomedicine readily restored vascular homeostasis, in both acute and chronic CVDs. It served as a promising solution to overcome the challenges associated with NO-based therapies and may inspire future research studies on new therapies for CVDs.
Collapse
Affiliation(s)
- Kuangshi Zhou
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou 310016, China
| | - Chengchen Huang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou 310016, China
| | - Jian Li
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou 310016, China
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Wenbin Dai
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Zhaoyang Lu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou 310016, China
| | - Fan Jia
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou 310016, China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Min Shang
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou 310016, China
| | - Jian Ji
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou 310058, P. R. China.
| | - Guosheng Fu
- Department of Cardiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China.
- Zhejiang Key Laboratory of Cardiovascular Intervention and Precision Medicine, Hangzhou 310016, China
- Engineering Research Center for Cardiovascular Innovative Devices of Zhejiang Province, Hangzhou 310016, China
| |
Collapse
|
6
|
Witzdam L, White T, Rodriguez-Emmenegger C. Steps Toward Recapitulating Endothelium: A Perspective on the Next Generation of Hemocompatible Coatings. Macromol Biosci 2024; 24:e2400152. [PMID: 39072925 DOI: 10.1002/mabi.202400152] [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: 03/31/2024] [Revised: 06/26/2024] [Indexed: 07/30/2024]
Abstract
Endothelium, the lining in this blood vessel, orchestrates three main critical functions such as protecting blood components, modulating of hemostasis by secreting various inhibitors, and directing clot digestion (fibrinolysis) by activating tissue plasminogen activator. No other surface can perform these tasks; thus, the contact of blood and blood-contacting medical devices inevitably leads to the activation of coagulation, often causing device failure, and thromboembolic complications. This perspective, first, discusses the biological mechanisms of activation of coagulation and highlights the efforts of advanced coatings to recapitulate one characteristic of endothelium, hereafter single functions of endothelium and noting necessity of the synergistic integration of its three main functions. Subsequently, it is emphasized that to overcome the challenges of blood compatibility an endothelium-mimicking system is needed, proposing a synergy of bottom-up synthetic biology, particularly synthetic cells, with passive- and bioactive surface coatings. Such integration holds promise for developing advanced biomaterials capable of recapitulating endothelial functions, thereby enhancing the hemocompatibility and performance of blood-contacting medical devices.
Collapse
Affiliation(s)
- Lena Witzdam
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, Barcelona, 08028, Spain
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, 52074, Aachen, Germany
- Institute of Technical and Macromolecular Chemistry, RWTH Aachen University, Worringerweg 2, 52074, Aachen, Germany
| | - Tom White
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, Barcelona, 08028, Spain
| | - Cesar Rodriguez-Emmenegger
- Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Carrer de Baldiri Reixac, 10, 12, Barcelona, 08028, Spain
- DWI - Leibniz Institute for Interactive Materials, Forckenbeckstraße 50, 52074, Aachen, Germany
- Catalan Institution for Research and Advanced Studies (ICREA), Passeig Lluís Companys 23, Barcelona, 08010, Spain
- Biomedical Research Networking, Center in Bioengineering, Biomaterials and Nanomedicine, The Institute of Health Carlos III, Madrid, 28029, Spain
| |
Collapse
|
7
|
Cheng Y, Huo Y, Yu Y, Duan P, Dong X, Yu Z, Cheng Q, Dai H, Pan Z. A photothermal responsive system accelerating nitric oxide release to enhance bone repair by promoting osteogenesis and angiogenesis. Mater Today Bio 2024; 28:101180. [PMID: 39221216 PMCID: PMC11364911 DOI: 10.1016/j.mtbio.2024.101180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/27/2024] [Accepted: 08/03/2024] [Indexed: 09/04/2024] Open
Abstract
Managing bone defects remains a formidable clinical hurdle, primarily attributed to the inadequate orchestration of vascular reconstruction and osteogenic differentiation in both spatial and temporal dimensions. This challenge persists due to the constrained availability of autogenous grafts and the limited regenerative capacity of allogeneic or synthetic bone substitutes, thus necessitating continual exploration and innovation in the realm of functional and bioactive bone graft materials. While synthetic scaffolds have emerged as promising carriers for bone grafts, their efficacy is curtailed by deficiencies in vascularization and osteoinductive potential. Nitric oxide (NO) plays a key role in revascularization and bone tissue regeneration, yet studies related to the use of NO for the treatment of bone defects remain scarce. Herein, we present a pioneering approach leveraging a photothermal-responsive system to augment NO release. This system comprises macromolecular mPEG-P nanoparticles encapsulating indocyanine green (ICG) (NO-NPs@ICG) and a mPEG-PA-PP injectable thermosensitive hydrogel carrier. By harnessing the synergistic photothermal effects of near-infrared radiation and ICG, the system achieves sustained NO release, thereby activating the soluble guanylate cyclase (SGC)-cyclic guanosine monophosphate (cGMP) signaling pathway both in vitro and in vivo. This orchestrated cascade culminates in the facilitation of angiogenesis and osteogenesis, thus expediting the reparative processes in bone defects. In a nutshell, the NO release-responsive system elucidated in this study presents a pioneering avenue for refining the bone tissue microenvironment and fostering enhanced bone regeneration.
Collapse
Affiliation(s)
- Yannan Cheng
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Yuanfang Huo
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan, 430070, China
| | - Yongle Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Ping Duan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Xianzhen Dong
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan, 430070, China
| | - Zirui Yu
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| | - Qiang Cheng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan, 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan, 430070, China
- Shenzhen Research Institute of Wuhan University of Technology, Shenzhen, 518000, China
| | - Zhenyu Pan
- Department of Orthopedics Trauma and Microsurgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, China
| |
Collapse
|
8
|
Jia P, Zou Y, Jiang J. S-Nitrosylated CuS Hybrid Hydrogel Patches with Robust Antibacterial and Repair-Promoting Activity for Infected Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307629. [PMID: 38073365 DOI: 10.1002/smll.202307629] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/07/2023] [Indexed: 05/25/2024]
Abstract
Development of wound dressing with robust antibacterial and repair-promoting activity has always been an urgent biomedical task during the past years. The therapeutic effect of current hydrogel dressings containing single bioactive agent like nanoparticle or gas is still unsatisfactory for the treatment of infected wound. Herein, a CuS/NO co-loaded hydrogel (CuS/NO Gel) is proposed, which is constructed by sequential polymerization, reduction, and S-nitrosylation of CuS hybrid hydrogel with disulfide bonds. These CuS/NO Gel patches show good mechanical stability, high photothermal activity and excellent biocompatibility. When being applied to treat infected wound, CuS/NO Gel can not only eliminate infection effectively by the synergistic effect of mild photothermal heating and boosted NO release in infection phase, but also promote vascularization and collagen deposition due to the synchronous supply of Cu ion nutrients and low concentration NO signaling molecules in wound repair phase. Compared to hydrogel dressings with individual active agent (CuS Gel or NO Gel), CuS/NO Gel exhibits better antibacterial and repair-promoting activity both in vitro and in vivo. Therefore, this CuS/NO Gel holds great promise in the future clinical treatment against infected wound.
Collapse
Affiliation(s)
- Pengpeng Jia
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- i-Lab, CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Yu Zou
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- i-Lab, CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| | - Jiang Jiang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei, 230026, China
- i-Lab, CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, 215123, China
| |
Collapse
|
9
|
Bhowmik R, Roy M. Recent advances on the development of NO-releasing molecules (NORMs) for biomedical applications. Eur J Med Chem 2024; 268:116217. [PMID: 38367491 DOI: 10.1016/j.ejmech.2024.116217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 02/02/2024] [Accepted: 02/02/2024] [Indexed: 02/19/2024]
Abstract
Nitric oxide (NO) is an important biological messenger as well as a signaling molecule that participates in a broad range of physiological events and therapeutic applications in biological systems. However, due to its very short half-life in physiological conditions, its therapeutic applications are restricted. Efforts have been made to develop an enormous number of NO-releasing molecules (NORMs) and motifs for NO delivery to the target tissues. These NORMs involve organic nitrate, nitrite, nitro compounds, transition metal nitrosyls, and several nanomaterials. The controlled release of NO from these NORMs to the specific site requires several external stimuli like light, sound, pH, heat, enzyme, etc. Herein, we have provided a comprehensive review of the biochemistry of nitric oxide, recent advancements in NO-releasing materials with the appropriate stimuli of NO release, and their biomedical applications in cancer and other disease control.
Collapse
Affiliation(s)
- Rintu Bhowmik
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur, India
| | - Mithun Roy
- Department of Chemistry, National Institute of Technology Manipur, Langol, 795004, Imphal West, Manipur, India.
| |
Collapse
|
10
|
Ali R, Sen S, Hameed R, Nazir A, Verma S. Strategies for gaseous neuromodulator release in chemical neuroscience: Experimental approaches and translational validation. J Control Release 2024; 365:132-160. [PMID: 37972768 DOI: 10.1016/j.jconrel.2023.11.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 11/03/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
Gasotransmitters are a group of short-lived gaseous signaling molecules displaying diverse biological functions depending upon their localized concentration. Nitric oxide (NO), hydrogen sulfide (H2S), and carbon monoxide (CO) are three important examples of endogenously produced gasotransmitters that play a crucial role in human neurophysiology and pathogenesis. Alterations in their optimal physiological concentrations can lead to various severe pathophysiological consequences, including neurological disorders. Exogenous administration of gasotransmitters has emerged as a prominent therapeutic approach for treating such neurological diseases. However, their gaseous nature and short half-life limit their therapeutic delivery. Therefore, developing synthetic gasotransmitter-releasing strategies having control over the release and duration of these gaseous molecules has become imperative. However, the complex chemistry of synthesis and the challenges of specific quantified delivery of these gases, make their therapeutic application a challenging task. This review article provides a focused overview of emerging strategies for delivering gasotransmitters in a controlled and sustained manner to re-establish neurophysiological homeostasis.
