1
|
Wang H, Bi D, Yu B, Chen Q, Du S, Xie G, Zhu J, Zhang L. Photonic hydrogels combining the slow photon effect and NO gas therapy for synergetic enhanced photodynamic antibacterial therapy. J Colloid Interface Sci 2025; 682:1185-1194. [PMID: 39671952 DOI: 10.1016/j.jcis.2024.12.018] [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: 09/23/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 12/15/2024]
Abstract
Photodynamic therapy (PDT) offers potential for combating bacterial infections through the generation of reactive oxygen species (ROS). However, the antibacterial efficiency of PDT is largely impeded by the limited photon absorption of photosensitizers and the short diffusion length and lifespan of ROS. Herein, we present a light-harvesting platform based on l-arginine-modified photonic hydrogels loaded with new indocyanine green (PG@Arg/IR820) for synergizing the slow photon effect with NO gas therapy to enhance PDT antibacterial efficiency. Upon near-infrared (NIR) light irradiation, PG@Arg/IR820 can maximize the utilization of photons via the slow photon effect to generate sufficient ROS, which not only acts as the primary bactericidal agent in PDT but also triggers l-arginine to generate NO. NO exhibits a long diffusion distance and lifespan and can freely diffuse to inhibit distant bacterial growth, demonstrating a vital complementary advantage in bacterial inactivation by ROS. The synergistic effect of the slow photon effect combined with NO gas therapy allows PG@Arg/IR820 to intensify bacterial destruction and enhance PDT antibacterial efficiency. This antibacterial system sheds light on an advisable design principle for efficient antibacterial activities in photodynamic inactivation.
Collapse
Affiliation(s)
- Hui Wang
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Duohang Bi
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Bowen Yu
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Qiang Chen
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Shuo Du
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Ge Xie
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China
| | - Jintao Zhu
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| | - Lianbin Zhang
- Key Lab of Material Chemistry for Energy Conversion and Storage of Ministry of Education, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), Wuhan 430074, China.
| |
Collapse
|
2
|
Tian C, Liu X, Hao Y, Fu H, Shao X, Cai W. Flexible Tail of Antimicrobial Peptide PGLa Facilitates Water Pore Formation in Membranes. J Phys Chem B 2025. [PMID: 39847609 DOI: 10.1021/acs.jpcb.4c06190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
PGLa, an antimicrobial peptide (AMP), primarily exerts its antibacterial effects by disrupting bacterial cell membrane integrity. Previous theoretical studies mainly focused on the binding mechanism of PGLa with membranes, while the mechanism of water pore formation induced by PGLa peptides, especially the role of structural flexibility in the process, remains unclear. In this study, using all-atom simulations, we investigated the entire process of membrane deformation caused by the interaction of PGLa with an anionic cell membrane composed of dimyristoylphosphatidylcholine (DMPC) and dimyristoylphosphatidylglycerol (DMPG). Using a deep learning-based key intermediate identification algorithm, we found that the C-terminal tail plays a crucial role for PGLa insertion into the membrane, and that with its assistance, a variety of water pores formed inside the membrane. Mutation of the tail residues revealed that, in addition to electrostatic and hydrophobic interactions, the flexibility of the tail residues is crucial for peptide insertion and pore formation. The full extension of these flexible residues enhances peptide-peptide and peptide-membrane interactions, guiding the transmembrane movement of PGLa and the aggregation of PGLa monomers within the membrane, ultimately leading to the formation of water-filled pores in the membrane. Overall, this study provides a deep understanding of the transmembrane mechanism of PGLa and similar AMPs, particularly elucidating for the first time the importance of C-terminal flexibility in both insertion and oligomerization processes.
Collapse
Affiliation(s)
- Chunsuo Tian
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical, Biology College of Chemistry, Nankai University, Tianjin 300071, China
| | - Xuyang Liu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical, Biology College of Chemistry, Nankai University, Tianjin 300071, China
| | - Yuelei Hao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical, Biology College of Chemistry, Nankai University, Tianjin 300071, China
| | - Haohao Fu
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical, Biology College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Xueguang Shao
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical, Biology College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| | - Wensheng Cai
- Research Center for Analytical Sciences, Tianjin Key Laboratory of Biosensing and Molecular Recognition, State Key Laboratory of Medicinal Chemical, Biology College of Chemistry, Nankai University, Tianjin 300071, China
- Haihe Laboratory of Sustainable Chemical Transformations, Tianjin 300192, China
| |
Collapse
|
3
|
Lu Z, Fan W, Ye Y, Huang Y, Zhou X, Zhang Y, Cui W, Ji J, Yao K, Han H. Drug in Drug: Quorum Sensing Inhibitor in Star-Shaped Antibacterial Polypeptides for Inhibiting and Eradicating Corneal Bacterial Biofilms. ACS NANO 2025; 19:2268-2285. [PMID: 39772450 DOI: 10.1021/acsnano.4c12195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Biofilm-related bacterial keratitis is a severe ocular infection that can result in drastic vision impairment and even blindness. However, the therapeutic efficiency of clinical antibiotic eyedrops is often compromised because the bacteria in the biofilms resist bactericide via the community genetic regulation, namely, bacterial quorum sensing. Herein, quercetin (QCT)-loaded star-shaped antibacterial peptide polymer (SAPP), QCT@SAPP, is developed based on a "drug" in a "drug" strategy for inhibiting and eradicating Pseudomonas aeruginosa biofilms on the cornea. The natural antibacterial peptide-mimic SAPP with the positively charged amphipathic structure not only enables QCT@SAPP to penetrate the biofilms readily but also selectively adheres to the highly negatively charged P. aeruginosa, releasing the loaded QCT into the bacteria to regulate quorum sensing by inhibiting lasI, lasR, rhlR, and rhlI. Thanks to its robust bactericidal ability from SAPP, QCT@SAPP can eliminate more than 99.99% of biofilms. Additionally, QCT@SAPP displayed outstanding performance in relieving ocular inflammation by significantly downregulating pro-inflammatory cytokines and profiting from scavenging reactive oxygen species by releasing QCT, which finally helps to restore visual function. In conclusion, QCT@SAPP, with good compatibility, exerts excellent therapeutic effects in a bacterial keratitis mice model, making it a promising candidate for controlling bacterial biofilm-induced infections, including bacterial keratitis.
Collapse
Affiliation(s)
- Zhouyu Lu
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Wenjie Fan
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Yang Ye
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Yue Huang
- 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
| | - Xianchi Zhou
- 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
| | - Yin Zhang
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Wenyu Cui
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Jian Ji
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. 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
| | - Ke Yao
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| | - Haijie Han
- Eye Center, the Second Affiliated Hospital, School of Medicine, Zhejiang University, Zhejiang Provincial Key Laboratory of Ophthalmology, Zhejiang Provincial Clinical Research Center for Eye Diseases, Zhejiang Provincial Engineering Institute on Eye Diseases, Hangzhou 310009, P. R. China
| |
Collapse
|
4
|
Zhang L, Chen Y, Feng D, Xing Z, Wang Y, Bai Y, Shi D, Li H, Fan X, Xia J, Wang J. Recombinant collagen microneedles for transdermal delivery of antibacterial copper-DNA nanoparticles to treat skin and soft tissue infections. J Control Release 2025; 379:191-201. [PMID: 39793653 DOI: 10.1016/j.jconrel.2025.01.016] [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: 10/22/2024] [Revised: 12/29/2024] [Accepted: 01/07/2025] [Indexed: 01/13/2025]
Abstract
Skin and soft tissue infections (SSTI) include bacterial infections of the skin, muscles, and connective tissue such as ligaments and tendons. SSTI in patients with immunocompromising diseases may lead to chronic, hard-to-heal infected wounds, resulting in disability, amputation, or even death. To treat SSTI and rebuild the defensive barrier of the skin, here we utilize recombinant type XVII collagen protein (rCol XVII) to construct biodegradable, regenerative collagen microneedles (rCol-MNs) for transdermal delivery of antibacterial agents. Spheroidal copper-DNA antibacterial nanoparticles (Cu-CpG NPs; CpG represents short single-stranded synthetic DNA molecules of cytosine and guanine) are synthesized with copper ions and CpG oligodeoxynucleotides (ODNs), followed by polydopamine (PDA) coating to obtain Cu-CpG@PDA. Doping Cu-CpG@PDA into rCol-MNs yields Cu-CpG@PDA-loaded rCol-MNs. These microneedles combine the photothermal conversion property of PDA, antibacterial properties of copper ions, innate immune activation of CpG ODNs, and skin regenerating ability of rCol XVII, allowing the treatment of SSTI and also regenerating the damaged skin. In a mouse model, we show that the Cu-CpG@PDA-loaded rCol-MNs rescue skin wound infections, facilitate the orderly deposition of collagen at the wound site, and promote the healing of infected full-thickness wounds without noticeable scar formation. rCol-MNs serve as a transdermal delivery vehicle and, simultaneously, a reservoir of skin-regenerating recombinant collagen, bringing combined benefits of infection control and skin regeneration. SIGNIFICANCE STATEMENT: Treatment of soft tissue infection requires the delivery of antibacterial agents into the soft tissue or dermis while providing a regenerating environment for open wounds. Here, we devise recombinant collagen microneedles (rCol-MNs) to meet both requirements.
Collapse
Affiliation(s)
- Li Zhang
- School of Pharmacy, Changzhou University, Changzhou 213164, China; Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Yifan Chen
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Danna Feng
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Zheng Xing
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Yuhui Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Yang Bai
- School of Pharmacy, Changzhou University, Changzhou 213164, China
| | - Dongjian Shi
- Key Laboratory of Synthetic and Biological Colloids, Ministry of Education, Jiangnan University, Wuxi 214122, China
| | - Haihang Li
- Jiangsu Trautec Medical Technology Co., Ltd, Changzhou 213200, PR China
| | - Xiaoju Fan
- Jiangsu Trautec Medical Technology Co., Ltd, Changzhou 213200, PR China
| | - Jiang Xia
- Department of Chemistry, the Chinese University of Hong Kong, Shatin, Hong Kong Special Administrative Region.
| | - Jianhao Wang
- School of Pharmacy, Changzhou University, Changzhou 213164, China; School of Medical and Health Engineering, Changzhou University, Changzhou 213164, PR China.
| |
Collapse
|
5
|
Li M, Zhao P, Wang J, Zhang X, Li J. Functional antimicrobial peptide-loaded 3D scaffolds for infected bone defect treatment with AI and multidimensional printing. MATERIALS HORIZONS 2025; 12:20-36. [PMID: 39484845 DOI: 10.1039/d4mh01124d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Infection is the most prevalent complication of fractures, particularly in open fractures, and often leads to severe consequences. The emergence of bacterial resistance has significantly exacerbated the burden of infection in clinical practice, making infection control a significant treatment challenge for infectious bone defects. The implantation of a structural stent is necessary to treat large bone defects despite the increased risk of infection. Therefore, there is a need for the development of novel antibacterial therapies. The advancement in antibacterial biomaterials and new antimicrobial drugs offers fresh perspectives on antibacterial treatment. Although antimicrobial 3D scaffolds are currently under intense research focus, relying solely on material properties or antibiotic action remains insufficient. Antimicrobial peptides (AMPs) are one of the most promising new antibacterial therapy approaches. This review discusses the underlying mechanisms behind infectious bone defects and presents research findings on antimicrobial peptides, specifically emphasizing their mechanisms and optimization strategies. We also explore the potential prospects of utilizing antimicrobial peptides in treating infectious bone defects. Furthermore, we propose that artificial intelligence (AI) algorithms can be utilized for predicting the pharmacokinetic properties of AMPs, including absorption, distribution, metabolism, and excretion, and by combining information from genomics, proteomics, metabolomics, and clinical studies with computational models driven by machine learning algorithms, scientists can gain a comprehensive understanding of AMPs' mechanisms of action, therapeutic potential, and optimizing treatment strategies tailored to individual patients, and through interdisciplinary collaborations between computer scientists, biologists, and clinicians, the full potential of AI in accelerating the discovery and development of novel AMPs will be realized. Besides, with the continuous advancements in 3D/4D/5D/6D technology and its integration into bone scaffold materials, we anticipate remarkable progress in the field of regenerative medicine. This review summarizes relevant research on the optimal future for the treatment of infectious bone defects, provides guidance for future novel treatment strategies combining multi-dimensional printing with new antimicrobial agents, and provides a novel and effective solution to the current challenges in the field of bone regeneration.