Collapse
Affiliation(s)
- Rafat Ali
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Shantanu Sen
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India
| | - Rohil Hameed
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow 226031, UP, India.
| | - Sandeep Verma
- Department of Chemistry, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India; Center for Nanoscience, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India; Mehta Family Center for Engineering in Medicine, Indian Institute of Technology Kanpur, Kanpur 208016, UP, India.
| |
Collapse
|
11
|
Sheet PS, Lautner G, Meyerhoff ME, Schwendeman SP. Mechanistic analysis of the photolytic decomposition of solid-state S-nitroso-N-acetylpenicillamine. Nitric Oxide 2024; 142:38-46. [PMID: 37979933 PMCID: PMC12103906 DOI: 10.1016/j.niox.2023.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 09/28/2023] [Accepted: 11/02/2023] [Indexed: 11/20/2023]
Abstract
S-Nitroso-N-acetylpenicillamine (SNAP) is among the most common nitric oxide (NO)-donor molecules and its solid-state photolytic decomposition has potential for inhaled nitric oxide (iNO) therapy. The photochemical NO release kinetics and mechanism were investigated by exposing solid-state SNAP to a narrow-band LED as a function of nominal wavelength and intensity of incident light. The photolytic efficiency, decomposition products, and the photolytic pathways of the SNAP were examined. The maximum light penetration depth through the solid layer of SNAP was determined by an optical microscope and found to be within 100-200 μm, depending on the wavelength of light. The photolysis of solid-state SNAP to generate NO along with the stable thiyl (RS·) radical was confirmed using Electron Spin Resonance (ESR) spectroscopy. The fate of the RS· radical in the solid phase was studied both in the presence and absence of O2 using NMR, IR, ESR, and UPLC-MS. The changes in the morphology of SNAP due to its photolysis were examined using PXRD and SEM. The stable thiyl radical formed from the photolysis of solid SNAP was found to be reactive with another adjacent thiyl radical to form a disulfide (RSSR) or with oxygen to form various sulfonyl and sulfonyl peroxyl radicals {RS(O)xO·, x = 0 to 7}. However, the thiyl radical did not recombine with NO to reform the SNAP. From the PXRD data, it was found that the SNAP loses its crystallinity by generating the NO after photolysis. The initial release of NO during photolysis was increased with increased intensity of light, whereas the maximum light penetration depth was unaffected by light intensity. The knowledge gained about the photochemical reactions of SNAP may provide important insight in designing portable photoinduced NO-releasing devices for iNO therapy.
Collapse
Affiliation(s)
- Partha S Sheet
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Gergely Lautner
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
| | - Mark E Meyerhoff
- Department of Chemistry, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
12
|
Mu Z, Jin T, Chu T, Lu H, Chen Y, Li S, Zeng B, Huang C, Lei K, Cai X, Deng H, Hu R. Functionalized MoS 2-nanosheets with NIR-Triggered nitric oxide delivery and photothermal activities for synergistic antibacterial and regeneration-promoting therapy. J Nanobiotechnology 2023; 21:463. [PMID: 38044437 PMCID: PMC10694958 DOI: 10.1186/s12951-023-02167-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/18/2023] [Indexed: 12/05/2023] Open
Abstract
Bacterial infection in skin and soft tissue has emerged as a critical concern. Overreliance on antibiotic therapy has led to numerous challenges, including the emergence of multidrug-resistant bacteria and adverse drug reactions. It is imperative to develop non-antibiotic treatment strategies that not only exhibit potent antibacterial properties but also promote rapid wound healing and demonstrate biocompatibility. Herein, a novel multimodal synergistic antibacterial system (SNO-CS@MoS2) was developed. This system employs easily surface-modified thin-layer MoS2 as photothermal agents and loaded with S-nitrosothiol-modified chitosan (SNO-CS) via electrostatic interactions, thus realizing the combination of NO gas therapy and photothermal therapy (PTT). Furthermore, this surface modification renders SNO-CS@MoS2 highly stable and capable of binding with bacteria. Through PTT's thermal energy, SNO-CS@MoS2 rapidly generates massive NO, collaborating with PTT to achieve antibacterial effects. This synergistic therapy can swiftly disrupt the bacterial membrane, causing protein leakage and ATP synthesis function damage, ultimately eliminating bacteria. Notably, after effectively eliminating all bacteria, the residual SNO-CS@MoS2 can create trace NO to promote fibroblast migration, proliferation, and vascular regeneration, thereby accelerating wound healing. This study concluded that SNO-CS@MoS2, a novel multifunctional nanomaterial with outstanding antibacterial characteristics and potential to promote wound healing, has promising applications in infected soft tissue wound treatment.
Collapse
Affiliation(s)
- Zhixiang Mu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Ting Jin
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Tengda Chu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Hongyang Lu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Yuanqi Chen
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Sisi Li
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - BaiRui Zeng
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Chen Huang
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Kezheng Lei
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China
| | - Xiaojun Cai
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China.
| | - Hui Deng
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China.
| | - Rongdang Hu
- School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, P.R. China.
| |
Collapse
|
13
|
Wang K, Mao W, Song X, Chen M, Feng W, Peng B, Chen Y. Reactive X (where X = O, N, S, C, Cl, Br, and I) species nanomedicine. Chem Soc Rev 2023; 52:6957-7035. [PMID: 37743750 DOI: 10.1039/d2cs00435f] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/26/2023]
Abstract
Reactive oxygen, nitrogen, sulfur, carbonyl, chlorine, bromine, and iodine species (RXS, where X = O, N, S, C, Cl, Br, and I) have important roles in various normal physiological processes and act as essential regulators of cell metabolism; their inherent biological activities govern cell signaling, immune balance, and tissue homeostasis. However, an imbalance between RXS production and consumption will induce the occurrence and development of various diseases. Due to the considerable progress of nanomedicine, a variety of nanosystems that can regulate RXS has been rationally designed and engineered for restoring RXS balance to halt the pathological processes of different diseases. The invention of radical-regulating nanomaterials creates the possibility of intriguing projects for disease treatment and promotes advances in nanomedicine. In this comprehensive review, we summarize, discuss, and highlight very-recent advances in RXS-based nanomedicine for versatile disease treatments. This review particularly focuses on the types and pathological effects of these reactive species and explores the biological effects of RXS-based nanomaterials, accompanied by a discussion and the outlook of the challenges faced and future clinical translations of RXS nanomedicines.
Collapse
Affiliation(s)
- Keyi Wang
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China.
| | - Weipu Mao
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, P. R. China
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Ming Chen
- Department of Urology, Affiliated Zhongda Hospital of Southeast University, Nanjing, 210009, P. R. China
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Bo Peng
- Department of Urology, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, 200072, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
14
|
Andrabi SM, Sharma NS, Karan A, Shahriar SMS, Cordon B, Ma B, Xie J. Nitric Oxide: Physiological Functions, Delivery, and Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303259. [PMID: 37632708 PMCID: PMC10602574 DOI: 10.1002/advs.202303259] [Citation(s) in RCA: 156] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Indexed: 08/28/2023]
Abstract
Nitric oxide (NO) is a gaseous molecule that has a central role in signaling pathways involved in numerous physiological processes (e.g., vasodilation, neurotransmission, inflammation, apoptosis, and tumor growth). Due to its gaseous form, NO has a short half-life, and its physiology role is concentration dependent, often restricting its function to a target site. Providing NO from an external source is beneficial in promoting cellular functions and treatment of different pathological conditions. Hence, the multifaceted role of NO in physiology and pathology has garnered massive interest in developing strategies to deliver exogenous NO for the treatment of various regenerative and biomedical complexities. NO-releasing platforms or donors capable of delivering NO in a controlled and sustained manner to target tissues or organs have advanced in the past few decades. This review article discusses in detail the generation of NO via the enzymatic functions of NO synthase as well as from NO donors and the multiple biological and pathological processes that NO modulates. The methods for incorporating of NO donors into diverse biomaterials including physical, chemical, or supramolecular techniques are summarized. Then, these NO-releasing platforms are highlighted in terms of advancing treatment strategies for various medical problems.
Collapse
Affiliation(s)
- Syed Muntazir Andrabi
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Navatha Shree Sharma
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Anik Karan
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - S. M. Shatil Shahriar
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Brent Cordon
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bing Ma
- Cell Therapy Manufacturing FacilityMedStar Georgetown University HospitalWashington, DC2007USA
| | - Jingwei Xie
- Department of Surgery‐Transplant and Mary & Dick Holland Regenerative Medicine ProgramCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of Mechanical and Materials EngineeringCollege of EngineeringUniversity of Nebraska LincolnLincolnNE68588USA
| |
Collapse
|
15
|
Zhou R, Chang M, Shen M, Cong Y, Chen Y, Wang Y. Sonocatalytic Optimization of Titanium-Based Therapeutic Nanomedicine. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301764. [PMID: 37395421 PMCID: PMC10477905 DOI: 10.1002/advs.202301764] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 05/28/2023] [Indexed: 07/04/2023]
Abstract
Recent considerable technological advances in ultrasound-based treatment modality provides a magnificent prospect for scientific communities to conquer the related diseases, which is featured with remarkable tissue penetration, non-invasive and non-thermal characteristics. As one of the critical elements that influences treatment outcomes, titanium (Ti)-based sonosensitizers with distinct physicochemical properties and exceptional sonodynamic efficiency have been applied extensively in the field of nanomedical applications. To date, a myriad of methodologies has been designed to manipulate the sonodynamic performance of titanium-involved nanomedicine and further enhance the productivity of reactive oxygen species for disease treatments. In this comprehensive review, the sonocatalytic optimization of diversified Ti-based nanoplatforms, including defect engineering, plasmon resonance modulation, heterojunction, modulating tumor microenvironment, as well as the development of synergistic therapeutic modalities is mainly focused. The state-of-the-art Ti-based nanoplatforms ranging from preparation process to the extensive medical applications are summarized and highlighted, with the goal of elaborating on future research prospects and providing a perspective on the bench-to-beside translation of these sonocatalytic optimization tactics. Furthermore, to spur further technological advancements in nanomedicine, the difficulties currently faced and the direction of sonocatalytic optimization of Ti-based therapeutic nanomedicine are proposed and outlooked.
Collapse
Affiliation(s)
- Ruirui Zhou
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| | - Meiqi Chang
- Laboratory CenterShanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071P. R. China
| | - Mengjun Shen
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| | - Yang Cong
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| | - Yu Chen
- Materdicine LabSchool of Life SciencesShanghai UniversityShanghai200444P. R. China
| | - Yin Wang
- Department of UltrasoundShanghai Pulmonary HospitalSchool of MedicineTongji UniversityShanghai200433P. R. China
| |
Collapse
|
16
|
Ma W, Liu X, Yang M, Hong Q, Meng L, Zhang Q, Chen J, Pan C. Fabrication of CO-releasing surface to enhance the blood compatibility and endothelialization of TiO 2 nanotubes on titanium surface. BIOMATERIALS ADVANCES 2023; 149:213393. [PMID: 36966654 DOI: 10.1016/j.bioadv.2023.213393] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/10/2023] [Accepted: 03/15/2023] [Indexed: 05/02/2023]
Abstract
Although the construction of nanotube arrays with the micro-nano structures on the titanium surfaces has demonstrated a great promise in the field of blood-contacting materials and devices, the limited surface hemocompatibility and delayed endothelial healing should be further improved. Carbon monoxide (CO) gas signaling molecule within the physiological concentrations has excellent anticoagulation and the ability to promote endothelial growth, exhibiting the great potential for the blood-contact biomaterials, especially the cardiovascular devices. In this study, the regular titanium dioxide nanotube arrays were firstly prepared in situ on the titanium surface by anodic oxidation, followed by the immobilization of the complex of sodium alginate/carboxymethyl chitosan (SA/CS) on the self-assembled modified nanotube surface, the CO-releasing molecule (CORM-401) was finally grafted onto the surface to create a CO-releasing bioactive surface to enhance the biocompatibility. The results of scanning electron microscopy (SEM), X-ray energy dispersion spectroscopy (EDS), and X-ray photoelectron spectroscopy (XPS) revealed that the CO-releasing molecules were successfully immobilized on the surface. The modified nanotube arrays not only exhibited excellent hydrophilicity but also could slowly release CO gas molecules, and the amount of CO release increased when cysteine was added. Furthermore, the nanotube array can promote albumin adsorption while inhibit fibrinogen adsorption to some extent, demonstrating its selective albumin adsorption; although this effect was somewhat reduced by the introduction of CORM-401, it can be significantly enhanced by the catalytic release of CO. The results of hemocompatibility and endothelial cell growth behaviors showed that, as compared with the CORM-401 modified sample, although the SA/CS-modified sample had better biocompatibility, in the case of cysteine-catalyzed CO release, the released CO could not only reduce the platelet adhesion and activation as well as hemolysis rate, but also promote endothelial cell adhesion and proliferation as well as vascular endothelial growth factor (VEGF) and nitric oxide (NO) expression. As a result, the research of the present study demonstrated that the releasing CO from TiO2 nanotubes can simultaneously enhance the surface hemocompatibility and endothelialization, which could open a new route to enhance the biocompatibility of the blood-contacting materials and devices, such as the artificial heart valve and cardiovascular stents.