Collapse
Affiliation(s)
- Mengmeng Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Peizhang Zhao
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jingwen Wang
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xincai Zhang
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA.
| | - Jun Li
- Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
- Trauma Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| |
Collapse
|
6
|
Halder A, Pasupuleti R, Sivagnanam S, Das P, Mukherjee O. Boc-Protected Phenylalanine and Tryptophan-Based Dipeptides: A Broad Spectrum Anti-Bacterial Agent. Biopolymers 2025; 116:e23649. [PMID: 39718897 DOI: 10.1002/bip.23649] [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: 08/16/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024]
Abstract
Dipeptides were constructed using hydrophobic amino acid residues following AMP prediction. After that Boc-modification was performed on the screened peptides and finally Boc-Phe-Trp-OMe and Boc-Trp-Trp-OMe were synthesized. Even though no inhibition zones were observed in agar well diffusion assays, minimum inhibitory concentration (MIC) analysis revealed anti-bacterial activity against both Gram-positive and Gram-negative bacteria, with MIC90 ranging from 230 to 400 μg/mL. The crystal violet assay confirmed the dipeptides' biofilm eradication and disruption capabilities. Furthermore, membrane permeabilization assays indicated outer and inner membrane permeabilization, while SEM analysis revealed the formation of fibril and spherical nanostructures, likely contributing to this effect. The peptides also exhibited resistance to protein adsorption, non-cytotoxicity, and non-hemolytic properties, making them promising broad-spectrum anti-bacterial agents with biofilm eradication and disruption potential. This study concludes that Boc-protected phenylalanine- and tryptophan-based dipeptides can self-assemble and can be used as broad-spectrum anti-bacterial agents. The self-assembly of these peptides offers a versatile platform for designing biomaterials with tailored properties and functionalities. Research exploring the anti-bacterial potential of Boc-protected dipeptides has been limited, prompting our investigation to shed light on this overlooked area. Our analysis of synthesized Boc-protected dipeptides revealed notable anti-bacterial activity, marking a significant advancement. This finding suggests that these dipeptides could emerge as potent, broad-spectrum anti-bacterial agents, addressing the urgent need for effective treatments against bacterial resistance and opening new avenues in therapy. This study not only enhances our understanding of these dipeptides but also highlights their potential as innovative and efficacious anti-bacterial agents, making a substantial impact in the clinical field.
Collapse
Affiliation(s)
- Arpita Halder
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India
| | | | | | - Priyadip Das
- Department of Chemistry, SRMIST, Kattankulathur, Chennai, Tamil Nadu, India
| | - Oindrilla Mukherjee
- Department of Biotechnology, National Institute of Technology Durgapur, Durgapur, West Bengal, India
| |
Collapse
|
7
|
Xu X, Yu YB. Role of antimicrobial peptides in gastrointestinal diseases: Recent advances. Shijie Huaren Xiaohua Zazhi 2024; 32:865-871. [DOI: 10.11569/wcjd.v32.i12.865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/26/2024] [Accepted: 10/21/2024] [Indexed: 12/28/2024] Open
Abstract
Gastrointestinal diseases, especially gastrointestinal inflammation and tumors, affect millions of people world-wide, adversely affecting the health and quality of life of individuals. In recent years, with the continuous advances of relevant research, the diagnosis and treatment of gastrointestinal diseases have made great progress. However, traditional therapies still have drawbacks such as poor efficacy and side effects. Antimicrobial peptides, as part of the innate immune defense of many organisms, not only have broad-spectrum antibacterial activity and immune modulating function, which can assist in maintaining homeostasis within the gastrointestinal tract, but also can specifically kill tumor cells, showing good prospects in the treatment of gastrointestinal diseases. In this review, we briefly outline the related studies on the role of antimicrobial peptides in gastrointestinal diseases in the last decade, and discuss the application potential and challenges that they face.
Collapse
Affiliation(s)
- Xia Xu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250000, Shandong Province, China
| | - Yan-Bo Yu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan 250000, Shandong Province, China
| |
Collapse
|
8
|
Chen S, Xi J, Xu Y, Zhang Y, Du W, Meng X, Zhang J, Hu X, Wang Y. Extracellular Matrix-Inspired Antibacterial Fibrous Hydrogels Containing Polyhexamethylene Biguanide and Gd 3. Macromol Rapid Commun 2024:e2400696. [PMID: 39632411 DOI: 10.1002/marc.202400696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 10/30/2024] [Indexed: 12/07/2024]
Abstract
Traditional bulk hydrogels containing antibiotics or metal ions often fall short in effectively treating wound infections due to mechanical limitations, bacterial resistance, and potential cytotoxicity. To address these challenges, an extracellular matrix (ECM)-inspired antibacterial fibrous hydrogel featuring an anisotropic topological structure is developed that closely mimics the natural ECM environment. A novel antibacterial agent, PHMB-VAN-Gd (PVG), is synthesized by reacting polyhexamethylene biguanide (PHMB) with O-Vanillin (VAN) to form the Schiff base ligand PHMB-VAN (PV), followed by coordination with gadolinium ions (Gd3⁺). Employing silk fibroin (SF) as the matrix, the PVG complex is incorporated into fibrous hydrogels through electrospinning, generating structures that replicate the fibrous architecture of the ECM. The resulting SF-PVG fibrous hydrogels exhibited robust antibacterial activity, effectively inhibiting bacterial growth and biofilm formation. Furthermore, the aligned fiber orientation and substantial mechanical strength of these hydrogels facilitated cellular functions, promoting cell attachment and proliferation. This study underscores the significant potential of SF-PVG hydrogels for wound infection treatment.
Collapse
Affiliation(s)
- Siyuan Chen
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jingyun Xi
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuanyuan Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Yuxin Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Wenli Du
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xinyue Meng
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Jieyu Zhang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| | - Xuefeng Hu
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan, 610065, China
| |
Collapse
|
9
|
Zhang Y, Xu T, Li T, Chen H, Xu G, Hu W, Li Y, Dong Y, Liu Z, Han B. A three-phase strategy of bionic drug reservoir scaffold by 3D printing and layer-by-layer modification for chronic relapse management in traumatic osteomyelitis. Mater Today Bio 2024; 29:101356. [PMID: 39687799 PMCID: PMC11648807 DOI: 10.1016/j.mtbio.2024.101356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/11/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
We have developed a novel three-phase strategy for osteomyelitis treatment, structured into three distinct phases: the "strong antimicrobial" phase, the "monitoring and osteogenesis" phase and the "bone repair" phase. To implement this staged therapeutic strategy, we engineered a bionic drug reservoir scaffold carrying a dual-drug combination of antimicrobial peptides (AMPs) and simvastatin (SV). The scaffold integrated a bilayer gel drug-carrying structure, based on an induced membrane and combined with a 3D-printed rigid bone graft using a layer-by-layer modification strategy. The mechanical strength of the composite scaffold (73.40 ± 22.44 MPa) is comparable to that of cancellous bone. This scaffold enables controlled, sequential drug release through a spatial structure design and nanoparticle drug-carrying strategy. AMPs are released rapidly, with the release efficiency of 74.90 ± 8.19 % at 14 days (pH = 7.2), thus enabling rapid antimicrobial therapy. Meanwhile, SV is released over a prolonged period, with a release efficiency of 98.98 ± 0.05 % over 40 days in vitro simulations, promoting sustained osteogenesis and facilitating the treatment of intracellular infections by activating macrophage extracellular traps (METs). The antimicrobial, osteogenic and immunomodulatory effects of the scaffolds were verified through in vitro and in vivo experiments. It was demonstrated that composite scaffolds were able to combat the chronic recurrence of osteomyelitis after debridement, by providing rapid sterilization, stimulating METs formation, and supporting osteogenic repair.
Collapse
Affiliation(s)
- Yutong Zhang
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Tongtong Xu
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Tieshu Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
- Changchun University of Chinese Medicine, Changchun, 130117, China
- Affiliated Hospital of Yangzhou University, Yangzhou, 225009, China
| | - Hening Chen
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Guangzhe Xu
- Changchun University of Chinese Medicine, Changchun, 130117, China
| | - Wenxin Hu
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yongting Li
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Yue Dong
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| | - Zhihui Liu
- Department of Prosthodontics, Hospital of Stomatology, Jilin University, Changchun, 130021, China
| | - Bing Han
- School of Pharmaceutical Sciences, Jilin University, Changchun, 130021, China
| |
Collapse
|
10
|
Zhang K, Yang N, Teng D, Mao R, Hao Y, Wang J. Expression and characterization of the new antimicrobial peptide AP138L-arg26 anti Staphylococcus aureus. Appl Microbiol Biotechnol 2024; 108:111. [PMID: 38229298 DOI: 10.1007/s00253-023-12947-w] [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: 08/08/2023] [Revised: 10/18/2023] [Accepted: 10/25/2023] [Indexed: 01/18/2024]
Abstract
The low activity and yield of antimicrobial peptides (AMPs) are pressing problems. The improvement of activity and yield through modification and heterologous expression, a potential way to solve the problem, is a research hot-pot. In this work, a new plectasin-derived variant L-type AP138 (AP138L-arg26) was constructed for the study of recombination expression and druggablity. As a result, the total protein concentration of AP138L-arg26 was 3.1 mg/mL in Pichia pastoris X-33 supernatant after 5 days of induction expression in a 5-L fermenter. The recombinant peptide AP138L-arg26 has potential antibacterial activity against selected standard and clinical Gram-positive bacteria (G+, minimum inhibitory concentration (MIC) 2-16 µg/mL) and high stability under different conditions (temperature, pH, ion concentration) and 2 × MIC of AP138L-arg26 could rapidly kill Staphylococcus aureus (S. aureus) (> 99.99%) within 1.5 h. It showed a high safety in vivo and in vivo and a long post-antibiotic effect (PAE, 1.91 h) compared with vancomycin (1.2 h). Furthermore, the bactericidal mechanism was revealed from two dimensions related to its disruption of the cell membrane resulting in intracellular potassium leakage (2.5-fold higher than control), and an increase in intracellular adenosine triphosphate (ATP), and reactive oxygen species (ROS), the decrease of lactate dehydrogenase (LDH) and further intervening metabolism in S. aureus. These results indicate that AP138L-arg26 as a new peptide candidate could be used for more in-depth development in the future. KEY POINTS: • The AP138L-arg26 was expressed in the P. pastoris expression system with high yield • The AP138 L-arg26 showed high stability and safety in vitro and in vivo • The AP138L-arg26 killed S. aureus by affecting cell membranes and metabolism.
Collapse
Affiliation(s)
- Kun Zhang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Na Yang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China.
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China.
| | - Da Teng
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Ruoyu Mao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Ya Hao
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China
| | - Jianhua Wang
- Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, No. 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, People's Republic of China.
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, People's Republic of China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, People's Republic of China.
| |
Collapse
|
11
|
Chaudhary S, Ali Z, Pantoja‐Angles A, Abdelrahman S, Juárez COB, Rao GS, Hong P, Hauser C, Mahfouz M. High-yield, plant-based production of an antimicrobial peptide with potent activity in a mouse model. PLANT BIOTECHNOLOGY JOURNAL 2024; 22:3392-3405. [PMID: 39264967 PMCID: PMC11606426 DOI: 10.1111/pbi.14460] [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: 03/15/2024] [Revised: 08/18/2024] [Accepted: 08/21/2024] [Indexed: 09/14/2024]
Abstract
Plants offer a promising chassis for the large-scale, cost-effective production of diverse therapeutics, including antimicrobial peptides (AMPs). However, key advances will reduce production costs, including simplifying the downstream processing and purification steps. Here, using Nicotiana benthamiana plants, we present an improved modular design that enables AMPs to be secreted via the endomembrane system and sequestered in an extracellular compartment, the apoplast. Additionally, we translationally fused an AMP to a mutated small ubiquitin-like modifier sequence, thereby enhancing peptide yield and solubilizing the peptide with minimal aggregation and reduced occurrence of necrotic lesions in the plant. This strategy resulted in substantial peptide accumulation, reaching around 2.9 mg AMP per 20 g fresh weight of leaf tissue. Furthermore, the purified AMP demonstrated low collateral toxicity in primary human skin cells, killed pathogenic bacteria by permeabilizing the membrane and exhibited anti-infective efficacy in a preclinical mouse (Mus musculus) model system, reducing bacterial loads by up to three orders of magnitude. A base-case techno-economic analysis demonstrated the economic advantages and scalability of our plant-based platform. We envision that our work can establish plants as efficient bioreactors for producing preclinical-grade AMPs at a commercial scale, with the potential for clinical applications.
Collapse
Affiliation(s)
- Shahid Chaudhary
- Laboratory for Genome Engineering and Synthetic Biology, Division of Biological SciencesKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Zahir Ali
- Laboratory for Genome Engineering and Synthetic Biology, Division of Biological SciencesKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Aarón Pantoja‐Angles
- Laboratory for Genome Engineering and Synthetic Biology, Division of Biological SciencesKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Sherin Abdelrahman
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and EngineeringKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
- Computational Bioscience Research CenterKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
- Red Sea Research CenterKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Cynthia Olivia Baldelamar Juárez
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and EngineeringKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
- Computational Bioscience Research CenterKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
- Red Sea Research CenterKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Gundra Sivakrishna Rao
- Laboratory for Genome Engineering and Synthetic Biology, Division of Biological SciencesKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Pei‐Ying Hong
- Water Desalination and Reuse Center, Division of Biological Sciences and EngineeringKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Charlotte Hauser
- Laboratory for Nanomedicine, Division of Biological and Environmental Science and EngineeringKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
- Computational Bioscience Research CenterKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
- Red Sea Research CenterKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| | - Magdy Mahfouz
- Laboratory for Genome Engineering and Synthetic Biology, Division of Biological SciencesKing Abdullah University of Science and Technology (KAUST)ThuwalJeddahSaudi Arabia
| |
Collapse
|
12
|
Arévalo-Jaimes BV, Salinas-Pena M, Ponte I, Jordan A, Roque A, Torrents E. Antimicrobial and antibiofilm activity of human recombinant H1 histones against bacterial infections. mSystems 2024; 9:e0070424. [PMID: 39470247 PMCID: PMC11575268 DOI: 10.1128/msystems.00704-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Histones possess significant antimicrobial potential, yet their activity against biofilms remains underexplored. Moreover, concerns regarding adverse effects limit their clinical implementation. We investigated the antibacterial efficacy of human recombinant histone H1 subtypes against Pseudomonas aeruginosa PAO1, both planktonic and in biofilms. After the in vitro tests, toxicity and efficacy were assessed in a P. aeruginosa PAO1 infection model using Galleria mellonella larvae. Histones were also evaluated in combination with ciprofloxacin (Cpx) and gentamicin (Gm). Our results demonstrate antimicrobial activity of all three histones against P. aeruginosa PAO1, with H1.0 and H1.4 showing efficacy at lower concentrations. The bactericidal effect was associated with a mechanism of membrane disruption. In vitro studies using static and dynamic models showed that H1.4 had antibiofilm potential by reducing cell biomass. Neither H1.0 nor H1.4 showed toxicity in G. mellonella larvae, and both increased larvae survival when infected with P. aeruginosa PAO1. Although in vitro synergism was observed between ciprofloxacin and H1.0, no improvement over the antibiotic alone was noted in vivo. Differences in antibacterial and antibiofilm activity were attributed to sequence and structural variations among histone subtypes. Moreover, the efficacy of H1.0 and H1.4 was influenced by the presence and strength of the extracellular matrix. These findings suggest histones hold promise for combating acute and chronic infections caused by pathogens such as P. aeruginosa.IMPORTANCEThe constant increase of multidrug-resistant bacteria is a critical global concern. The inefficacy of current therapies to treat bacterial infections is attributed to multiple mechanisms of resistance, including the capacity to form biofilms. Therefore, the identification of novel and safe therapeutic strategies is imperative. This study confirms the antimicrobial potential of three histone H1 subtypes against both Gram-negative and Gram-positive bacteria. Furthermore, histones H1.0 and H1.4 demonstrated in vivo efficacy without associated toxicity in an acute infection model of Pseudomonas aeruginosa PAO1 in Galleria mellonella larvae. The bactericidal effect of these proteins also resulted in biomass reduction of P. aeruginosa PAO1 biofilms. Given the clinical significance of this opportunistic pathogen, our research provides a comprehensive initial evaluation of the efficacy, toxicity, and mechanism of action of a potential new therapeutic approach against acute and chronic bacterial infections.
Collapse
Affiliation(s)
- Betsy Verónica Arévalo-Jaimes
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| | | | - Inmaculada Ponte
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Albert Jordan
- Molecular Biology Institute of Barcelona (IBMB-CSIC), Barcelona, Spain
| | - Alicia Roque
- Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Eduard Torrents
- Bacterial infections and antimicrobial therapies group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- Microbiology Section, Department of Genetics, Microbiology and Statistics, Faculty of Biology, University of Barcelona, Barcelona, Spain
| |
Collapse
|
13
|
Lombardi L, Li J, Williams DR. Peptide-Based Biomaterials for Combatting Infections and Improving Drug Delivery. Pharmaceutics 2024; 16:1468. [PMID: 39598591 PMCID: PMC11597775 DOI: 10.3390/pharmaceutics16111468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 11/14/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
This review explores the potential of peptide-based biomaterials to enhance biomedical applications through self-assembly, biological responsiveness, and selective targeting. Peptides are presented as versatile agents for antimicrobial activity and drug delivery, with recent approaches incorporating antimicrobial peptides into self-assembling systems to improve effectiveness and reduce resistance. The review also covers peptide-based nanocarriers for cancer drug delivery, highlighting their improved stability, targeted delivery, and reduced side effects. The focus of this work is on the bioactive properties of peptides, particularly in infection control and drug delivery, rather than on their structural design or material characteristics. Additionally, it examines the role of peptidomimetics in broadening biomaterial applications and enhancing resistance to enzymatic degradation. Finally, the review discusses the commercial prospects and challenges of translating peptide biomaterials into clinical applications.
Collapse
Affiliation(s)
- Lucia Lombardi
- Department of Chemical Engineering, South Kensington Campus, Imperial College London, London SW7 2AZ, UK
| | | | | |
Collapse
|
14
|
Wang Y, Miao G, Wang S, Zhou F. Design, synthesis, and evaluation of pyranochromene derivatives as membrane targeting antibacterials against Gram-positive bacteria. Bioorg Med Chem Lett 2024; 113:129949. [PMID: 39243868 DOI: 10.1016/j.bmcl.2024.129949] [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: 08/08/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/09/2024]
Abstract
The rapid growth of bacterial resistance has created obstacles for the effective treatment with conventional antibiotics, simultaneously posing a major threat to public health. In this study, a class of novel amphipathic pyranochromene derivatives were designed and synthesized by mimicking the amphiphilic characteristics of AMPs. Bioactivity screening identified a lead compound 5a with broad-spectrum antibacterial activity against Gram-positive stains (MICs = 1-4 μg/mL) and low hemolytic toxicity (HC50 = 111.6 μg/mL). Additionally, compound 5a displayed rapid bactericidal action, and was unlikely to induce bacterial resistance. Mechanistic investigation further demonstrated that compound 5a was able to disrupt the transmembrane potential and increased membrane permeability of S. aureus, which in turn causes leakage of cell contents such as DNA and proteins, ultimately leading to bacterial death. These findings indicated that compound 5a is a promising lead to combat bacterial infection caused by Gram-positive bacteria.
Collapse
Affiliation(s)
- Yinhu Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China.
| | - Guoqing Miao
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Shuo Wang
- State Key Laboratory for Macromolecule Drugs and Large-scale Manufacturing, School of Pharmaceutical Sciences and Food Engineering, Liaocheng University, Liaocheng 252059, China
| | - Fen Zhou
- Department of Pharmacy, Liaocheng People's Hospital, Liaocheng, China.
| |
Collapse
|
15
|
Coelho AA, Barbosa LCDS, da Costa AC, Kipnis A, Junqueira-Kipnis AP. Novel Synthetic Peptide Agelaia-12 Has Improved Activity Against Mycobacterium abscessus Complex. Pathogens 2024; 13:994. [PMID: 39599547 PMCID: PMC11597844 DOI: 10.3390/pathogens13110994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Fast-growing mycobacteria cause difficult-to-treat infections due to their high intrinsic resistance to antibiotics as well as disinfectant agents. Mycobacterium abscessus complex (MAC) is the main cause of nontuberculous mycobacteria diseases. In this work, we evaluated the activity of the novel synthetic antimicrobial peptide, Agelaia-12, against Mycobacterium abscessus and M. massiliense. Agelaia-12 showed a minimum inhibitory concentration (MIC) of 25 μM detected against M. abscessus and M. massiliense with no cytotoxicity. The scanning electronic microscopy analysis of mycobacterial treated with Agelaia-12 demonstrated the presence of filamentous structures and aggregation of the cells. Congo red binding assay of M. abscessus exhibited altered dye accumulation after treatment with Agelaia-12. Treatment of M. abscessus- or M. massiliense-infected murine macrophages with Agelaia-12 decreased the mycobacterial load by 92% for the tested strains. Additionally, IFN-y KO mice infected with M. abscessus or M. massiliense and treated with Agelaia-12 showed a 98% reduction in lung bacterial load. Thus, the synthetic peptide Agelaia-12 may be a promising biomolecule for the treatment of mycobacteriosis, and its structural properties may serve as a foundational model for the design and development of novel pharmaceutical agents aimed at combating this disease.
Collapse
Affiliation(s)
- Arthur Alves Coelho
- Department of Biosciences and Technology, Tropical Pathology and Public Health Institute, Federal University of Goiás, Goiânia 74605-050, GO, Brazil; (A.A.C.); (L.C.d.S.B.); (A.C.d.C.); (A.K.)
| | - Lília Cristina de Souza Barbosa
- Department of Biosciences and Technology, Tropical Pathology and Public Health Institute, Federal University of Goiás, Goiânia 74605-050, GO, Brazil; (A.A.C.); (L.C.d.S.B.); (A.C.d.C.); (A.K.)
| | - Adeliane Castro da Costa
- Department of Biosciences and Technology, Tropical Pathology and Public Health Institute, Federal University of Goiás, Goiânia 74605-050, GO, Brazil; (A.A.C.); (L.C.d.S.B.); (A.C.d.C.); (A.K.)
- Health Sciences Academic Unit, Federal University of Jataí, Jatái 75801-615, GO, Brazil
| | - André Kipnis
- Department of Biosciences and Technology, Tropical Pathology and Public Health Institute, Federal University of Goiás, Goiânia 74605-050, GO, Brazil; (A.A.C.); (L.C.d.S.B.); (A.C.d.C.); (A.K.)
| | - Ana Paula Junqueira-Kipnis
- Department of Biosciences and Technology, Tropical Pathology and Public Health Institute, Federal University of Goiás, Goiânia 74605-050, GO, Brazil; (A.A.C.); (L.C.d.S.B.); (A.C.d.C.); (A.K.)
| |
Collapse
|
16
|
Canè C, Tammaro L, Duilio A, Di Somma A. Investigation of the Mechanism of Action of AMPs from Amphibians to Identify Bacterial Protein Targets for Therapeutic Applications. Antibiotics (Basel) 2024; 13:1076. [PMID: 39596769 PMCID: PMC11591259 DOI: 10.3390/antibiotics13111076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 11/08/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Antimicrobial peptides (AMPs) from amphibians represent a promising source of novel antibacterial agents due to their potent and broad-spectrum antimicrobial activity, which positions them as valid alternatives to conventional antibiotics. This review provides a comprehensive analysis of the mechanisms through which amphibian-derived AMPs exert their effects against bacterial pathogens. We focus on the identification of bacterial protein targets implicated in the action of these peptides and on biological processes altered by the effect of AMPs. By examining recent advances in countering multidrug-resistant bacteria through multi-omics approaches, we elucidate how AMPs interact with bacterial membranes, enter bacterial cells, and target a specific protein. We discuss the implications of these interactions in developing targeted therapies and overcoming antibiotic resistance (ABR). This review aims to integrate the current knowledge on AMPs' mechanisms, identify gaps in our understanding, and propose future directions for research to harness amphibian AMPs in clinical applications.