Collapse
Affiliation(s)
- Wenfu Ma
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Xuhui Liu
- The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an 223003, China
| | - Minhui Yang
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Qingxiang Hong
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Lingjie Meng
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Qiuyang Zhang
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China.
| | - Jie Chen
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China
| | - Changjiang Pan
- Faculty of Mechanical and Material Engineering, Jiangsu Provincial Engineering Research Center for Biomaterials and Advanced Medical Devices, Huaiyin Institute of Technology, Huai'an 223003, China.
| |
Collapse
|
17
|
Ji H, Li Y, Su B, Zhao W, Kizhakkedathu JN, Zhao C. Advances in Enhancing Hemocompatibility of Hemodialysis Hollow-Fiber Membranes. ADVANCED FIBER MATERIALS 2023; 5:1-43. [PMID: 37361105 PMCID: PMC10068248 DOI: 10.1007/s42765-023-00277-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 02/19/2023] [Indexed: 06/28/2023]
Abstract
Hemodialysis, the most common modality of renal replacement therapy, is critically required to remove uremic toxins from the blood of patients with end-stage kidney disease. However, the chronic inflammation, oxidative stress as well as thrombosis induced by the long-term contact of hemoincompatible hollow-fiber membranes (HFMs) contribute to the increase in cardiovascular diseases and mortality in this patient population. This review first retrospectively analyzes the current clinical and laboratory research progress in improving the hemocompatibility of HFMs. Details on different HFMs currently in clinical use and their design are described. Subsequently, we elaborate on the adverse interactions between blood and HFMs, involving protein adsorption, platelet adhesion and activation, and the activation of immune and coagulation systems, and the focus is on how to improve the hemocompatibility of HFMs in these aspects. Finally, challenges and future perspectives for improving the hemocompatibility of HFMs are also discussed to promote the development and clinical application of new hemocompatible HFMs. Graphical Abstract
Collapse
Affiliation(s)
- Haifeng Ji
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 People’s Republic of China
- Department of Pathology and Lab Medicine & Center for Blood Research & Life Science Institute, 2350 Health Sciences Mall, Life Sciences Centre, The School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Yupei Li
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041 China
- Institute for Disaster Management and Reconstruction, Sichuan University, Chengdu, 610207 China
| | - Baihai Su
- Department of Nephrology, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Weifeng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 People’s Republic of China
| | - Jayachandran N. Kizhakkedathu
- Department of Pathology and Lab Medicine & Center for Blood Research & Life Science Institute, 2350 Health Sciences Mall, Life Sciences Centre, The School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3 Canada
| | - Changsheng Zhao
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065 People’s Republic of China
| |
Collapse
|
18
|
Navale GR, Singh S, Ghosh K. NO donors as the wonder molecules with therapeutic potential: Recent trends and future perspectives. Coord Chem Rev 2023. [DOI: 10.1016/j.ccr.2023.215052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
|
19
|
Zhang Y, Liu W, Wang X, Liu Y, Wei H. Nanozyme-Enabled Treatment of Cardio- and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2204809. [PMID: 36192166 DOI: 10.1002/smll.202204809] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/18/2022] [Indexed: 06/16/2023]
Abstract
Cardio- and cerebrovascular diseases are two major vascular-related diseases that lead to death worldwide. Reactive oxygen species (ROS) play a vital role in the occurrence and exacerbation of diseases. Excessive ROS induce cellular context damage and lead to tissue dysfunction. Nanozymes, as emerging enzyme mimics, offer a unique perspective for therapy through multifunctional activities, achieving essential results in the treatment of ROS-related cardio- and cerebrovascular diseases by directly scavenging excess ROS or regulating pathologically related molecules. This review first introduces nanozyme-enabled therapeutic mechanisms at the cellular level. Then, the therapies for several typical cardio- and cerebrovascular diseases with nanozymes are discussed, mainly including cardiovascular diseases, ischemia reperfusion injury, and neurological disorders. Finally, the challenges and outlooks for the application of nanozymes are also presented. This review will provide some instructive perspectives on nanozymes and promote the development of enzyme-mimicking strategies in cardio- and cerebrovascular disease therapy.
Collapse
Affiliation(s)
- Yihong Zhang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Wanling Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Xiaoyu Wang
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- Department of Chemistry and Material Science, College of Science, Nanjing Forestry University, Nanjing, Jiangsu, 210037, China
| | - Yufeng Liu
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
| | - Hui Wei
- College of Engineering and Applied Sciences, Nanjing National Laboratory of Microstructures, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing, Jiangsu, 210023, China
- State Key Laboratory of Analytical Chemistry for Life Science, School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing, Jiangsu, 210023, China
| |
Collapse
|
20
|
Li G, Yang T, Liu Y, Su H, Liu W, Fang D, Jin L, Jin F, Xu T, Duan C. The proteins derived from platelet-rich plasma improve the endothelialization and vascularization of small diameter vascular grafts. Int J Biol Macromol 2023; 225:574-587. [PMID: 36395946 DOI: 10.1016/j.ijbiomac.2022.11.116] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/26/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022]
Abstract
Vascular transplantation has become an ideal substitute for heart and peripheral vascular bypass therapy and tissue-engineered vascular grafts (TEVGs) present an attractive potential solution for vascular surgery. However, small diameter (Ф < 6 mm) vascular do not have ideal TEVGs for clinical use. Platelet-rich plasma (PRP), a key source of bioactive molecules, has been confirmed to promote tissue repair and regeneration. In this study, we prepared PRP-loaded TEVGs (PRP-TEVGs) by electrospinning, investigated the characterization of TEVGs, and verified the effect of PRP-TEVGs in vivo and in vitro experiments. The results suggested that PRP-TEVGs had good biocompatibility, released growth factors stably, promoted cell proliferation and migration significantly, up-regulated the expression of endothelial NO synthase (eNOS) in functional vascular endothelial cells (VECs), and maintained the stability of the endothelial structure. In vivo experiments suggest that PRP can promote rapid endothelialization and reconstruction of TEVGs. Overall, this finding indicated that PRP could promote the rapid vascular endothelialization of small-diameter TEVGs by improving contractile vascular smooth muscle cells (VSMCs) regeneration, and maintaining the integrity and functionality of VECs.
Collapse
Affiliation(s)
- Guangxu Li
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Yang
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Yanchao Liu
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Hengxian Su
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Wenchao Liu
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Dazhao Fang
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Lei Jin
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Fa Jin
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China
| | - Tao Xu
- Tsinghua-Berkeley Shenzhen Institute (TBSI), Tsinghua University, Shenzhen 518055, China; East China Institute of Digital Medical Engineering, Shangrao 334000, China.
| | - Chuanzhi Duan
- Neurosurgery Center, Department of Cerebrovascular Surgery, The National Key Clinical Specialty, The Engineering Technology Research Center of Education Ministry of China on Diagnosis and Treatment of Cerebrovascular Disease, Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, The Neurosurgery Institute of Guangdong Province, Zhujiang Hospital, Southern Medical University, Guangzhou 510282, China.
| |
Collapse
|
21
|
Zhao Y, Luo L, Huang L, Zhang Y, Tong M, Pan H, Shangguan J, Yao Q, Xu S, Xu H. In situ hydrogel capturing nitric oxide microbubbles accelerates the healing of diabetic foot. J Control Release 2022; 350:93-106. [PMID: 35973472 DOI: 10.1016/j.jconrel.2022.08.018] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 07/18/2022] [Accepted: 08/10/2022] [Indexed: 01/15/2023]
Abstract
Diabetic foot ulcer (DFU) is a devastating complication in diabetes patients, imposing a high risk of amputation and economic burden on patients. Sustained inflammation and angiogenesis hindrance are thought to be two key drivers of the pathogenesis of such ulcers. Nitric oxide (NO) has been proven to accelerate the healing of acute or chronic wounds by modulating inflammation and angiogenesis. However, the use of gas-based therapeutics is difficult for skin wounds. Herein, therapeutic NO gas was first prepared as stable microbubbles, followed by incorporation into a cold Poloxamer-407 (P407) solution. Exposed to the DFU wound, the cold P407 solution would rapidly be transformed into a semisolid hydrogel under body temperature and accordingly capture NO microbubbles. The NO microbubble-captured hydrogel (PNO) was expected to accelerate wound healing in diabetic feet. The NO microbubbles had an average diameter of 0.8 ± 0.4 μm, and most of which were captured by the in situ P407 hydrogel. Moreover, the NO microbubbles were evenly distributed inside the hydrogel and kept for a longer time. In addition, the gelling temperature of 30% (w/v) P407 polymer (21 °C) was adjusted to 31 °C for the PNO gel, which was near the temperature of the skin surface. Rheologic studies showed that the PNO gel had mechanical strength comparable with that of the P407 hydrogel. The cold PNO solution was conveniently sprayed or smeared on the wound of DFU and rapidly gelled. In vivo studies showed that PNO remarkably accelerated wound healing in rats with DFU. Moreover, the sustained inflammation at the DFU wound was largely reversed by PNO, as reflected by the decreased levels of proinflammatory cytokines (IL-1β, IL-6 and TNF-α) and the increased levels of anti-inflammatory cytokines (IL-10, IL-22 and IL-13). Meanwhile, angiogenesis was significantly promoted by PNO, resulting in rich blood perfusion at the DFU wounds. The therapeutic mechanism of PNO was highly associated with polarizing macrophages and maintaining the homeostasis of the extracellular matrix. Collectively, PNO gel may be a promising vehicle of therapeutic NO gas for DFU treatment.