Collapse
Affiliation(s)
- Carolina Canè
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy; (C.C.); (L.T.)
| | - Lidia Tammaro
- CEINGE Biotecnologie Avanzate Franco Salvatore, 80145 Naples, Italy; (C.C.); (L.T.)
| | - Angela Duilio
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cinthia 4, 80126 Napoli, Italy;
- National Institute of Biostructures and Biosystems (INBB), Via dei Carpegna 19, 00165 Roma, Italy
| | - Angela Di Somma
- Department of Chemical Sciences, University of Naples “Federico II”, Via Cinthia 4, 80126 Napoli, Italy;
| |
Collapse
|
17
|
Tancer R, Pawar S, Wang Y, Ventura CR, Wiedman G, Xue C. Improved Broad Spectrum Antifungal Drug Synergies with Cryptomycin, a Cdc50-Inspired Antifungal Peptide. ACS Infect Dis 2024; 10:3973-3993. [PMID: 39475550 PMCID: PMC11555678 DOI: 10.1021/acsinfecdis.4c00681] [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: 08/23/2024] [Revised: 10/15/2024] [Accepted: 10/22/2024] [Indexed: 11/09/2024]
Abstract
Fungal infections in humans are difficult to treat, with very limited drug options. Due to a confluence of factors, there is an urgent need for innovation in the antifungal drug space, particularly to combat increasing antifungal drug resistance. Our previous studies showed that Cdc50, a subunit of fungal lipid translocase (flippase), is essential for Cryptococcus neoformans virulence and required for antifungal drug resistance, suggesting that fungal lipid flippase could be a novel drug target. Here, we characterized an antifungal peptide, Cryptomycinamide (KKOO-NH2), derived from a 9-amino acid segment of the C. neoformans Cdc50 protein. A fungal killing assay indicated that KKOO-NH2 is fungicidal against C. neoformans. The peptide has antifungal activity against multiple major fungal pathogens with a minimum inhibitory concentration (MIC) of 8 μg/mL against C. neoformans and Candida glabrata, 16 μg/mL against Candida albicans and C. auris, and 32 μg/mL against Aspergillus fumigatus. The peptide has low cytotoxicity against host cells based on our hemolysis assays and vesicle leakage assays. Strikingly, the peptide exhibits strong drug synergy with multiple antifungal drugs, including amphotericin B, itraconazole, and caspofungin, depending on the specific species on which the combinations were assayed. The fluorescently labeled peptide was detected to localize to the plasma membrane, likely inhibiting key interactions of Cdc50 with membrane proteins such as P4 ATPases. Cryptococcus cells exposed to sub-MIC of peptide showed increased reactive oxygen species production and intracellular calcium levels, indicating a peptide-induced stress response. Decreased intracellular proliferation within macrophages was observed after 30 min of peptide exposure and 24 h coincubation with macrophages, providing a potential translational mechanism to explore further in vivo. In aggregate, the synergistic activity of our KKOO-NH2 peptide may offer a potential novel candidate for combination therapy with existing antifungal drugs.
Collapse
Affiliation(s)
- Robert
J. Tancer
- Public
Health Research Institute and Department of Microbiology, Biochemistry,
and Molecular Genetics, New Jersey Medical
School, Rutgers University, Newark, New Jersey 07103, United States
| | - Siddhi Pawar
- Public
Health Research Institute and Department of Microbiology, Biochemistry,
and Molecular Genetics, New Jersey Medical
School, Rutgers University, Newark, New Jersey 07103, United States
| | - Yina Wang
- Public
Health Research Institute and Department of Microbiology, Biochemistry,
and Molecular Genetics, New Jersey Medical
School, Rutgers University, Newark, New Jersey 07103, United States
| | - Cristina R. Ventura
- Department
of Chemistry and Biochemistry, Seton Hall
University, South
Orange, New Jersey 07079, United States
| | - Gregory Wiedman
- Department
of Chemistry and Biochemistry, Seton Hall
University, South
Orange, New Jersey 07079, United States
| | - Chaoyang Xue
- Public
Health Research Institute and Department of Microbiology, Biochemistry,
and Molecular Genetics, New Jersey Medical
School, Rutgers University, Newark, New Jersey 07103, United States
| |
Collapse
|
18
|
Lima LF, Oliveira KBSD, Osiro KO, Cunha VA, Franco OL. Application of antimicrobial peptides in the poultry industry. Vet Microbiol 2024; 298:110267. [PMID: 39383680 DOI: 10.1016/j.vetmic.2024.110267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 08/22/2024] [Accepted: 09/24/2024] [Indexed: 10/11/2024]
Abstract
Poultry meat production and exportation contribute significantly to the global economy. However, various infections affect poultry production and consequently affect the economy. Nowadays, antibiotics are widely used in infection treatment and prevention. Antibiotic overuse is problematic because may cause antimicrobial resistance, which can be transferred to humans directly or indirectly, affecting public health. In addition, since antibiotics for animal growth stimulation are banned, it is important to search for new molecules to overcome these difficulties. As an alternative, antimicrobial peptides (AMPs) can show immunomodulatory, antimicrobial, and growth stimulation, which makes these molecules interesting as alternatives to antibiotic use. Studying AMPs can provide new ideas for treating the most important infections that affect poultry. Besides, this can assist in reducing the resistance problem. This review aims to examine recent studies about AMPs used against pathogens that can affect the poultry industry.
Collapse
Affiliation(s)
- Letícia Ferreira Lima
- Centro de Análises Proteômicas e Bioquímica (CAPB), Programa de Pós-Graduação Em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília (UCB), Brasília 70790-160, Brazil
| | - Kamila Botelho Sampaio de Oliveira
- Centro de Análises Proteômicas e Bioquímica (CAPB), Programa de Pós-Graduação Em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília (UCB), Brasília 70790-160, Brazil; S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco (UCDB), Campo Grande 79117-900, Brazil
| | - Karen Ofuji Osiro
- Centro de Análises Proteômicas e Bioquímica (CAPB), Programa de Pós-Graduação Em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília (UCB), Brasília 70790-160, Brazil
| | - Victor Albuquerque Cunha
- S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco (UCDB), Campo Grande 79117-900, Brazil
| | - Octávio Luiz Franco
- Centro de Análises Proteômicas e Bioquímica (CAPB), Programa de Pós-Graduação Em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília (UCB), Brasília 70790-160, Brazil; S-Inova Biotech, Programa de Pós-Graduação Em Biotecnologia, Universidade Católica Dom Bosco (UCDB), Campo Grande 79117-900, Brazil; Programa de Pós-Graduação Em Patologia Molecular, Universidade de Brasília (UnB), Brasília 70910-900, Brazil.
| |
Collapse
|
19
|
Ma X, Yang N, Mao R, Hao Y, Li Y, Guo Y, Teng D, Huang Y, Wang J. Self-assembly antimicrobial peptide for treatment of multidrug-resistant bacterial infection. J Nanobiotechnology 2024; 22:668. [PMID: 39478570 PMCID: PMC11526549 DOI: 10.1186/s12951-024-02896-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/02/2024] [Indexed: 11/02/2024] Open
Abstract
The wide-spreading of multidrug resistance poses a significant threat to human and animal health. Although antimicrobial peptides (AMPs) show great potential application, their instability has severely limited their clinical application. Here, self-assembled AMPs composed of multiple modules based on the principle of associating natural marine peptide N6 with ß-sheet-forming peptide were designed. It is noteworthy that one of the designed peptides, FFN could self-assemble into nanoparticles at 35.46 µM and achieve a dynamic transformation from nanoparticles to nanofibers in the presence of bacteria, resulting in a significant increase in stability in trypsin and tissues by 1.72-57.5 times compared to that of N6. Additionally, FFN exhibits a broad spectrum of antibacterial activity against multidrug-resistant (MDR) gram-positive (G+) and gram-negative (G-) bacteria with Minimum inhibitory concentrations (MICs) as low as 2 µM by membrane destruction and complemented by nanofiber capture. In vivo mouse mastitis infection model further confirmed the therapeutic potential and promising biosafety of the self-assembled peptide FFN, which can effectively alleviate mastitis caused by MDR Escherichia coli (E. coli) and Staphylococcus aureus (S. aureus), and eliminate pathogenic bacteria. In conclusion, the design of peptide-based nanomaterials presents a novel approach for the delivery and clinical translation of AMPs, promoting their application in medicine and animal husbandry.
Collapse
Affiliation(s)
- Xuanxuan Ma
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing, 100193, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China
| | - Na Yang
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China
| | - Ruoyu Mao
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China
| | - Ya Hao
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China
| | - Yuanyuan Li
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China
| | - Ying Guo
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China
| | - Da Teng
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China.
| | - Yinhua Huang
- State Key Laboratory of Farm Animal Biotech Breeding, College of Biology Sciences, China Agricultural University, Beijing, 100193, China.
| | - Jianhua Wang
- Innovative Team of Antimicrobial Peptides and Alternatives to Antibiotics, Gene Engineering Laboratory, Feed Research Institute, Chinese Academy of Agricultural Sciences, 12 Zhongguancun Nandajie St., Haidian District, Beijing, 100081, China.
- Key Laboratory of Feed Biotechnology, Ministry of Agriculture and Rural Affairs, Beijing, 100081, China.
| |
Collapse
|
20
|
Fang L, Yang T, Wang H, Cao J. Multiplex antimicrobial activities of the self-assembled amphiphilic polypeptide β nanofiber KF-5 against vaginal pathogens. Biol Direct 2024; 19:96. [PMID: 39438996 PMCID: PMC11495241 DOI: 10.1186/s13062-024-00546-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024] Open
Abstract
BACKGROUND Vaginal infections caused by multidrug-resistant pathogens such as Candida albicans and Gardnerella spp. represent a significant health challenge. Current treatments often fail because of resistance and toxicity. This study aimed to synthesize and characterize a novel amphiphilic polypeptide, KF-5, and evaluate its antibacterial and antifungal activities, biocompatibility, and potential mechanisms of action. RESULTS The KF-5 peptide was synthesized via solid-phase peptide synthesis and self-assembled into nanostructures with filamentous and hydrogel-like configurations. Characterization by scanning electron microscopy (SEM), transmission electron microscopy (TEM), and atomic force microscopy (AFM) confirmed the unique nanostructural properties of KF-5. KF-5 (125, 250, or 500 µg/ml) demonstrated potent antibacterial and antifungal activities, with significant inhibitory effects on drug-resistant Candida albicans and Gardnerella spp. (P < 0.05). In vitro assays revealed that 500 µg/ml KF-5 disrupted microbial cell membranes, increased membrane permeability, and induced lipid oxidation, leading to cell death (P < 0.05). Cytotoxicity tests revealed minimal toxicity in human vaginal epithelial cells, keratinocytes, and macrophages, with over 95% viability at high concentrations. Molecular dynamics simulations indicated that KF-5 interacts with phospholipid bilayers through electrostatic interactions, causing membrane disruption. In vivo studies using a mouse model of vaginal infection revealed that 0.5, 1, and 2 mg/ml KF-5 significantly reduced fungal burden and inflammation, and histological analysis confirmed the restoration of vaginal mucosal integrity (P < 0.01). Compared with conventional antifungal treatments such as miconazole, KF-5 exhibited superior efficacy (P < 0.01). CONCLUSIONS KF-5 demonstrates significant potential as a safe and effective antimicrobial agent for treating vaginal infections. Its ability to disrupt microbial membranes while maintaining biocompatibility with human cells highlights its potential for clinical application. These findings provide a foundation for further development of KF-5 as a therapeutic option for combating drug-resistant infections.
Collapse
Affiliation(s)
- Ling Fang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Xishan People's Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, 214105, Jiangsu, China
- Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, Jiangsu, China
| | - Tiancheng Yang
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China
- Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, Jiangsu, China
| | - Haojue Wang
- Xishan People's Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Southeast University, Wuxi, 214105, Jiangsu, China.
| | - Jun Cao
- Nanjing Medical University, Nanjing, 211166, Jiangsu, China.
- Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, Jiangsu, China.
| |
Collapse
|
21
|
Tang Z, Deng L, Zhang J, Jiang T, Xiang H, Chen Y, Liu H, Cai Z, Cui W, Xiong Y. Intelligent Hydrogel-Assisted Hepatocellular Carcinoma Therapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0477. [PMID: 39691767 PMCID: PMC11651419 DOI: 10.34133/research.0477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 12/19/2024]
Abstract
Given the high malignancy of liver cancer and the liver's unique role in immune and metabolic regulation, current treatments have limited efficacy, resulting in a poor prognosis. Hydrogels, soft 3-dimensional network materials comprising numerous hydrophilic monomers, have considerable potential as intelligent drug delivery systems for liver cancer treatment. The advantages of hydrogels include their versatile delivery modalities, precision targeting, intelligent stimulus response, controlled drug release, high drug loading capacity, excellent slow-release capabilities, and substantial potential as carriers of bioactive molecules. This review presents an in-depth examination of hydrogel-assisted advanced therapies for hepatocellular carcinoma, encompassing small-molecule drug therapy, immunotherapy, gene therapy, and the utilization of other biologics. Furthermore, it examines the integration of hydrogels with conventional liver cancer therapies, including radiation, interventional therapy, and ultrasound. This review provides a comprehensive overview of the numerous advantages of hydrogels and their potential to enhance therapeutic efficacy, targeting, and drug delivery safety. In conclusion, this review addresses the clinical implementation of hydrogels in liver cancer therapy and future challenges and design principles for hydrogel-based systems, and proposes novel research directions and strategies.
Collapse
Affiliation(s)
- Zixiang Tang
- Department of Hepatobiliary Surgery, Academician (Expert) Workstation, Sichuan Digestive System Disease Clinical Medical Research Center,
Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, P. R. China
| | - Lin Deng
- Department of Clinical Medicine,
North Sichuan Medical College, Nanchong 637000, P. R. China
| | - Jing Zhang
- Department of Gastroenterology,
Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, P. R. China
| | - Tao Jiang
- Department of Hepatobiliary Surgery, Academician (Expert) Workstation, Sichuan Digestive System Disease Clinical Medical Research Center,
Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, P. R. China
| | - Honglin Xiang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanyang Chen
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Huzhe Liu
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Zhengwei Cai
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai 200025, P. R. China
| | - Yongfu Xiong
- Department of Hepatobiliary Surgery, Academician (Expert) Workstation, Sichuan Digestive System Disease Clinical Medical Research Center,
Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, P. R. China
| |
Collapse
|
22
|
Mitra S, Chen MT, Stedman F, Hernandez J, Kumble G, Kang X, Zhang C, Tang G, Daugherty I, Liu W, Ocloo J, Klucznik KR, Li AA, Heinrich F, Deslouches B, Tristram-Nagle S. How Unnatural Amino Acids in Antimicrobial Peptides Change Interactions with Lipid Model Membranes. J Phys Chem B 2024; 128:9772-9784. [PMID: 39328031 PMCID: PMC11472314 DOI: 10.1021/acs.jpcb.4c04152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/05/2024] [Accepted: 09/12/2024] [Indexed: 09/28/2024]
Abstract
This study investigates the potential of antimicrobial peptides (AMPs) as alternatives to combat antibiotic resistance, with a focus on two AMPs containing unnatural amino acids (UAAs), E2-53R (16 AAs) and LE-54R (14 AAs). In both peptides, valine is replaced by norvaline (Nva), and tryptophan is replaced by 1,2,3,4-tetrahydroisoquinoline-3-carboxylic acid (Tic). Microbiological studies reveal their potent activity against both Gram-negative (G(-)) and Gram-positive (G(+)) bacteria without any toxicity to eukaryotic cells at test concentrations up to 32 μM. Circular dichroism (CD) spectroscopy indicates that these peptides maintain α-helical structures when interacting with G(-) and G(+) lipid model membranes (LMMs), a feature linked to their efficacy. X-ray diffuse scattering (XDS) demonstrates a softening of G(-), G(+) and eukaryotic (Euk33) LMMs and a nonmonotonic decrease in chain order as a potential determinant for bacterial membrane destabilization. Additionally, XDS finds a significant link between both peptides' interfacial location in G(-) and G(+) LMMs and their efficacy. Neutron reflectometry (NR) confirms the AMP locations determined using XDS. Lack of toxicity in eukaryotic cells may be related to their loss of α-helicity and their hydrocarbon location in Euk33 LMMs. Both AMPs with UAAs offer a novel strategy to wipe out antibiotic-resistant strains while maintaining human cells. These findings are compared with previously published data on E2-35, which consists of the natural amino acids arginine, tryptophan, and valine.
Collapse
Affiliation(s)
- Saheli Mitra
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Mei-Tung Chen
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Francisca Stedman
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Jedidiah Hernandez
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Grace Kumble
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Xi Kang
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Churan Zhang
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Grace Tang
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Ian Daugherty
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Wanqing Liu
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Jeremy Ocloo
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Kevin Raphael Klucznik
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Alexander Anzhi Li
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Frank Heinrich
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
- Center
for Neutron Research, National Institute
of Standards and Technology, Gaithersburg, Maryland 20899, United States
| | - Berthony Deslouches
- Department
of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, Pennsylvania 15261, United States
| | - Stephanie Tristram-Nagle
- Biological
Physics Group, Physics Department, Carnegie
Mellon University, Pittsburgh, Pennsylvania 15213, United States
| |
Collapse
|
23
|
Zare-Zardini H, Saberian E, Jenča A, Ghanipour-Meybodi R, Jenča A, Petrášová A, Jenčová J. From defense to offense: antimicrobial peptides as promising therapeutics for cancer. Front Oncol 2024; 14:1463088. [PMID: 39445062 PMCID: PMC11496142 DOI: 10.3389/fonc.2024.1463088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Accepted: 09/09/2024] [Indexed: 10/25/2024] Open
Abstract
Antimicrobial peptides (AMPs), naturally occurring components of innate immunity, are emerging as a promising new class of anticancer agents. This review explores the potential of AMPs as a novel class of anticancer agents. AMPs, naturally occurring peptides with broad-spectrum antimicrobial activity, exhibit several characteristics that make them attractive candidates for cancer therapy, including selectivity for cancer cells, broad-spectrum activity, and immunomodulatory effects. Analysis of a dataset of AMPs with anticancer activity reveals that their effectiveness is influenced by various structural properties, including net charge, length, Boman index, and hydrophobicity. These properties contribute to their ability to target and disrupt cancer cell membranes, interfere with intracellular processes, and modulate the immune response. The review highlights the promising potential of AMPs as a new frontier in cancer treatment, offering hope for more effective and less toxic therapies. AMPs demonstrate promising potential in cancer therapy through multiple mechanisms, including direct cytotoxicity, immune response modulation, and targeting of the tumor microenvironment, as evidenced by extensive preclinical studies in animal models showing tumor regression, metastasis inhibition, and improved survival rates. AMPs show significant potential as cancer therapeutics through their direct cytotoxicity, immune response modulation, and tumor microenvironment targeting, with promising results from preclinical studies and early-phase clinical trials. Future research should focus on optimizing AMP properties, developing novel delivery strategies, and exploring synergistic combination therapies to fully realize their potential as effective cancer treatments, while addressing challenges related to stability, delivery, and potential toxicity.
Collapse
Affiliation(s)
- Hadi Zare-Zardini
- Department of Biomedical Engineering, Meybod University, Meybod, Iran
| | - Elham Saberian
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | | | - Andrej Jenča
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Adriána Petrášová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| | - Janka Jenčová
- Klinika of Stomatology and Maxillofacial Surgery Akadémia Košice Bacikova, Pavel Jozef Šafárik University (UPJS LF), Kosice, Slovakia
| |
Collapse
|
24
|
Phuong HBT, Ngan HD, Thi HP, Thanh BNT, Dang TT, Ho TNT, Thanh TT, Hong MN, Xuan HL. Dual Antimicrobial and Anticancer Activity of Membrane-Active Peptide BP52. Protein J 2024; 43:1025-1034. [PMID: 39190120 DOI: 10.1007/s10930-024-10231-y] [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] [Accepted: 08/21/2024] [Indexed: 08/28/2024]
Abstract
The linear undecapeptide BP52 was previously reported to have antibacterial activity against phytopathogenic bacteria species. Due to the structural similarities to naturally occurring cationic helical antimicrobial peptides, it was speculated that this peptide could potentially target microbial pathogens and cancer cells found in mammals. Consequently, this study aims to further investigate the structural and biological properties of this peptide. Our findings indicate that BP52 exhibits strong antimicrobial and anticancer activity while displaying relatively low levels of hemolytic activity. Hence, this study suggests that BP52 could be a potential lead compound for drug discovery against infectious diseases and cancer. Besides, new insights into the relationships between the structure and the multifunctional properties of antimicrobial peptides were also explored.
Collapse
Affiliation(s)
- Hai Bui Thi Phuong
- Faculty of Pharmacy, Phenikaa University, Hanoi, 12116, Vietnam
- Faculty of Biotechnology, Chemistry and Environmental Engineering, Phenikaa University, Hanoi, 12116, Vietnam
| | - Hoa Doan Ngan
- Faculty of Medical Technology, Phenikaa University, Hanoi, 12116, Vietnam
| | - Hue Pham Thi
- Bioresource Research Center, Phenikaa University, Hanoi, 12116, Vietnam
| | | | - Tien T Dang
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1B TL29, District 12, Ho Chi Minh City, Vietnam
| | - Thao N T Ho
- VN-UK Institute for Research and Executive Education, The University of Danang, Danang, 550000, Vietnam
| | | | - Minh Nguyen Hong
- Faculty of Biotechnology, Chemistry and Environmental Engineering, Phenikaa University, Hanoi, 12116, Vietnam
- Bioresource Research Center, Phenikaa University, Hanoi, 12116, Vietnam
| | - Huy Luong Xuan
- Faculty of Pharmacy, Phenikaa University, Hanoi, 12116, Vietnam.
| |
Collapse
|
25
|
Ratrey P, Bhattacharya S, Coffey L, Thompson D, Hudson SP. Solid lipid nanoparticle formulation maximizes membrane-damaging efficiency of antimicrobial nisin Z peptide. Colloids Surf B Biointerfaces 2024; 245:114255. [PMID: 39303385 DOI: 10.1016/j.colsurfb.2024.114255] [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: 06/13/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Solid lipid nanoparticles (SLNs) can protect and deliver naturally derived or synthetic biologically active products to target sites in vivo. Here, an SLN formulation produces a measured four-fold reduction in inhibitory concentration of an antimicrobial peptide nisin Z against S. aureus as compared to the free peptide, indicating the successful delivery and enhanced effectiveness of the SLN-encapsulated bacteriocin. Spherical SLNs of size 79.47 ± 2.01 nm and zeta potential of -9.8 ± 0.3 mV were synthesised. The lipid formulation maximizes the membrane-damaging mode of action of the free peptide with more and larger-sized pores formed on bacterial membranes treated with nisin Z SLNs as measured from scanning electron microscopy and transmission electron microscopy. Flow cytometry measurements precisely quantified an enhanced dye leakage from pre-labeled bacterial cells when treated with nisin Z-loaded SLNs compared to free peptide. The lipid formulation accelerated cell death by killing all the cells within half an hour compared to the equivalent concentration of free peptide which was not bactericidal. Molecular dynamics simulations revealed a mechanism of SLN facilitated binding to the lipid II bacterial cell wall precursor via enhanced adsorption of nisin Z at the inner bacterial cell membrane bilayer. These findings confirmed the potential of SLN formulations for the effective delivery of therapeutic peptides for next-generation antibiotics that are active at low concentrations with the potential to mitigate antimicrobial resistance.