Collapse
Affiliation(s)
- Yingzheng Zhao
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China.
| | - Lanzi Luo
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Lantian Huang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Yingying Zhang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Mengqi Tong
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Hanxiao Pan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Jianxun Shangguan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Qing Yao
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China
| | - Shihao Xu
- Department of Ultrasonography, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province 325000, China.
| | - Helin Xu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China; CiXi Biomedical Research Institute of Wenzhou Medical University, China.
| |
Collapse
|
22
|
Nitric oxide releasing alginate microspheres for antimicrobial application. Int J Biol Macromol 2022; 224:1244-1251. [DOI: 10.1016/j.ijbiomac.2022.10.210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 10/21/2022] [Accepted: 10/22/2022] [Indexed: 11/05/2022]
|
23
|
White CJ, Lehnert N, Meyerhoff ME. Electrochemical Generation of Nitric Oxide for Medical Applications. ELECTROCHEMICAL SCIENCE ADVANCES 2022; 2:e2100156. [PMID: 36386004 PMCID: PMC9642980 DOI: 10.1002/elsa.202100156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/06/2021] [Indexed: 12/31/2022] Open
Abstract
Over the past 30 years, the significance of nitric oxide (NO) has become increasingly apparent in mammalian physiology. It is biosynthesized by three isoforms of nitric oxide synthases (NOS): neuronal (nNOS), endothelial (eNOS), and inducible (iNOS). Neuronal and eNOS both produce low levels of NO (nM) as a signaling agent and vasodilator, respectively. Inducible (iNOS) is present in activated macrophages at sites of infection to generate acutely toxic (μM) levels of NO as part of the mammalian immune defense mechanism. These discoveries have led to numerous animal and clinical studies to evaluate the potential therapeutic utility of NO in various medical operations/treatments, primarily using NO gas (via gas-cylinders) as the NO source. In this review, we focus specifically on recent advances in the electrochemical generation of NO (E-NOgen) as an alternative means to generate NO from cheap and inert sources, and the fabrication and testing of biomedical devices that utilize E-NOgen to controllably generate NO for medical applications.
Collapse
Affiliation(s)
- Corey J White
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI 48109-1055, USA
| | - Nicolai Lehnert
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI 48109-1055, USA
| | - Mark E Meyerhoff
- Department of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, MI 48109-1055, USA
| |
Collapse
|
24
|
Cao Y, Yin J, Shi Y, Cheng J, Fang Y, Huang C, Yu W, Liu M, Yang Z, Zhou H, Liu H, Wang J, Zhao G. Starch and chitosan-based antibacterial dressing for infected wound treatment via self-activated NO release strategy. Int J Biol Macromol 2022; 220:1177-1187. [PMID: 36030977 DOI: 10.1016/j.ijbiomac.2022.08.152] [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: 04/23/2022] [Revised: 08/10/2022] [Accepted: 08/23/2022] [Indexed: 11/16/2022]
Abstract
In this work, a positively charged chitosan-grafted-polyarginine (CS-N-PArg) as the macro-molecular NO donor, and a negatively charged acetalated starch (AcSt-O-PAsp) as a glucose donor, have been synthesized. To achieve the multi-enzymatic cascade system for local generation of self-supply glucose to increase the H2O2 concentration for the subsequent oxidization of L-Arg into NO, the designed positively charged CS-N-PArg, negatively charged AcSt-O-PAsp, glucoamylase (GA) and glucose oxidase (GOx) are absorbed and assembled in the pore of the gelatin sponge via electrostatic interaction to establish a smart antibacterial dressings (CS/St + GOx/GA). Once stimulated by Escherichia coli (E. coli)-infected wounds (a slightly acidic environment), the cascade reaction system can sequentially induce to generate glucose, H2O2 and NO, which exhibits a meaningful alternative idea for a high-performance antibacterial therapy.
Collapse
Affiliation(s)
- Yufei Cao
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Juanjuan Yin
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Yuting Shi
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Ju Cheng
- School of Basic Medical Science, Lanzhou University, Lanzhou 730000, PR China
| | - Yu Fang
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Congshu Huang
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Wenwen Yu
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China
| | - Mingsheng Liu
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Zheng Yang
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China
| | - Haicun Zhou
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China
| | - Hongbin Liu
- The Second Clinical Medical College, Lanzhou University, Lanzhou 730000, PR China
| | - Jianrong Wang
- Department of Oral Health, Gansu Provincial Maternity and Child-care Hospital, Lanzhou 730050, PR China.
| | - Guanghui Zhao
- State Key Laboratory of Applied Organic Chemistry, Institute of Biochemical Engineering & Environmental Technology, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
25
|
Chen Z, Zheng S, Shen Z, Cheng J, Xiao S, Zhang G, Liu S, Hu J. Oxygen-Tolerant Photoredox Catalysis Triggers Nitric Oxide Release for Antibacterial Applications. Angew Chem Int Ed Engl 2022; 61:e202204526. [PMID: 35579256 DOI: 10.1002/anie.202204526] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Indexed: 12/30/2022]
Abstract
Photoredox catalysis has emerged as a robust tool for chemical synthesis. However, it remains challenging to implement photoredox catalysis under physiological conditions due to the complex microenvironment and the quenching of photocatalyst by biologically relevant molecules such as oxygen. Here, we report that UV-absorbing N,N'-dinitroso-1,4-phenylenediamine derivatives can be selectively activated by fac-Ir(ppy)3 photocatalyst within micellar nanoparticles under visible light irradiation (e.g., 500 nm) through photoredox catalysis in aerated aqueous solutions to form quinonediimine (QDI) residues with concomitant release of NO. Notably, the formation of QDI derivatives can actively scavenge the reactive oxygen species generated by fac-Ir(ppy)3 , thus avoiding oxygen quenching of the photocatalyst. Further, we exemplify that the oxygen-tolerant photoredox catalysis-mediated NO release can not only kill planktonic bacteria in vitro but also efficiently treat MRSA infections in vivo.
Collapse
Affiliation(s)
- Zhenhua Chen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Shaoqiu Zheng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Zhiqiang Shen
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jian Cheng
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Shiyan Xiao
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Guoying Zhang
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Shiyong Liu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Jinming Hu
- Department of Pharmacy, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, and CAS Key Laboratory of Soft Matter Chemistry, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, China
| |
Collapse
|
26
|
Kumar R, Chug MK, Brisbois EJ. Long-Term Storage Stability and Nitric Oxide Release Behavior of ( N-Acetyl- S-nitrosopenicillaminyl)- S-nitrosopenicillamine-Incorporated Silicone Rubber Coatings. ACS APPLIED MATERIALS & INTERFACES 2022; 14:30595-30606. [PMID: 35759508 PMCID: PMC9708111 DOI: 10.1021/acsami.2c06712] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Physical incorporation of nitric oxide (NO) releasing materials in biomedical grade polymer matrices to fabricate antimicrobial coatings and devices is an economically viable process. However, achieving long-term NO release with a minimum or no leaching of the NO donor from the polymer matrix is still a challenging task. Herein, (N-acetyl-S-nitrosopenicillaminyl)-S-nitrosopenicillamine (SNAP-SNAP), a penicillamine dipeptide NO-releasing molecule, is incorporated into a commercially available biomedical grade silicone rubber (SR) to fabricate a NO-releasing coating (SNAP-SNAP/SR). The storage stabilities of the SNAP-SNAP powder and SNAP-SNAP/SR coating were analyzed at different temperatures. The SNAP-SNAP/SR coatings with varying wt % of SNAP-SNAP showed a tunable and sustained NO release for up to 6 weeks. Further, S-nitroso-N-acetylpenicillamine (SNAP), a well-explored NO-releasing molecule, was incorporated into a biomedical grade silicone polymer to fabricate a NO-releasing coating (SNAP/SR) and a comparative analysis of the NO release and S-nitrosothiol (RSNO) leaching behavior of 10 wt % SNAP-SNAP/SR and 10 wt % SNAP/SR was studied. Interestingly, the 10 wt % SNAP-SNAP/SR coatings exhibited ∼36% higher NO release and 4 times less leaching of NO donors than the 10 wt % SNAP/SR coatings. Further, the 10 wt % SNAP-SNAP/SR coatings exhibited promising antibacterial properties against Staphylococcus aureus and Escherichia coli due to the persistent release of NO. The 10 wt % SNAP-SNAP/SR coatings were also found to be biocompatible against NIH 3T3 mouse fibroblast cells. These results corroborate the sustained stability and NO-releasing properties of the SNAP-SNAP in a silicone polymer matrix and demonstrate the potential for the SNAP-SNAP/SR polymer in the fabrication of long-term indwelling biomedical devices and implants to enhance biocompatibility and resist device-related infections.
Collapse
Affiliation(s)
- Rajnish Kumar
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Manjyot Kaur Chug
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| | - Elizabeth J Brisbois
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens, Georgia 30602, United States
| |
Collapse
|
27
|
Yi B, Zhou B, Song Z, Yu L, Wang W, Liu W. Step-wise CAG@PLys@PDA-Cu2+ modification on micropatterned nanofibers for programmed endothelial healing. Bioact Mater 2022; 25:657-676. [PMID: 37056258 PMCID: PMC10086768 DOI: 10.1016/j.bioactmat.2022.07.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 06/23/2022] [Accepted: 07/08/2022] [Indexed: 11/26/2022] Open
Abstract
Native-like endothelium regeneration is a prerequisite for material-guided small-diameter vascular regeneration. In this study, a novel strategy is proposed to achieve phase-adjusted endothelial healing by step-wise modification of parallel-microgroove-patterned (i.e., micropatterned) nanofibers with polydopamine-copper ion (PDA-Cu2+) complexes, polylysine (PLys) molecules, and Cys-Ala-Gly (CAG) peptides (CAG@PLys@PDA-Cu2+). Using electrospun poly(l-lactide-co-caprolactone) random nanofibers as the demonstrating biomaterial, step-wise modification of CAG@PLys@PDA-Cu2+ significantly enhanced substrate wettability and protein adsorption, exhibited an excellent antithrombotic surface and outstanding phase-adjusted capacity of endothelium regeneration involving cell adhesion, endothelial monolayer formation, and the regenerated endothelium maturation. Upon in vivo implantation for segmental replacement of rabbit carotid arteries, CAG@PLys@PDA-Cu2+ modified grafts (2 mm inner diameter) with micropatterns on inner surface effectively accelerated native-like endothelium regeneration within 1 week, with less platelet aggregates and inflammatory response compared to those on non-modified grafts. Prolonged observations at 6- and 12-weeks post-implantation demonstrated a positive vascular remodeling with almost fully covered endothelium and mature smooth muscle layer in the modified vascular grafts, accompanied with well-organized extracellular matrix. By contrast, non-modified vascular grafts induced a disorganized tissue formation with a high risk of thrombogenesis. In summary, step-wise modification of CAG@PLys@PDA-Cu2+ on micropatterned nanofibers can significantly promote endothelial healing without inflicting thrombosis, thus confirming a novel strategy for developing functional vascular grafts or other blood-contacting materials/devices.