Collapse
Affiliation(s)
- Poonam Ratrey
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Shayon Bhattacharya
- Department of Physics, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Laura Coffey
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Damien Thompson
- Department of Physics, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| | - Sarah P Hudson
- Department of Chemical Sciences, SSPC the SFI Research Centre for Pharmaceuticals, Bernal Institute, University of Limerick, Limerick V94 T9PX, Ireland.
| |
Collapse
|
26
|
Luo R, Xu H, Lin Q, Chi J, Liu T, Jin B, Ou J, Xu Z, Peng T, Quan G, Lu C. Emerging Trends in Dissolving-Microneedle Technology for Antimicrobial Skin-Infection Therapies. Pharmaceutics 2024; 16:1188. [PMID: 39339224 PMCID: PMC11435303 DOI: 10.3390/pharmaceutics16091188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 08/26/2024] [Accepted: 09/05/2024] [Indexed: 09/30/2024] Open
Abstract
Skin and soft-tissue infections require significant consideration because of their prolonged treatment duration and propensity to rapidly progress, resulting in severe complications. The primary challenge in their treatment stems from the involvement of drug-resistant microorganisms that can form impermeable biofilms, as well as the possibility of infection extending deep into tissues, thereby complicating drug delivery. Dissolving microneedle patches are an innovative transdermal drug-delivery system that effectively enhances drug penetration through the stratum corneum barrier, thereby increasing drug concentration at the site of infection. They offer highly efficient, safe, and patient-friendly alternatives to conventional topical formulations. This comprehensive review focuses on recent advances and emerging trends in dissolving-microneedle technology for antimicrobial skin-infection therapy. Conventional antibiotic microneedles are compared with those based on emerging antimicrobial agents, such as quorum-sensing inhibitors, antimicrobial peptides, and antimicrobial-matrix materials. The review also highlights the potential of innovative microneedles incorporating chemodynamic, nanoenzyme antimicrobial, photodynamic, and photothermal antibacterial therapies. This review explores the advantages of various antimicrobial therapies and emphasizes the potential of their combined application to improve the efficacy of microneedles. Finally, this review analyzes the druggability of different antimicrobial microneedles and discusses possible future developments.
Collapse
Affiliation(s)
- Rui Luo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Huihui Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Qiaoni Lin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Jiaying Chi
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Tingzhi Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Bingrui Jin
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Jiayu Ou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Zejun Xu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Tingting Peng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Guilan Quan
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
| | - Chao Lu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Department of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Pharmacy, College of Pharmacy, Jinan University, Guangzhou 511436, China
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA
| |
Collapse
|
27
|
Xu H, Che Y, Zhou R, Wang L, Huang J, Kong W, Liu C, Guo L, Tang Y, Wang X, Yang X, Wang E, Xu C. Research progress of natural polysaccharide-based and natural protein-based hydrogels for bacteria-infected wound healing. CHEMICAL ENGINEERING JOURNAL 2024; 496:153803. [DOI: 10.1016/j.cej.2024.153803] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
28
|
Stepanova M, Levit M, Egorova T, Nashchekina Y, Sall T, Demyanova E, Guryanov I, Korzhikova-Vlakh E. Poly(2-Deoxy-2-Methacrylamido-D-Glucose)-Based Complex Conjugates of Colistin, Deferoxamine and Vitamin B12: Synthesis and Biological Evaluation. Pharmaceutics 2024; 16:1080. [PMID: 39204425 PMCID: PMC11359296 DOI: 10.3390/pharmaceutics16081080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024] Open
Abstract
Growing resistance to traditional antibiotics poses a global threat to public health. In this regard, modification of known antibiotics, but with limited applications due to side effects, is one of the extremely promising approaches at present. In this study, we proposed the synthesis of novel complex polymeric conjugates of the peptide antibiotic colistin (CT). A biocompatible and water-soluble synthetic glycopolymer, namely, poly(2-deoxy-2-methacrylamido-D-glucose) (PMAG), was used as a polymer carrier. In addition to monoconjugates containing CT linked to PMAG by hydrolyzable and stable bonds, a set of complex conjugates also containing the siderophore deferoxamine (DFOA) and vitamin B12 was developed. The structures of the conjugates were confirmed by 1H NMR and FTIR-spectroscopy, while the compositions of conjugates were determined by UV-Vis spectrophotometry and HPLC analysis. The buffer media with pH 7.4, corresponding to blood or ileum pH, and 5.2, corresponding to the intestinal pH after ingestion or pH in the focus of inflammation, were used to study the release of CT. The resulting conjugates were examined for cytotoxicity and antimicrobial activity. All conjugates showed less cytotoxicity than free colistin. A Caco-2 cell permeability assay was carried out for complex conjugates to simulate the drug absorption in the intestine. In contrast to free CT, which showed very low permeability through the Caco-2 monolayer, the complex polymeric conjugates of vitamin B12 and CT provided significant transport. The antimicrobial activity of the conjugates depended on the conjugate composition. It was found that conjugates containing CT linked to the polymer by a hydrolyzable bond were found to be more active than conjugates with a non-hydrolyzable bond between CT and PMAG. Conjugates containing DFOA complexed with Fe3+ were characterized by enhanced antimicrobial activity against Pseudomonas aeruginosa compared to other conjugates.
Collapse
Affiliation(s)
- Mariia Stepanova
- Institute of Macromolecular Compounds of Russian Academy of Sciences, 199004 St. Petersburg, Russia; (M.S.); (M.L.)
| | - Mariia Levit
- Institute of Macromolecular Compounds of Russian Academy of Sciences, 199004 St. Petersburg, Russia; (M.S.); (M.L.)
| | - Tatiana Egorova
- Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.E.); (E.D.)
| | - Yulia Nashchekina
- Institute of Cytology of Russian Academy of Sciences, 194064 St. Petersburg, Russia;
| | - Tatiana Sall
- Institute of Experimental Medicine, 197022 St. Petersburg, Russia;
| | - Elena Demyanova
- Institute of Highly Pure Biopreparations, 197110 St. Petersburg, Russia; (T.E.); (E.D.)
| | - Ivan Guryanov
- Institute of Chemistry, St. Petersburg State University, 198504 St. Petersburg, Russia
| | - Evgenia Korzhikova-Vlakh
- Institute of Macromolecular Compounds of Russian Academy of Sciences, 199004 St. Petersburg, Russia; (M.S.); (M.L.)
| |
Collapse
|
29
|
Zhang Y, Liu LH, Xu B, Zhang Z, Yang M, He Y, Chen J, Zhang Y, Hu Y, Chen X, Sun Z, Ge Q, Wu S, Lei W, Li K, Cui H, Yang G, Zhao X, Wang M, Xia J, Cao Z, Jiang A, Wu YR. Screening antimicrobial peptides and probiotics using multiple deep learning and directed evolution strategies. Acta Pharm Sin B 2024; 14:3476-3492. [PMID: 39234615 PMCID: PMC11372459 DOI: 10.1016/j.apsb.2024.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 03/25/2024] [Accepted: 05/06/2024] [Indexed: 09/06/2024] Open
Abstract
Owing to their limited accuracy and narrow applicability, current antimicrobial peptide (AMP) prediction models face obstacles in industrial application. To address these limitations, we developed and improved an AMP prediction model using Comparing and Optimizing Multiple DEep Learning (COMDEL) algorithms, coupled with high-throughput AMP screening method, finally reaching an accuracy of 94.8% in test and 88% in experiment verification, surpassing other state-of-the-art models. In conjunction with COMDEL, we employed the phage-assisted evolution method to screen Sortase in vivo and developed a cell-free AMP synthesis system in vitro, ultimately increasing AMPs yields to a range of 0.5-2.1 g/L within hours. Moreover, by multi-omics analysis using COMDEL, we identified Lactobacillus plantarum as the most promising candidate for AMP generation among 35 edible probiotics. Following this, we developed a microdroplet sorting approach and successfully screened three L. plantarum mutants, each showing a twofold increase in antimicrobial ability, underscoring their substantial industrial application values.
Collapse
Affiliation(s)
- Yu Zhang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Li-Hua Liu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
- Biology Department and Institute of Marine Sciences, College of Science, Shantou University, Shantou 515063, China
| | - Bo Xu
- School of Basic Medical Sciences, Hubei University of Science and Technology, Xianning 437100, China
| | - Zhiqian Zhang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Min Yang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Yiyang He
- School of Education, Jianghan University, Wuhan 430056, China
| | - Jingjing Chen
- Yeasen Biotechnology (Shanghai) Co., Ltd., Shanghai 200000, China
| | - Yang Zhang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Yucheng Hu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Xipeng Chen
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Zitong Sun
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Qijun Ge
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Song Wu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Wei Lei
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Kaizheng Li
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Hua Cui
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Gangzhu Yang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Xuemei Zhao
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Man Wang
- Yeasen Biotechnology (Shanghai) Co., Ltd., Shanghai 200000, China
| | - Jiaqi Xia
- School of Basic Medicine, Jiamusi University, Jiamusi 154000, China
| | - Zhen Cao
- Yeasen Biotechnology (Shanghai) Co., Ltd., Shanghai 200000, China
| | - Ao Jiang
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| | - Yi-Rui Wu
- Tidetron Bioworks Technology (Guangzhou) Co., Ltd., Guangzhou Qianxiang Bioworks Co., Ltd., Guangzhou 510000, China
| |
Collapse
|
30
|
Li X, Zhang L, Liu Z, Wang R, Jiao T. Recent progress in hydrogels combined with phototherapy for bacterial infection: A review. Int J Biol Macromol 2024; 274:133375. [PMID: 38914386 DOI: 10.1016/j.ijbiomac.2024.133375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 06/17/2024] [Accepted: 06/21/2024] [Indexed: 06/26/2024]
Abstract
Phototherapy has become one of the most effective antibacterial methods due to its associated lack of drug resistance and its good antibacterial effect. For the purpose of avoiding the aggregation and premature release of photosensitive/photothermal agents during phototherapy, they can be mixed into three-dimensional hydrogels. The combination of hydrogels and phototherapy combines the merits of both hydrogels and phototherapy, overcomes the disadvantages of traditional antibacterial methodologies, and has broad application prospects. This review presents recent advancements in phototherapeutic antibacterial hydrogels including photodynamic antibacterial hydrogels, photothermal antibacterial hydrogels, photodynamic and photothermal synergistic antibacterial hydrogels, and other synergistic antibacterial hydrogels involving phototherapy.
Collapse
Affiliation(s)
- Xinyu Li
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, PR China
| | - Lexin Zhang
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, PR China
| | - Zhiwei Liu
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, PR China.
| | - Ran Wang
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, PR China.
| | - Tifeng Jiao
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, PR China.
| |
Collapse
|
31
|
Fathi F, Alizadeh B, Tabarzad MV, Tabarzad M. Important structural features of antimicrobial peptides towards specific activity: Trends in the development of efficient therapeutics. Bioorg Chem 2024; 149:107524. [PMID: 38850782 DOI: 10.1016/j.bioorg.2024.107524] [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: 12/18/2023] [Revised: 04/29/2024] [Accepted: 06/02/2024] [Indexed: 06/10/2024]
Abstract
Proteins and peptides, as polypeptide chains, have usually got unique conformational structures for effective biological activity. Antimicrobial peptides (AMPs) are a group of bioactive peptides, which have been increasingly studied during recent years for their promising antibacterial, antifungal, antiviral and anti-inflammatory activity, as well as, other esteemed bioactivities. Numerous AMPs have been separated from a wide range of natural resources, or produced in vitro through chemical synthesis and recombinant protein expression. Natural AMPs have had limited clinical application due to several drawbacks, such as their short half-life due to protease degradation, lack of activity at physiological salt concentrations, toxicity to mammalian cells, and the absence of suitable methods of delivery for the AMPs that are targeted and sustained. The creation of synthetic analogs of AMPs would both avoid the drawbacks of the natural analogs and maintain or even increase the antimicrobial effectiveness. The structure-activity relationship of discovered AMPs or their derivatives facilitates the development of synthetic AMPs. This review discovered that the relationship between the activity of AMPs and their positive net charge, hydrophobicity, and amino acid sequence and the relationship between AMPs' function and other features like their topology, glycosylation, and halogenation.
Collapse
Affiliation(s)
- Fariba Fathi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Bahareh Alizadeh
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Vahid Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
32
|
Wang X, Wang Q, Wang J, Wang X, Yin L, Wang C, Fan G, Pan J. A Novel Lipopeptide-Functionalized Metal-Organic Framework for Periodontitis Therapy through the Htra1/FAK/YAP Pathway. Biomater Res 2024; 28:0057. [PMID: 39076893 PMCID: PMC11283871 DOI: 10.34133/bmr.0057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 06/20/2024] [Indexed: 07/31/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease characterized by plaque accumulation, resulting in immune microenvironment disorders and resorption of alveolar bone. To promote bone healing under inflammatory environments, a functional biomaterial based on disease pathophysiology is designed. A novel fatty acid C10-modified polypeptide, C10-KR8, is discovered to have excellent abilities in modulating macrophage repolarization and promoting bone regeneration in periodontitis. To build a multifunctional material localized drug delivery system, C10-KR8@ZIF-8 (C10-KR8-loaded zeolitic imidazolate framework-8) nanoparticles are constructed to sustainedly release the C10-KR8 peptide and Zn elements. By synergistic effects of providing a dynamic immuno-modulatory environment and promoting osteogenesis under pathological conditions, the obtained pH-responsive nanoparticles display excellent bone regeneration capability. Furthermore, coimmunoprecipitation/liquid chromatography-tandem mass spectrometry analysis and proteomics analysis revealed that the C10-KR8 peptide directly interacts with the high-temperature requirement protein A1 (Htra1), and C10-KR8@ZIF-8 nanoparticles promote the osteogenic differentiation of bone mesenchymal stem cells by activating the focal adhesion kinase (FAK)/phosphatidylinositide 3-kinase (PI3K)/AKT pathway and enhancing the nuclear localization of Yes-associated protein (YAP). Taken together, this study demonstrates C10-KR8 peptide regulate osteoimmunology and bone regeneration by Htra1/FAK/YAP pathway and that ZIF-8-based peptide loading platform is a promising strategy for periodontitis.
Collapse
Affiliation(s)
- Xuechun Wang
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Qing Wang
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Jian Wang
- Department of General Dentistry, Shanghai Ninth People’s Hospital, School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology,
Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, Shanghai, 200011, China
| | - Xuan Wang
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Linling Yin
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| | - Changping Wang
- Department of Orthopaedics, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Guangjian Fan
- Precision Research Center for Refractory Diseases, Institute for Clinical Research, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jinsong Pan
- Department of Stomatology, Shanghai General Hospital,
Shanghai Jiao Tong University School of Medicine, Shanghai, 201620, China
| |
Collapse
|
33
|
Li C, Cai Y, Luo L, Tian G, Wang X, Yan A, Wang L, Wu S, Wu Z, Zhang T, Chen W, Zhang Z. TC-14, a cathelicidin-derived antimicrobial peptide with broad-spectrum antibacterial activity and high safety profile. iScience 2024; 27:110404. [PMID: 39092176 PMCID: PMC11292558 DOI: 10.1016/j.isci.2024.110404] [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: 03/27/2024] [Revised: 05/27/2024] [Accepted: 06/26/2024] [Indexed: 08/04/2024] Open
Abstract
Cathelicidins, a major class of antimicrobial peptides (AMPs), hold considerable potential for antimicrobial drug development. In the present study, we identified a novel cathelicidin AMP (TC-33) derived from the Chinese tree shrew. Despite TC-33 demonstrating weak antimicrobial activity, the novel peptide TC-14, developed based on its active region, exhibited a 432-fold increase in antimicrobial activity over the parent peptide. Structural analysis revealed that TC-14 adopted an amphipathic α-helical conformation. The bactericidal mechanism of TC-14 involved targeting and disrupting the bacterial membrane, leading to rapid membrane permeabilization and rupture. Furthermore, TC-14 exhibited a high-safety profile, as evidenced by the absence of cytotoxic and hemolytic activities, as well as high biocompatibility and safety in vivo. Of note, its potent antimicrobial activity provided significant protection in a murine model of skin infection. Overall, this study presents TC-14 as a promising drug candidate for antimicrobial drug development.
Collapse
Affiliation(s)
- Chenxi Li
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, Yunnan, China
| | - Ying Cai
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Lin Luo
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, Yunnan, China
| | - Gengzhou Tian
- Department of Breast Surgery, First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650032, China
| | - Xingyu Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - An Yan
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Liunan Wang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Sijing Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
- School of Life Sciences, Yunnan University, Kunming 650500, China
| | - Zhongxiang Wu
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Tianyu Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| | - Wenlin Chen
- Third Department of Breast Surgery, Peking University Cancer Hospital Yunnan, Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital), Kunming 650118, Yunnan, China
| | - Zhiye Zhang
- Institute of Medical Biology, Chinese Academy of Medical Sciences & Peking Union Medical College, Kunming 650031, Yunnan, China
| |
Collapse
|
34
|
Zhu J, Zhang H, Lan H, Bi B, Peng X, Li D, Wang H, Zhu K, Shao F, Yin M. Enhancing breast cancer treatment: mesoporous dopamine nanoparticles in synergy with chrysin for photothermal therapy. Front Oncol 2024; 14:1427858. [PMID: 39045563 PMCID: PMC11263883 DOI: 10.3389/fonc.2024.1427858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Accepted: 06/19/2024] [Indexed: 07/25/2024] Open
Abstract
Introduction Breast cancer is one of the most prevalent cancers, primarily affecting women. Among its subtypes, estrogen receptor-positive (ER+) breast cancer is particularly common. Inhibiting estrogen's effects is crucial for treating ER+ breast cancer, but current therapies often have significant side effects and limitations. Chrysin, a natural flavonoid, has shown potential in reducing estrogen receptor expression, but its poor water solubility hampers clinical application. This study explores the use of mesoporous dopamine nanoparticles (mPDA) to enhance the delivery and efficacy of Chrysin, combined with photothermal therapy (PTT), for breast cancer treatment. Methods Chrysin-loaded mPDA nanoparticles (Chrysin@mPDA) were synthesized and characterized for their morphology, drug-loading efficiency, stability, and photothermal properties. Network pharmacology was used to predict Chrysin's mechanisms in breast cancer, which were validated through gene expression analysis in cell experiments. The therapeutic efficacy of Chrysin@mPDA with and without PTT was evaluated in a mouse model of breast cancer, with tumor volume and weight measured. Immunohistochemical analysis was conducted to assess estrogen receptor expression and immune cell infiltration in tumor tissues. Results Chrysin@mPDA nanoparticles demonstrated a high drug-loading capacity and excellent stability. Photothermal studies confirmed the nanoparticles' ability to generate heat upon laser exposure, significantly enhancing Chrysin release in acidic conditions with laser irradiation. Network pharmacology identified key target genes affected by Chrysin, including ESR1, BRCA1, CTNNB1, and BAX, which were validated through qPCR. In vivo, the combination of Chrysin@mPDA and PTT significantly reduced tumor volume and weight, decreased estrogen receptor-positive cells, and increased infiltration of CD3+CD4+ and CD3+CD8+ T cells in tumor tissues. Discussion The study highlights the potential of Chrysin-loaded mPDA nanoparticles combined with PTT as an effective strategy for breast cancer treatment. This approach addresses the limitations of Chrysin's solubility and enhances its therapeutic efficacy through synergistic mechanisms. The dual action of Chrysin in modulating gene expression and PTT in inducing localized hyperthermia and immune response suggests a promising avenue for improved breast cancer prognosis and reduced recurrence.
Collapse
Affiliation(s)
- Jing Zhu
- Health Management Center, Zigong First People’s Hospital, Zigong, China
| | - Heng Zhang
- Medical Imaging Center, Dazhou Central Hospital, Dazhou, China
- Center for Precision Health, School of Medical and Health Science, Edith Cowan University, Perth, WA, Australia
| | - Haomiao Lan
- Department of Thyroid and Breast Surgery, Zigong First People’s Hospital, Zigong, China
| | - Bing Bi
- Health Management Center, Zigong First People’s Hospital, Zigong, China
| | - Xianfeng Peng
- Department of Nuclear Medicine, Zigong First People’s Hospital, Zigong, China
| | - Dandan Li
- Department of Nuclear Medicine, Zigong First People’s Hospital, Zigong, China
| | - Haili Wang
- Department of Nuclear Medicine, Zigong First People’s Hospital, Zigong, China
| | - Ke Zhu
- Department of Nuclear Medicine, Zigong First People’s Hospital, Zigong, China
| | - Fuqiang Shao
- Department of Nuclear Medicine, Zigong First People’s Hospital, Zigong, China
| | - Minggang Yin
- Department of Clinical Laboratory, Zigong First People’s Hospital, Zigong, China
| |
Collapse
|
35
|
Xie Z, Jiang W, Liu H, Chen L, Xuan C, Wang Z, Shi X, Lin Z, Gao X. Antimicrobial Peptide- and Dentin Matrix-Functionalized Hydrogel for Vital Pulp Therapy via Synergistic Bacteriostasis, Immunomodulation, and Dentinogenesis. Adv Healthc Mater 2024; 13:e2303709. [PMID: 38431770 DOI: 10.1002/adhm.202303709] [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: 10/25/2023] [Revised: 02/20/2024] [Indexed: 03/05/2024]
Abstract
The preservation of vital pulps is crucial for maintaining the physiological functions of teeth; however, vital pulp therapy (VPT) of pulpitis teeth remains a substantial challenge due to uncontrolled infection, excessive inflammation, and limited regenerative potential. Current pulp capping agents have restricted effects in the infectious and inflammatory microenvironment. To address this, a multifunctional hydrogel (TGH/DM) with antibacterial, immunomodulatory, and mineralization-promoting effects is designed. The antimicrobial peptide (AMP) and demineralized dentin matrix are incorporated into the hydrogel, achieving sustainable delivery of AMP and a cocktail of growth factors. In vitro results show that TGH/DM could kill endodontic microbiota, ameliorate inflammatory responses of human dental pulp stem cells (hDPSCs), and prompt odontogenic differentiation of inflammatory hDPSCs via activation of peroxisome proliferator-activated receptor gamma. In vivo results suggest that TGH/DM is capable of inducing M2 phenotype transformation of macrophages in mice and fostering the regeneration of the dentin-pulp complex in inflamed pulps of beagle dogs. Overall, this study first proposes the synergistic regulation of AMP and tissue-specific extracellular matrix for the treatment of pulpitis, and the advanced hydrogel provides a facile and effective way for VPT.
Collapse
Affiliation(s)
- Zhuo Xie
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Wentao Jiang
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Hui Liu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Lingling Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Chengkai Xuan
- School of Biomedical Science and Engineering, National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zhenxing Wang
- School of Biomedical Science and Engineering, National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Xuetao Shi
- School of Biomedical Science and Engineering, National Engineering Research Centre for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, P. R. China
| | - Zhengmei Lin
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| | - Xianling Gao
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, 510055, P. R. China
| |
Collapse
|
36
|
van der Walt M, Möller DS, van Wyk RJ, Ferguson PM, Hind CK, Clifford M, Do Carmo Silva P, Sutton JM, Mason AJ, Bester MJ, Gaspar ARM. QSAR Reveals Decreased Lipophilicity of Polar Residues Determines the Selectivity of Antimicrobial Peptide Activity. ACS OMEGA 2024; 9:26030-26049. [PMID: 38911757 PMCID: PMC11191095 DOI: 10.1021/acsomega.4c01277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/15/2024] [Accepted: 05/20/2024] [Indexed: 06/25/2024]
Abstract
Antimicrobial resistance has increased rapidly, causing daunting morbidity and mortality rates worldwide. Antimicrobial peptides (AMPs) have emerged as promising alternatives to traditional antibiotics due to their broad range of targets and low tendency to elicit resistance. However, potent antimicrobial activity is often accompanied by excessive cytotoxicity toward host cells, leading to a halt in AMP therapeutic development. Here, we present multivariate analyses that correlate 28 peptide properties to the activity and toxicity of 46 diverse African-derived AMPs and identify the negative lipophilicity of polar residues as an essential physiochemical property for selective antimicrobial activity. Twenty-seven active AMPs are identified, of which the majority are of scorpion or frog origin. Of these, thirteen are novel with no previously reported activities. Principal component analysis and quantitative structure-activity relationships (QSAR) reveal that overall hydrophobicity, lipophilicity, and residue side chain surface area affect the antimicrobial and cytotoxic activity of an AMP. This has been well documented previously, but the present QSAR analysis additionally reveals that a decrease in the lipophilicity, contributed by those amino acids classified as polar, confers selectivity for a peptide to pathogen over mammalian cells. Furthermore, an increase in overall peptide charge aids selectivity toward Gram-negative bacteria and fungi, while selectivity toward Gram-positive bacteria is obtained through an increased number of small lipophilic residues. Finally, a conservative increase in peptide size in terms of sequence length and molecular weight also contributes to improved activity without affecting toxicity. Our findings suggest a novel approach for the rational design or modification of existing AMPs to increase pathogen selectivity and enhance therapeutic potential.