Collapse
|
28
|
Chen Z, Zheng S, Shen Z, Cheng J, Xiao S, Zhang G, Liu S, Hu J. Oxygen‐Tolerant Photoredox Catalysis Triggers Nitric Oxide Release for Antibacterial Applications. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202204526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Zhenhua Chen
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| | - Shaoqiu Zheng
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| | - Zhiqiang Shen
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| | - Jian Cheng
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| | - Shiyan Xiao
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| | - Guoying Zhang
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| | - Shiyong Liu
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| | - Jinming Hu
- Department of Pharmacy The First Affiliated Hospital of USTC Division of Life Sciences and Medicine and CAS Key Laboratory of Soft Matter Chemistry Department of Polymer Science and Engineering University of Science and Technology of China Hefei Anhui 230026 China
| |
Collapse
|
29
|
Brady B, Li W, Farooque N, Ehrhardt C, Meyerhoff ME, Wang X. S-Nitrosothiol-Impregnated Silicone Catheter for Colorimetric Sensing of Indole and E. coli: Toward On-Body Detection of Urinary Tract Infections. ACS Sens 2022; 7:1712-1719. [PMID: 35604028 DOI: 10.1021/acssensors.2c00439] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Although there are many techniques to detect pathogenic bacteria, most of them are only suited for in vitro diagnostics. We report a urinary catheter-based colorimetric sensor for potential on-body detection of E. coli, the most prevalent bacterial species in urinary tract infections associated with the use of urinary catheters. In urine, indole is secreted by E. coli and reacts with a nitrosating agent incorporated in a silicone catheter. A red dimeric product, indoxyl red, is generated within silicone rubber to allow for color-based indole sensing with high sensitivity, linearity, and specificity. This reaction is initiated by the nitrosation reaction of indole at its C-3 position via reagents like sodium nitrite or S-nitroso-N-acetyl-penicillamine under aerobic conditions. The generated 3-nitrosoindole undergoes tautomerization, dimerization, and deoximation to form indoxyl red with high absorbance at 537 nm. In contrast to other indole sensors, the presented method can be applied in real catheters to detect indole and E. coli in biofluids such as urine. The is because (1) S-nitroso-N-acetyl-penicillamine, the nitrosating agent, can be impregnated into silicone elastomers, (2) indole from urine is extracted into silicone due to its hydrophobicity, and (3) the high acidity and oxygen solubility of silicone facilitates the sensing reaction within the silicone matrix. This silicone-based colorimetric sensor clearly differentiates E. coli below and above 105 CFU/mL, which is the threshold concentration of bacteriuria. We expect that early diagnosis of urinary tract infections using the naked eye is possible by functionalizing an exposed section of urinary catheters with the proposed molecular probe.
Collapse
Affiliation(s)
- Brock Brady
- Department of Chemistry, Virginia Commonwealth University, 1001 West Main Street, Richmond, Virginia 23284, United States
| | - Wuwei Li
- Department of Chemistry, Virginia Commonwealth University, 1001 West Main Street, Richmond, Virginia 23284, United States
| | - Nashwan Farooque
- Department of Chemistry, Virginia Commonwealth University, 1001 West Main Street, Richmond, Virginia 23284, United States
| | - Christopher Ehrhardt
- Department of Forensic Science, Virginia Commonwealth University, 1015 Floyd Avenue, Richmond, Virginia 23284, United States
| | - Mark E. Meyerhoff
- Department of Chemistry, University of Michigan, 930 North University, Ann Arbor, Michigan 48109, United States
| | - Xuewei Wang
- Department of Chemistry, Virginia Commonwealth University, 1001 West Main Street, Richmond, Virginia 23284, United States
| |
Collapse
|
30
|
Ahmed R, Augustine R, Chaudhry M, Akhtar UA, Zahid AA, Tariq M, Falahati M, Ahmad IS, Hasan A. Nitric oxide-releasing biomaterials for promoting wound healing in impaired diabetic wounds: State of the art and recent trends. Pharmacotherapy 2022; 149:112707. [PMID: 35303565 DOI: 10.1016/j.biopha.2022.112707] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/31/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022]
Abstract
Impaired diabetic wounds are serious pathophysiological complications associated with persistent microbial infections including failure in the closure of wounds, and the cause of a high frequency of lower limb amputations. The healing of diabetic wounds is attenuated due to the lack of secretion of growth factors, prolonged inflammation, and/or inhibition of angiogenic activity. Diabetic wound healing can be enhanced by supplying nitric oxide (NO) endogenously or exogenously. NO produced inside the cells by endothelial nitric oxide synthase (eNOS) naturally aids wound healing through its beneficial vasculogenic effects. However, during hyperglycemia, the activity of eNOS is affected, and thus there becomes an utmost need for the topical supply of NO from exogenous sources. Thus, NO-donors that can release NO are loaded into wound healing patches or wound coverage matrices to treat diabetic wounds. The burst release of NO from its donors is prevented by encapsulating them in polymeric hydrogels or nanoparticles for supplying NO for an extended duration of time to the diabetic wounds. In this article, we review the etiology of diabetic wounds, wound healing strategies, and the role of NO in the wound healing process. We further discuss the challenges faced in translating NO-donors as a clinically viable nanomedicine strategy for the treatment of diabetic wounds with a focus on the use of biomaterials for the encapsulation and in vivo controlled delivery of NO-donors.
Collapse
Affiliation(s)
- Rashid Ahmed
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar; Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, Mirpur 10250, AJK, Pakistan; Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA
| | - Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar
| | - Maryam Chaudhry
- Department of Continuing Education, University of Oxford, OX1 2JD Oxford, United Kingdom
| | - Usman A Akhtar
- Department of Chemical Engineering, College of Engineering, Qatar University, Doha 2713, Qatar
| | - Alap Ali Zahid
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar
| | - Muhammad Tariq
- Department of Biotechnology, Faculty of Natural and Applied Sciences, Mirpur University of Science and Technology, Mirpur 10250, AJK, Pakistan
| | - Mojtaba Falahati
- Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, 3015GE Rotterdam, The Netherlands
| | - Irfan S Ahmad
- Nick Holonyak Jr. Micro and Nanotechnology Laboratory, University of Illinois at Urbana Champaign, IL, USA; Department of Agricultural and Biological Engineering, University of Illinois at Urbana Champaign, IL, USA; Carle Illinois College of Medicine, University of Illinois at Urbana Champaign, IL, USA
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| |
Collapse
|
31
|
Li M, Qiu W, Wang Q, Li N, Liu L, Wang X, Yu J, Li X, Li F, Wu D. Nitric Oxide-Releasing Tryptophan-Based Poly(ester urea)s Electrospun Composite Nanofiber Mats with Antibacterial and Antibiofilm Activities for Infected Wound Healing. ACS APPLIED MATERIALS & INTERFACES 2022; 14:15911-15926. [PMID: 35373564 DOI: 10.1021/acsami.1c24131] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Bacterial biofilms on wounds can lead to ongoing inflammation and delayed reepithelialization, which brings a heavy burden to the medical systems. Nitric oxide based treatment has attracted attention because it is a promising strategy to eliminate biofilms and heal infected wounds. Herein, a series of tryptophan-based poly(ester urea)s with good biodegradation and biocompatibility were developed for the preparation of composite mats by electrospinning. Furthermore, the mats were grafted with a nitric oxide donor (nitrosoglutathione, GSNO) to provide one type of NO loading cargo. The mats were found to have a prolonged NO release profile for 408 h with a maximum release of 1.0 μmol/L, which had a significant effect on killing bacteria and destructing biofilms. The designed mats were demonstrated to promote the growth of cells, regulate inflammatory factors, and significantly improve collagen deposition in the wound, eventually accelerating wound-size reduction. Thus, the studies presented herein provide insights into the production of NO-releasing wound dressings and support the application of full-thickness wound healing.
Collapse
Affiliation(s)
- Mengna Li
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Weiwang Qiu
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Qian Wang
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Na Li
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Lu Liu
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
| | - Xueli Wang
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 201620, China
| | - Jianyong Yu
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 201620, China
| | - Xiaoran Li
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 201620, China
| | - Faxue Li
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 201620, China
| | - Dequn Wu
- Key Laboratory of Textile Science and Technology, Ministry of Education, College of Textiles, Donghua University, Shanghai 201620, China
- Innovation Center for Textile Science and Technology, Donghua University, Shanghai 201620, China
| |
Collapse
|
32
|
Beurton J, Boudier A, Barozzi Seabra A, Vrana NE, Clarot I, Lavalle P. Nitric Oxide Delivering Surfaces: An Overview of Functionalization Strategies and Efficiency Progress. Adv Healthc Mater 2022; 11:e2102692. [PMID: 35358359 DOI: 10.1002/adhm.202102692] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 02/27/2022] [Indexed: 12/15/2022]
Abstract
An overview on the design of nitric oxide (NO) delivering surfaces for biomedical purposes is provided, with a focus on the advances of the past 5 years. A localized supply of NO is of a particular interest due to the pleiotropic biological effects of this diatomic compound. Depending on the generated NO flux, the surface can mimic a physiological release profile to provide an activity on the vascular endothelium or an antibacterial activity. Three requirements are considered to describe the various strategies leading to a surface delivering NO. Firstly, the coating must be selected in accordance with the properties of the substrate (nature, shape, dimensions…). Secondly, the releasing and/or generating kinetics of NO should match the targeted biological application. Currently, the most promising structures are developed to provide an adaptable NO supply driven by pathophysiological needs. Finally, the biocompatibility and the stability of the surface must also be considered regarding the expected residence time of the device. A critical point of view is proposed to help readers in the design of the NO delivering surface according to its expected requirement and therapeutic purpose.
Collapse
Affiliation(s)
- Jordan Beurton
- Université de Lorraine CITHEFOR Nancy F‐54000 France
- Institut National de la Santé et de la Recherche Médicale Inserm UMR_S 1121 Biomaterials and Bioengineering Strasbourg F‐67085 France
- Université de Strasbourg Faculté de Chirurgie Dentaire de Strasbourg Strasbourg F‐67000 France
| | | | - Amedea Barozzi Seabra
- Center for Natural and Human Sciences (CCNH) Federal University of ABC (UFABC) Santo André SP CEP 09210‐580 Brazil
| | | | - Igor Clarot
- Université de Lorraine CITHEFOR Nancy F‐54000 France
| | - Philippe Lavalle
- Université de Strasbourg Faculté de Chirurgie Dentaire de Strasbourg Strasbourg F‐67000 France
- Center for Natural and Human Sciences (CCNH) Federal University of ABC (UFABC) Santo André SP CEP 09210‐580 Brazil
- SPARTHA Medical 14B Rue de la Canardiere Strasbourg 67100 France
| |
Collapse
|
33
|
Chen Y, Huang Y, Jin Q. Polymeric nanoplatforms for the delivery of antibacterial agents. MACROMOL CHEM PHYS 2022. [DOI: 10.1002/macp.202100440] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Yongcheng Chen
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education Department of Polymer Science and Engineering Zhejiang University Hangzhou Zhejiang 310027 PR China
| | - Yue Huang
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education Department of Polymer Science and Engineering Zhejiang University Hangzhou Zhejiang 310027 PR China
| | - Qiao Jin
- MOE Key Laboratory of Macromolecule Synthesis and Functionalization of Ministry of Education Department of Polymer Science and Engineering Zhejiang University Hangzhou Zhejiang 310027 PR China
| |
Collapse
|
34
|
Li W, Wang D, Lao KU, Wang X. Buffer concentration dramatically affects the stability of S-nitrosothiols in aqueous solutions. Nitric Oxide 2022; 118:59-65. [PMID: 34848361 DOI: 10.1016/j.niox.2021.11.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/19/2021] [Accepted: 11/25/2021] [Indexed: 01/17/2023]
Abstract
S-nitrosothiols (RSNOs) are an important group of nitric oxide (NO)-donating compounds with low toxicity and wide biomedical applications. In this paper, we, for the first time, demonstrate that the concentration of buffer remarkably affects the stability of RSNOs including naturally occurring S-nitrosoglutathione (GSNO) and synthetic S-nitroso-N-acetylpenicillamine (SNAP). For a solution with a high concentration of GSNO (e.g., 50 mM) and an initial near-neutral pH, the optimal buffer concentration is close to the GSNO concentration under our experimental conditions. A lower buffer concentration does not have adequate buffer capacity to resist the pH drop caused by GSNO decomposition. The decreased solution pH further accelerates GSNO decomposition because GSNO is most stable at near-neutral pH according to our density functional theory (DFT) calculations. A higher-than-optimal buffer concentration also reduces the GSNO stability because buffer ingredients including phosphate, Tris base, and HEPES consume NO/N2O3. In contrast to GSNO, the highest SNAP stability is obtained when the starting solution at a neutral pH does not contain buffer species, and the stability decreases as the buffer concentration increases. This is because SNAP is more stable at mildly acidic pH and the SNAP decomposition-induced pH drop stabilizes the donor. When the RSNO concentration is low (e.g., 1 mM), the buffer concentration also matters because any excess buffer accelerates the donor decomposition. Since the effect of buffer concentration was previously overlooked and suboptimal buffer concentrations were often used, this paper will aid in the formulation of RSNO solutions to obtain the maximum stability for prolonged storage and sustained NO release.
Collapse
Affiliation(s)
- Wuwei Li
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St., Richmond, VA, 23284, USA
| | - Danyang Wang
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St., Richmond, VA, 23284, USA
| | - Ka Un Lao
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St., Richmond, VA, 23284, USA
| | - Xuewei Wang
- Department of Chemistry, Virginia Commonwealth University, 1001 W Main St., Richmond, VA, 23284, USA.
| |
Collapse
|
35
|
Ren X, Wang Y, Jia L, Guo X, He X, Zhao Z, Gao D, Yang Z. Intelligent Nanomedicine Approaches Using Medical Gas-Mediated Multi-Therapeutic Modalities Against Cancer. J Biomed Nanotechnol 2022; 18:24-49. [PMID: 35180898 DOI: 10.1166/jbn.2022.3224] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The emerging area of gas-mediated cancer treatment has received widespread attention in the medical community. Featuring unique physical, chemical, and biological properties, nanomaterials can facilitate the delivery and controllable release of medicinal gases at tumor sites, and also serve as ideal platforms for the integration of other therapeutic modalities with gas therapy to augment cancer therapeutic efficacy. This review presents an overview of anti-cancer mechanisms of several therapeutic gases: nitric oxide (NO), hydrogen sulfide (H₂S), carbon monoxide (CO), oxygen (O₂), and hydrogen (H₂). Controlled release behaviors of gases under different endogenous and exogenous stimuli are also briefly discussed, followed by their synergistic effects with different therapeutic modes. Moreover, the potential challenges and future prospects regarding gas therapy based on nanomaterials are also described, aiming to facilitate the advancement of gas therapeutic nanomedicine in new frontiers for highly efficient cancer treatment.
Collapse
Affiliation(s)
- Xuechun Ren
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Ying Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Liangliang Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiaoqing Guo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xinyu He
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhipeng Zhao
- School of Physical Education, Xizang Minzu University, Xianyang, 712000, Shaanxi, China
| | - Di Gao
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zhe Yang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| |
Collapse
|
36
|
Hu J, Fang Y, Huang X, Qiao R, Quinn JF, Davis TP. Engineering macromolecular nanocarriers for local delivery of gaseous signaling molecules. Adv Drug Deliv Rev 2021; 179:114005. [PMID: 34687822 DOI: 10.1016/j.addr.2021.114005] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/30/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
In addition to being notorious air pollutants, nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S) have also been known as endogenous gaseous signaling molecules (GSMs). These GSMs play critical roles in maintaining the homeostasis of living organisms. Importantly, the occurrence and development of many diseases such as inflammation and cancer are highly associated with the concentration changes of GSMs. As such, GSMs could also be used as new therapeutic agents, showing great potential in the treatment of many formidable diseases. Although clinically it is possible to directly inhale GSMs, the precise control of the dose and concentration for local delivery of GSMs remains a substantial challenge. The development of gaseous signaling molecule-releasing molecules provides a great tool for the safe and convenient delivery of GSMs. In this review article, we primarily focus on the recent development of macromolecular nanocarriers for the local delivery of various GSMs. Learning from the chemistry of small molecule-based donors, the integration of these gaseous signaling molecule-releasing molecules into polymeric matrices through physical encapsulation, post-modification, or direct polymerization approach renders it possible to fabricate numerous macromolecular nanocarriers with optimized pharmacokinetics and pharmacodynamics, revealing improved therapeutic performance than the small molecule analogs. The development of GSMs represents a new means for many disease treatments with unique therapeutic outcomes.
Collapse
|
37
|
Mazrad ZAI, Schelle B, Nicolazzo JA, Leiske MN, Kempe K. Nitrile-Functionalized Poly(2-oxazoline)s as a Versatile Platform for the Development of Polymer Therapeutics. Biomacromolecules 2021; 22:4618-4632. [PMID: 34647734 DOI: 10.1021/acs.biomac.1c00923] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In recent years, polymers bearing reactive groups have received significant interest for biomedical applications. Numerous functional polymer platforms have been introduced, which allow for the preparation of materials with tailored properties via post-polymerization modifications. However, because of their reactivity, many functional groups are not compatible with the initial polymerization. The nitrile group is a highly interesting and relatively inert functionality that has mainly received attention in radical polymerizations. In this Article, a nitrile-functionalized 2-oxazoline monomer (2-(4-nitrile-butyl)-2-oxazoline, BuNiOx) is introduced, and its compatibility with the cationic ring-opening polymerization is demonstrated. Subsequently, the versatility of nitrile-functionalized poly(2-oxazoline)s (POx) is presented. To this end, diverse (co)polymers are synthesized and characterized by nuclear resonance spectroscopy, size-exclusion chromatography, and mass spectrometry. Amphiphilic block copolymers are shown to efficiently encapsulate the hydrophobic drug curcumin (CUR) in aqueous solution, and the anti-inflammatory effect of the CUR-containing nanostructures is presented in BV-2 microglia. Furthermore, the availability of the BuNiOx repeating units for post-polymerization modifications with hydroxylamine to yield amidoxime (AO)-functionalized POx is demonstrated. These AO-containing POx were successfully applied for the complexation of Fe(III) in a quantitative manner. In addition, AO-functionalized POx were shown to release nitric oxide intracellularly in BV-2 microglia. Thus nitrile-functionalized POx represent a promising and robust platform for the design of polymer therapeutics for a wide range of applications.
Collapse
Affiliation(s)
- Zihnil A I Mazrad
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Baptiste Schelle
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Joseph A Nicolazzo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Meike N Leiske
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia
| | - Kristian Kempe
- ARC Centre of Excellence in Convergent Bio-Nano Science & Technology, Monash University, Parkville, Victoria 3052, Australia.,Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria 3052, Australia.,Materials Science and Engineering, Monash University, Clayton, Victoria 3800, Australia
| |
Collapse
|
38
|
Tang Y, Wang T, Feng J, Rong F, Wang K, Li P, Huang W. Photoactivatable Nitric Oxide-Releasing Gold Nanocages for Enhanced Hyperthermia Treatment of Biofilm-Associated Infections. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50668-50681. [PMID: 34669372 DOI: 10.1021/acsami.1c12483] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
With the increasing clinical use of invasive medical devices, various healthcare-associated infections (HAIs) caused by bacterial biofilm colonization of biomedical devices have posed serious threats to patients. The formation of biofilms makes it much more difficult and costly to treat infections. Here, we report a nitric oxide (NO)-releasing gold nanocage (AuNC@NO) that is stimulated by near-infrared (NIR) irradiation to deliver NO and generate hyperthermia for biofilm elimination. AuNC@NO was prepared by immobilizing a temperature-responsive NO donor onto gold nanocages (AuNCs) through thiol-gold interactions. AuNC@NO possesses stable and excellent photothermal conversion efficiency, as well as the characteristics of slow NO release at physiological temperature and on-demand quick NO release under NIR irradiation. Based on these features, AuNC@NO exhibits enhanced in vitro bactericidal and antibiofilm efficacy compared with AuNCs, which could achieve 4 orders of magnitude bacterial reduction and 85.4% biofilm elimination under NIR irradiation. In addition, we constructed an implant biofilm infection model and a subcutaneous biofilm infection model to evaluate the anti-infective effect of AuNC@NO. The in vivo results indicated that after 5 min of 0.5 W cm-2 NIR irradiation, NO release from AuNC@NO was significantly accelerated, which induced the dispersal of methicillin-resistant Staphylococcus aureus (MRSA) biofilms and synergized with photothermal therapy (PTT) to kill planktonic MRSA that had lost its biofilm protection. Meanwhile, the surrounding tissues showed little damage because of controlled photothermal temperature and toxicity. In view of the above-mentioned results, the AuNC@NO nanocomposite developed in this work reveals potential application prospects as a useful antibiofilm agent in the field of biofilm-associated infection treatment.
Collapse
Affiliation(s)
- Yizhang Tang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Tengjiao Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Jiahao Feng
- Queen Mary University of London Engineering School, Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Fan Rong
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Kun Wang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Peng Li
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
| | - Wei Huang
- Frontiers Science Center for Flexible Electronics (FSCFE), Xi'an Institute of Flexible Electronics (IFE) & Xi'an Institute of Biomedical Materials and Engineering (IBME), Northwestern Polytechnical University (NPU), 127 West Youyi Road, Xi'an, Shaanxi 710072, P. R. China
- State Key Laboratory for Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, P. R. China
- Key Laboratory of Flexible Electronics (KLOFE), Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, P. R. China
| |
Collapse
|
39
|
Li P, Jin D, Dou J, Wang L, Wang Y, Jin X, Han X, Kang IK, Yuan J, Shen J, Yin M. Nitric oxide-releasing poly(ε-caprolactone)/S-nitrosylated keratin biocomposite scaffolds for potential small-diameter vascular grafts. Int J Biol Macromol 2021; 189:516-527. [PMID: 34450147 DOI: 10.1016/j.ijbiomac.2021.08.147] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/16/2021] [Accepted: 08/18/2021] [Indexed: 02/08/2023]
Abstract
Rapid endothelialization and regulation of smooth muscle cell proliferation are crucial for small-diameter vascular grafts to address poor compliance, thromboembolism, and intimal hyperplasia, and achieve revascularization. As a gaseous signaling molecule, nitric oxide (NO) regulates cardiovascular homeostasis, inhibits blood clotting and intimal hyperplasia, and promotes the growth of endothelial cells. Due to the instability and burst release of small molecular NO donors, a novel biomacromolecular donor has generated increasing interest. In the study, a low toxic NO donor of S-nitrosated keratin (KSNO) was first synthesized and then coelectrospun with poly(ε-caprolactone) to afford NO-releasing small-diameter vascular graft. PCL/KSNO graft was capable to generate NO under the catalysis of ascorbic acid (Asc), so the graft selectively elevated adhesion and growth of human umbilical vein endothelial cells (HUVECs), while inhibited the proliferation of human aortic smooth muscle cells (HASMCs) in the presence of Asc. In addition, the graft displayed significant antibacterial properties and good blood compatibility. Animal experiments showed that the biocomposite graft could inhibit thrombus formation and preserve normal blood flow via single rabbit carotid artery replacement for 1 month. More importantly, a complete endothelium was observed on the lumen surface. Taken together, PCL/KSNO small-diameter vascular graft has potential applications in vascular tissue engineering with rapid endothelialization and vascular remolding.
Collapse
Affiliation(s)
- Pengfei Li
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Dawei Jin
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, PR China
| | - Jie Dou
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Lijuan Wang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Yanfang Wang
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Xingxing Jin
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Xiao Han
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China
| | - Inn-Kyu Kang
- Department of Polymer Science and Engineering, Kyungpook National University, Daegu 702-701, South Korea
| | - Jiang Yuan
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China.
| | - Jian Shen
- Jiangsu Collaborative Innovation Center of Biomedical Functional Materials, Jiangsu Key Laboratory of Bio-functional Materials, Department of Materials Science and Engineering, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, PR China.
| | - Meng Yin
- Department of Cardiothoracic Surgery, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, 1678 Dong Fang Road, Shanghai 200127, PR China.
| |
Collapse
|
40
|
Li W, Yang Y, Ehrhardt CJ, Lewinski N, Gascoyne D, Lucas G, Zhao H, Wang X. 3D Printing of Antibacterial Polymer Devices Based on Nitric Oxide Release from Embedded S-Nitrosothiol Crystals. ACS APPLIED BIO MATERIALS 2021; 4:7653-7662. [PMID: 35006705 DOI: 10.1021/acsabm.1c00887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Controlled release of drugs from medical implants is an effective approach to reducing foreign body reactions and infections. We report here on a one-step 3D printing strategy to create drug-eluting polymer devices with a drug-loaded bulk and a drug-free coating. The spontaneously formed drug-free coating dramatically reduces the surface roughness of the implantable devices and serves as a protective layer to suppress the burst release of drugs. A high viscosity liquid silicone that can be extruded based on its shear-thinning property and quickly vulcanize upon exposure to ambient moisture is used as the ink for 3D printing. S-Nitrosothiol type nitric oxide (NO) donors in their crystalline forms are selected as model drugs because of the potent antimicrobial, antithrombotic, and anti-inflammatory properties of NO. Direct ink writing of the homogenized polymer-drug mixtures generates rough and ill-defined device surfaces because of the exposed S-nitrosothiol microparticles. When a low-viscosity silicone (polydimethylsiloxane) is added into the ink, this silicone diffuses outward upon deposition to form a drug-free outermost layer without compromising the integrity of the printed structures. S-Nitrosoglutathione (GSNO) or S-nitroso-N-acetylpenicillamine (SNAP) embedded in the printed silicone matrix releases NO under physiological conditions from days to about one month. The microsized drug crystals are well-preserved in the ink preparation and printing processes, which is one reason for the sustained NO release. Biofilm and cytotoxicity experiments confirmed the antibacterial property and safety of the printed NO-releasing devices. This additive manufacturing platform does not require dissolution of drugs and involves no thermal or UV processes and, therefore, offers unique opportunities to produce drug-eluting silicone devices in a customized manner.
Collapse
Affiliation(s)
- Wuwei Li
- Department of Chemistry, Virginia Commonwealth University, 1001 W. Main Street, Richmond, Virginia 23284, United States
| | - Yuanhang Yang
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, BioTech One, 800 East Leigh Street, Richmond, Virginia 23219, United States
| | - Christopher J Ehrhardt
- Department of Forensic Science, Virginia Commonwealth University, 1015 Floyd Avenue, Richmond, Virginia 23284, United States
| | - Nastassja Lewinski
- Department of Chemical and Life Science Engineering, Virginia Commonwealth University, 601 W. Main Street, Richmond, Virginia 23284, United States
| | - David Gascoyne
- Momentive Performance Materials Inc., 260 Hudson River Road, Waterford, New York 12188, United States
| | - Gary Lucas
- Momentive Performance Materials Inc., 260 Hudson River Road, Waterford, New York 12188, United States
| | - Hong Zhao
- Department of Mechanical and Nuclear Engineering, Virginia Commonwealth University, BioTech One, 800 East Leigh Street, Richmond, Virginia 23219, United States
| | - Xuewei Wang
- Department of Chemistry, Virginia Commonwealth University, 1001 W. Main Street, Richmond, Virginia 23284, United States
| |
Collapse
|
41
|
Wang DC, Clark JR, Lee R, Nelson AH, Maresso AW, Acharya G, Shin CS. Development of Antimicrobial Nitric Oxide-Releasing Fibers. Pharmaceutics 2021; 13:pharmaceutics13091445. [PMID: 34575520 PMCID: PMC8468281 DOI: 10.3390/pharmaceutics13091445] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/05/2021] [Accepted: 09/07/2021] [Indexed: 11/27/2022] Open
Abstract
Nitric oxide (NO) is a highly reactive gas molecule, exhibiting antimicrobial properties. Because of its reactive nature, it is challenging to store and deliver NO efficiently as a therapeutic agent. The objective of this study was to develop NO-releasing polymeric fibers (NO-fibers), as an effective delivery platform for NO. NO-fibers were fabricated with biopolymer solutions of polyvinyl pyrrolidone (PVP) and ethylcellulose (EC), and derivatives of N-diazeniumdiolate (NONOate) as NO donor molecules, using an electrospinning system. We evaluated in vitro NO release kinetics, along with antimicrobial effects and cytotoxicity in microorganisms and human cell culture models. We also studied the long-term stability of NONOates in NO-fibers over 12 months. We demonstrated that the NO-fibers could release NO over 24 h, and showed inhibition of the growth of Pseudomonas aeruginosa (P. aeruginosa) and methicillin-resistant Staphylococcus aureus (MRSA), without causing cytotoxicity in human cells. NO-fibers were able to store NONOates for over 12 months at room temperature. This study presents the development of NO-fibers, and the feasibility of NO-fibers to efficiently store and deliver NO, which can be further developed as a bandage.
Collapse
Affiliation(s)
- Daniel C. Wang
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.W.); (R.L.); (A.H.N.)
| | - Justin R. Clark
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (J.R.C.); (A.W.M.)
| | - Richard Lee
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.W.); (R.L.); (A.H.N.)
| | - Adam H. Nelson
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.W.); (R.L.); (A.H.N.)
| | - Anthony W. Maresso
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA; (J.R.C.); (A.W.M.)
| | - Ghanashyam Acharya
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.W.); (R.L.); (A.H.N.)
- Correspondence: (G.A.); (C.S.S.)
| | - Crystal S. Shin
- Department of Surgery, Baylor College of Medicine, Houston, TX 77030, USA; (D.C.W.); (R.L.); (A.H.N.)
- Correspondence: (G.A.); (C.S.S.)
| |
Collapse
|
42
|
Paul S, Pan S, Mukherjee A, De P. Nitric Oxide Releasing Delivery Platforms: Design, Detection, Biomedical Applications, and Future Possibilities. Mol Pharm 2021; 18:3181-3205. [PMID: 34433264 DOI: 10.1021/acs.molpharmaceut.1c00486] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Gasotransmitters belong to the subfamily of endogenous gaseous signaling molecules, which find a wide range of biomedical applications. Among the various gasotransmitters, nitric oxide (NO) has an enormous effect on the cardiovascular system. Apart from this, NO showed a pivotal role in neurological, respiratory, and immunological systems. Moreover, the paradoxical concentration-dependent activities make this gaseous signaling molecule more interesting. The gaseous NO has negligible stability in physiological conditions (37 °C, pH 7.4), which restricts their potential therapeutic applications. To overcome this issue, various NO delivering carriers were reported so far. Unfortunately, most of these NO donors have low stability, short half-life, or low NO payload. Herein, we review the synthesis of NO delivering motifs, development of macromolecular NO donors, their advantages/disadvantages, and biological applications. Various NO detection analytical techniques are discussed briefly, and finally, a viewpoint about the design of polymeric NO donors with improved physicochemical characteristics is predicted.
Collapse
|
43
|
Yang Y, Zhou Y, Li Y, Guo L, Zhou J, Chen J. Injectable and self-healing hydrogel containing nitric oxide donor for enhanced antibacterial activity. REACT FUNCT POLYM 2021. [DOI: 10.1016/j.reactfunctpolym.2021.105003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
44
|
Shen Z, Zheng S, Xiao S, Shen R, Liu S, Hu J. Red-Light-Mediated Photoredox Catalysis Enables Self-Reporting Nitric Oxide Release for Efficient Antibacterial Treatment. Angew Chem Int Ed Engl 2021; 60:20452-20460. [PMID: 34196472 DOI: 10.1002/anie.202107155] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/22/2021] [Indexed: 12/12/2022]
Abstract
Nitric oxide (NO) serves as a key regulator of many physiological processes and as a potent therapeutic agent. The local delivery of NO is important to achieve target therapeutic outcomes due to the toxicity of NO at high concentrations. Although light stimulus represents a non-invasive tool with spatiotemporal precision to mediate NO release, many photoresponsive NO-releasing molecules can only respond to ultraviolet (UV) or near-UV visible light with low penetration and high phototoxicity. We report that coumarin-based NO donors with maximal absorbances at 328 nm can be activated under (deep) red-light (630 or 700 nm) irradiation in the presence of palladium(II) tetraphenyltetrabenzoporphyrin, enabling stoichiometric and self-reporting NO release with a photolysis quantum yield of 8 % via photoredox catalysis. This NO-releasing platform with ciprofloxacin loading can eradicate Pseudomonas aeruginosa biofilm in vitro and treat cutaneous abscesses in vivo.
Collapse
Affiliation(s)
- Zhiqiang Shen
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Shaoqiu Zheng
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Shiyan Xiao
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Ruan Shen
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Shiyong Liu
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China
| | - Jinming Hu
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, 230026, Anhui, China
| |
Collapse
|
45
|
Shen Z, Zheng S, Xiao S, Shen R, Liu S, Hu J. Red‐Light‐Mediated Photoredox Catalysis Enables Self‐Reporting Nitric Oxide Release for Efficient Antibacterial Treatment. Angew Chem Int Ed Engl 2021. [DOI: 10.1002/ange.202107155] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Affiliation(s)
- Zhiqiang Shen
- CAS Key Laboratory of Soft Matter Chemistry Hefei National Laboratory for Physical Science at the Microscale Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 Anhui China
| | - Shaoqiu Zheng
- CAS Key Laboratory of Soft Matter Chemistry Hefei National Laboratory for Physical Science at the Microscale Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 Anhui China
| | - Shiyan Xiao
- CAS Key Laboratory of Soft Matter Chemistry Hefei National Laboratory for Physical Science at the Microscale Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 Anhui China
| | - Ruan Shen
- CAS Key Laboratory of Soft Matter Chemistry Hefei National Laboratory for Physical Science at the Microscale Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 Anhui China
| | - Shiyong Liu
- CAS Key Laboratory of Soft Matter Chemistry Hefei National Laboratory for Physical Science at the Microscale Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 Anhui China
| | - Jinming Hu
- CAS Key Laboratory of Soft Matter Chemistry Hefei National Laboratory for Physical Science at the Microscale Department of Polymer Science and Engineering University of Science and Technology of China Hefei 230026 Anhui China
| |
Collapse
|
46
|
Wu M, Lu Z, Wu K, Nam C, Zhang L, Guo J. Recent advances in the development of nitric oxide-releasing biomaterials and their application potentials in chronic wound healing. J Mater Chem B 2021; 9:7063-7075. [PMID: 34109343 DOI: 10.1039/d1tb00847a] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Chronic wounds, such as pressure ulcers, vascular ulcers and diabetic foot ulcers (DFUs), often stay in a state of pathological inflammation and suffer from persistent infection, excess inflammation, and hypoxia, thus they are difficult to be healed. Nitric oxide (NO) plays a critical role in the regulation of various wound healing processes, including inflammatory response, cell proliferation, collagen formation, antimicrobial action and angiogenesis. The important role of NO in wound healing attracts intensive research focus on NO-based wound healing therapy. However, the application of NO gas therapy needs to resolve the intrinsic shortcomings of gas therapy, such as short storage and release times as well as temporal and spatial uncontrollability of the release mode. So far, various types of NO donors, including organic nitrates (RONO2), nitrites (RONO), S-nitrosothiols (RSNOs), nitrosamines, N-diazeniumdiolates (NONOates), and metal-NO complexes, have been developed to solidify gaseous NO and they were further encapsulated in or conjugated onto a variety of biomaterial vectors to develop NO delivery systems. NO synthetic enzyme mimics to catalyze the production and release of NO from l-arginine have also been developed. This paper reviews recent advances of NO donors, biomaterial vectors, thus-formed NO delivery systems, as well as recently emerged NO synthetic enzyme mimics. Furthermore, this review also summarizes the functions of NO releasing biomaterials that would benefit chronic wound healing, including antibacterial properties and the promotion of angiogenesis, as well as the convenient combination of light/thermal induced NO release with light/thermal therapies, and the prospects for future developing trends in this area.
Collapse
Affiliation(s)
- Min Wu
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
| | - Zhihui Lu
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
| | - Keke Wu
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
| | - Changwoo Nam
- Department of Organic Materials and Fiber Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si, Jeollabuk-do 54896, Republic of Korea.
| | - Lin Zhang
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
| | - Jinshan Guo
- Department of Histology and Embryology, NMPA Key Laboratory for Safety Evaluation of Cosmetics, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Southern Medical University, Guangzhou, China.
| |
Collapse
|
47
|
Ghalei S, Hopkins S, Douglass M, Garren M, Mondal A, Handa H. Nitric oxide releasing halloysite nanotubes for biomedical applications. J Colloid Interface Sci 2021; 590:277-289. [PMID: 33548611 PMCID: PMC7933102 DOI: 10.1016/j.jcis.2021.01.047] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/28/2020] [Accepted: 01/16/2021] [Indexed: 11/18/2022]
Abstract
Halloysite nanotubes (HNTs) are natural aluminosilicate clay that have been extensivelyexplored fordelivery of bioactive agents in biomedical applications because of their desirable features including unique hollow tubular structure, good biocompatibility, high mechanical strength, and extensive functionality. For the first time, in this work, functionalized HNTs are developed as a delivery platform for nitric oxide (NO), a gaseous molecule, known for its important roles in the regulation of various physiological processes. HNTs were first hydroxylated and modified with an aminosilane crosslinker, (3-aminopropyl) trimethoxysilane (APTMS), to enable the covalent attachment of a NO donor precursor, N-acetyl-d-penicillamine (NAP). HNT-NAP particles were then converted to NO-releasing S-nitroso-N-acetyl-penicillamine HNT-SNAP by nitrosation. The total NO loading on the resulting nanotubes was 0.10 ± 0.07 μmol/mg which could be released using different stimuli such as heat and light. Qualitative (Fourier-transform infrared spectroscopy and Nuclear magnetic resonance) and quantitative (Ninhydrin and Ellman) analyses were performed to confirm successful functionalization of HNTs at each step. Field emission scanning electron microscopy (FE-SEM) showed that the hollow tubular morphology of the HNTs was preserved after modification. HNT-SNAP showed concentration-dependent antibacterial effects against Gram-positive Staphylococcus aureus (S. aureus), resulting in up to 99.6% killing efficiency at a concentration of 10 mg/mL as compared to the control. Moreover, no significant cytotoxicity toward 3T3 mouse fibroblast cells was observed at concentrations equal or below 2 mg/mL of HNT-SNAP according to a WST-8-based cytotoxicity assay. The SNAP-functionalized HNTs represent a novel and efficient NO delivery system that holds the potential to be used, either alone or in combination with polymers for different biomedical applications.
Collapse
Affiliation(s)
- Sama Ghalei
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens 30602, United States
| | - Sean Hopkins
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens 30602, United States
| | - Megan Douglass
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens 30602, United States
| | - Mark Garren
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens 30602, United States
| | - Arnab Mondal
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens 30602, United States
| | - Hitesh Handa
- School of Chemical, Materials and Biomedical Engineering, University of Georgia, Athens 30602, United States.
| |
Collapse
|
48
|
Duan Y, He K, Zhang G, Hu J. Photoresponsive Micelles Enabling Codelivery of Nitric Oxide and Formaldehyde for Combinatorial Antibacterial Applications. Biomacromolecules 2021; 22:2160-2170. [PMID: 33884862 DOI: 10.1021/acs.biomac.1c00251] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
It is of particular interest to develop new antibacterial agents with low risk of drug resistance development and low toxicity toward mammalian cells to combat pathogen infections. Although gaseous signaling molecules (GSMs) such as nitric oxide (NO) and formaldehyde (FA) have broad-spectrum antibacterial performance and the low propensity of drug resistance development, many previous studies heavily focused on nanocarriers capable of delivering only one GSM. Herein, we developed a micellar nanoparticle platform that can simultaneously deliver NO and FA under visible light irradiation. An amphiphilic diblock copolymer of poly(ethylene oxide)-b-poly(4-((2-nitro-5-(((2-nitrobenzyl)oxy)methoxy)benzyl)(nitroso)amino)benzyl methacrylate) (PEO-b-PNNBM) was successfully synthesized through atom transfer radical polymerization (ATRP). The resulting diblock copolymer self-assembled into micellar nanoparticles without premature NO and FA leakage, whereas they underwent phototriggered disassembly with the corelease of NO and FA. We showed that the NO- and FA-releasing micellar nanoparticles exhibited a combinatorial antibacterial performance, efficiently killing both Gram-negative (e.g., Escherichia coli) and Gram-positive (e.g., Staphylococcus aureus) bacteria with low toxicity to mammalian cells and low hemolytic property. This work provides new insights into the development of GSM-based antibacterial agents.
Collapse
Affiliation(s)
- Yutian Duan
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Kewu He
- Imaging Center of the Third Affiliated Hospital of Anhui Medical University, 390 Huaihe Road, Hefei, 230031 Anhui, China
| | - Guoying Zhang
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| | - Jinming Hu
- CAS Key Laboratory of Soft Matter Chemistry, Hefei National Laboratory for Physical Science at the Microscale, Department of Polymer Science and Engineering, University of Science and Technology of China, Hefei, Anhui 230026, P. R. China
| |
Collapse
|
49
|
Strategies to Use Nanofiber Scaffolds as Enzyme-Based Biocatalysts in Tissue Engineering Applications. Catalysts 2021. [DOI: 10.3390/catal11050536] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Nanofibers are considered versatile materials with remarkable potential in tissue engineering and regeneration. In addition to their extracellular matrix-mimicking properties, nanofibers can be functionalized with specific moieties (e.g., antimicrobial nanoparticles, ceramics, bioactive proteins, etc.) to improve their overall performance. A novel approach in this regard is the use of enzymes immobilized onto nanofibers to impart biocatalytic activity. These nanofibers are capable of carrying out the catalysis of various biological processes that are essential in the healing process of tissue. In this review, we emphasize the use of biocatalytic nanofibers in various tissue regeneration applications. Biocatalytic nanofibers can be used for wound edge or scar matrix digestion, which reduces the hindrance for cell migration and proliferation, hence displaying applications in fast tissue repair, e.g., spinal cord injury. These nanofibers have potential applications in bone regeneration, mediating osteogenic differentiation, biomineralization, and matrix formation through direct enzyme activity. Moreover, enzymes can be used to undertake efficient crosslinking and fabrication of nanofibers with better physicochemical properties and tissue regeneration potential.
Collapse
|
50
|
Jeong H, Kim T, Earmme T, Hong J. Acceleration of Nitric Oxide Release in Multilayer Nanofilms through Cu(II) Ion Intercalation for Antibacterial Applications. Biomacromolecules 2021; 22:1312-1322. [PMID: 33617240 DOI: 10.1021/acs.biomac.0c01821] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Implant-derived bacterial infection is a prevalent cause of diseases, and no antibacterial coating currently exists that is biocompatible and that does not induce multidrug resistance. To this end, nitric oxide (NO) has been emerging as an effective antimicrobial agent that acts on a broad range of bacteria and elicits no known resistance. Here, a method for accelerating NO release from multilayered nanofilms has been developed for facilitating antibacterial activity. A previously reported multilayered nanofilm (nbi film) was fabricated by alternative deposition of branched polyethyleneimine (BPEI) and alginate via the layer-by-layer assembly method. N-Diazeniumdiolate, a chemical NO donor, was synthesized at the secondary amine moiety of BPEI within the film (nbi/NO film). Cu(II) ions can be incorporated into the film by forming chelating compounds with unreacted amines that have not been converted to NO donors. The increase of the amine protonation state in the chelate caused destabilization of the NO donor by reducing hydrogen bonding between the deprotonated amine and the NO donor. Thus, the Cu(II) ion-embedding film presented accelerated NO release and was further subjected to antibacterial testing to demonstrate the correlation between the NO release rate and the antibacterial activity. This study aimed to establish a novel paradigm for NO-releasing material design based on multilayered nanofilms by presenting the correlation between the NO release rate and the antibacterial effect.
Collapse
Affiliation(s)
- Hyejoong Jeong
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Taihyun Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Taeshik Earmme
- Department of Chemical Engineering, Hongik University, 94 Wausan-ro, Mapo-gu, Seoul 04066, Republic of Korea
| | - Jinkee Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|