Collapse
Affiliation(s)
- Mandelie van der Walt
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Dalton S. Möller
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Rosalind J. van Wyk
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Philip M. Ferguson
- Institute
of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Charlotte K. Hind
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - Melanie Clifford
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - Phoebe Do Carmo Silva
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - J. Mark Sutton
- Antimicrobial
Discovery Development and Diagnostics, Vaccine Evaluation and Development
Centre, UK Health Security Agency, Salisbury SP4 0JG, United Kingdom
| | - A. James Mason
- Institute
of Pharmaceutical Science, School of Cancer & Pharmaceutical Science, King’s College London, Franklin-Wilkins Building, 150 Stamford
Street, London SE1 9NH, United Kingdom
| | - Megan J. Bester
- Department
of Anatomy, Faculty of Health Sciences, University of Pretoria, Pretoria 0002, South Africa
| | - Anabella R. M. Gaspar
- Department
of Biochemistry, Genetics and Microbiology, Faculty of Natural and
Agricultural Sciences, University of Pretoria, Pretoria 0002, South Africa
| |
Collapse
|
37
|
Chen M, Hu Z, Shi J, Xie Z. Human β-defensins and their synthetic analogs: Natural defenders and prospective new drugs of oral health. Life Sci 2024; 346:122591. [PMID: 38548013 DOI: 10.1016/j.lfs.2024.122591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/08/2024] [Accepted: 03/24/2024] [Indexed: 04/14/2024]
Abstract
As a family of cationic host defense peptides, human β-defensins (HBDs) are ubiquitous in the oral cavity and are mainly synthesized primarily by epithelial cells, serving as the primary barrier and aiming to prevent microbial invasion, inflammation, and disease while maintaining physiological homeostasis. In recent decades, there has been great interest in their biological functions, structure-activity relationships, mechanisms of action, and therapeutic potential in oral diseases. Meanwhile, researchers are dedicated to improving the properties of HBDs for clinical application. In this review, we first describe the classification, structural characteristics, functions, and mechanisms of HBDs. Next, we cover the role of HBDs and their synthetic analogs in oral diseases, including dental caries and pulp infections, periodontitis, peri-implantitis, fungal/viral infections and oral mucosal diseases, and oral squamous cell carcinoma. Finally, we discuss the limitations and challenges of clinical translation of HBDs and their synthetic analogs, including, but not limited to, stability, bioavailability, antimicrobial activity, resistance, and toxicity. Above all, this review summarizes the biological functions, mechanisms of action, and therapeutic potential of both natural HBDs and their synthetic analogs in oral diseases, as well as the challenges associated with clinical translation, thus providing substantial insights into the laboratory development and clinical application of HBDs in oral diseases.
Collapse
Affiliation(s)
- Mumian Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zihe Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Jue Shi
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
38
|
Alves PM, Barrias CC, Gomes P, Martins MCL. How can biomaterial-conjugated antimicrobial peptides fight bacteria and be protected from degradation? Acta Biomater 2024; 181:98-116. [PMID: 38697382 DOI: 10.1016/j.actbio.2024.04.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/19/2024] [Accepted: 04/25/2024] [Indexed: 05/05/2024]
Abstract
The emergence of antibiotic-resistant bacteria is a serious threat to public health. Antimicrobial peptides (AMP) are a powerful alternative to antibiotics due to their low propensity to induce bacterial resistance. However, cytotoxicity and short half-lives have limited their clinical translation. To overcome these problems, AMP conjugation has gained relevance in the biomaterials field. Nevertheless, few studies describe the influence of conjugation on enzymatic protection, mechanism of action and antimicrobial efficacy. This review addresses this gap by providing a detailed comparison between conjugated and soluble AMP. Additionally, commonly employed chemical reactions and factors to consider when promoting AMP conjugation are reviewed. The overall results suggested that AMP conjugated onto biomaterials are specifically protected from degradation by trypsin and/or pepsin. However, sometimes, their antimicrobial efficacy was reduced. Due to limited conformational freedom in conjugated AMP, compared to their soluble forms, they appear to act initially by creating small protuberances on bacterial membranes that may lead to the alteration of membrane potential and/or formation of holes, triggering cell death. Overall, AMP conjugation onto biomaterials is a promising strategy to fight infection, particularly associated to the use of medical devices. Nonetheless, some details need to be addressed before conjugated AMP reach clinical practice. STATEMENT OF SIGNIFICANCE: Covalent conjugation of antimicrobial peptides (AMP) has been one of the most widely used strategies by bioengineers, in an attempt to not only protect AMP from proteolytic degradation, but also to prolong their residence time at the target tissue. However, an explanation for the mode of action of conjugated AMP is still lacking. This review extensively gathers works on AMP conjugation and puts forward a mechanism of action for AMP when conjugated onto biomaterials. The implications of AMP conjugation on antimicrobial activity, cytotoxicity and resistance to proteases are all discussed. A thorough review of commonly employed chemical reactions for this conjugation is also provided. Finally, details that need to be addressed for conjugated AMP to reach clinical practice are discussed.
Collapse
Affiliation(s)
- Pedro M Alves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; Faculdade de Engenharia, Universidade do Porto, Rua Dr. Roberto Frias, 4200-465 Porto, Portugal; LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal
| | - Paula Gomes
- LAQV-REQUIMTE, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Rua do Campo Alegre 687, 4169-007 Porto, Portugal
| | - M Cristina L Martins
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-135 Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira, 4050-313 Porto, Portugal.
| |
Collapse
|
39
|
Bae DH, Bae H, Yu HS, Dorjsembe B, No YH, Kim T, Kim NH, Kim JW, Kim J, Lee BS, Kim YJ, Park S, Khaleel ZH, Sa DH, Lee EC, Lee J, Ham J, Kim JC, Kim YH. Peptide-Drug Conjugate with Statistically Designed Transcellular Peptide for Psoriasis-Like Inflammation. Adv Healthc Mater 2024; 13:e2303480. [PMID: 38421096 DOI: 10.1002/adhm.202303480] [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: 10/11/2023] [Revised: 02/26/2024] [Indexed: 03/02/2024]
Abstract
Peptide-drug conjugates (PDCs) are a promising class of drug delivery systems that utilize covalently conjugated carrier peptides with therapeutic agents. PDCs offer several advantages over traditional drug delivery systems including enhanced target engagement, improved bioavailability, and increased cell permeability. However, the development of efficient transcellular peptides capable of effectively transporting drugs across biological barriers remains an unmet need. In this study, physicochemical criteria based on cell-penetrating peptides are employed to design transcellular peptides derived from an antimicrobial peptides library. Among the statistically designed transcellular peptides (SDTs), SDT7 exhibits higher skin permeability, faster kinetics, and improved cell permeability in human keratinocyte cells compared to the control peptide. Subsequently, it is found that 6-Paradol (PAR) exhibits inhibitory activity against phosphodiesterase 4, which can be utilized for an anti-inflammatory PDC. The transcellular PDC (SDT7-conjugated with PAR, named TM5) is evaluated in mouse models of psoriasis, exhibiting superior therapeutic efficacy compared to PAR alone. These findings highlight the potential of transcellular PDCs (TDCs) as a promising approach for the treatment of inflammatory skin disorders.
Collapse
Affiliation(s)
- Do Hyun Bae
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea
| | - Hayeon Bae
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Hyung-Seok Yu
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Banzragch Dorjsembe
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Young Hyun No
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Taejung Kim
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Nam Hyeong Kim
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Jin-Woo Kim
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Jiyool Kim
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Bok-Soo Lee
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Ye Ji Kim
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Seongchan Park
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Zinah Hilal Khaleel
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Deok Hyang Sa
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Eui-Chul Lee
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Jaecheol Lee
- School of Pharmacy, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- IMNEWRUN Inc., Suwon, 16419, Republic of Korea
| | - Jungyeob Ham
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Jin-Chul Kim
- Natural Products Research Institute, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon, 34113, Republic of Korea
| | - Yong Ho Kim
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Center for Neuroscience Imaging Research (CNIR), Institute for Basic Science (IBS), Suwon, 16419, Republic of Korea
- SKKU Advanced Institute of Nano Technology (SAINT), Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Nano Science and Technology, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- IMNEWRUN Inc., Suwon, 16419, Republic of Korea
- Department of Nano Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| |
Collapse
|
40
|
Liang Q, Liu Z, Liang Z, Zhu C, Li D, Kong Q, Mou H. Development strategies and application of antimicrobial peptides as future alternatives to in-feed antibiotics. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 927:172150. [PMID: 38580107 DOI: 10.1016/j.scitotenv.2024.172150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/14/2024] [Accepted: 03/30/2024] [Indexed: 04/07/2024]
Abstract
The use of in-feed antibiotics has been widely restricted due to the significant environmental pollution and food safety concerns they have caused. Antimicrobial peptides (AMPs) have attracted widespread attention as potential future alternatives to in-feed antibiotics owing to their demonstrated antimicrobial activity and environment friendly characteristics. However, the challenges of weak bioactivity, immature stability, and low production yields of natural AMPs impede practical application in the feed industry. To address these problems, efforts have been made to develop strategies for approaching the AMPs with enhanced properties. Herein, we summarize approaches to improving the properties of AMPs as potential alternatives to in-feed antibiotics, mainly including optimization of structural parameters, sequence modification, selection of microbial hosts, fusion expression, and industrially fermentation control. Additionally, the potential for application of AMPs in animal husbandry is discussed. This comprehensive review lays a strong theoretical foundation for the development of in-feed AMPs to achieve the public health globally.
Collapse
Affiliation(s)
- Qingping Liang
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Zhemin Liu
- Fundamental Science R&D Center of Vazyme Biotech Co. Ltd., Nanjing 210000, China
| | - Ziyu Liang
- Section of Neurobiology, Department of Biological Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Changliang Zhu
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Dongyu Li
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Qing Kong
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China
| | - Haijin Mou
- College of Food Science and Engineering, Ocean University of China, Qingdao 266404, China.
| |
Collapse
|
41
|
Tran DVH, Luong HX, Kim DH, Lee BJ, Kim YW. Lysine-homologue substitution: Impact on antimicrobial activity and proteolytic stability of cationic stapled heptapeptides. Bioorg Med Chem 2024; 106:117735. [PMID: 38714021 DOI: 10.1016/j.bmc.2024.117735] [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: 02/22/2024] [Revised: 04/19/2024] [Accepted: 04/19/2024] [Indexed: 05/09/2024]
Abstract
Numerous natural antimicrobial peptides (AMPs) exhibit a cationic amphipathic helical conformation, wherein cationic amino acids, such as lysine and arginine, play pivotal roles in antimicrobial activity by aiding initial attraction to negatively charged bacterial membranes. Expanding on our previous work, which introduced a de novo design of amphipathic helices within cationic heptapeptides using an 'all-hydrocarbon peptide stapling' approach, we investigated the impact of lysine-homologue substitution on helix formation, antimicrobial activity, hemolytic activity, and proteolytic stability of these novel AMPs. Our results demonstrate that substituting lysine with ornithine enhances both the antimicrobial activity and proteolytic stability of the stapled heptapeptide AMP series, while maintaining low hemolytic activity. This finding underscores lysine-homologue substitution as a valuable strategy for optimizing the therapeutic potential of diverse cationic AMPs.
Collapse
Affiliation(s)
- Duc V H Tran
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea; Intergrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Huy X Luong
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea; Intergrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea
| | - Do-Hee Kim
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Bong-Jin Lee
- College of Pharmacy, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Woo Kim
- College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea; Intergrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang 10326, Republic of Korea.
| |
Collapse
|
42
|
Hu Y, Yu L, Dai Q, Hu X, Shen Y. Multifunctional antibacterial hydrogels for chronic wound management. Biomater Sci 2024; 12:2460-2479. [PMID: 38578143 DOI: 10.1039/d4bm00155a] [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: 04/06/2024]
Abstract
Chronic wounds have gradually evolved into a global health challenge, comprising long-term non-healing wounds, local tissue necrosis, and even amputation in severe cases. Accordingly, chronic wounds place a considerable psychological and economic burden on patients and society. Chronic wounds have multifaceted pathogenesis involving excessive inflammation, insufficient angiogenesis, and elevated reactive oxygen species levels, with bacterial infection playing a crucial role. Hydrogels, renowned for their excellent biocompatibility, moisture retention, swelling properties, and oxygen permeability, have emerged as promising wound repair dressings. However, hydrogels with singular functions fall short of addressing the complex requirements associated with chronic wound healing. Hence, current research emphasises the development of multifunctional antibacterial hydrogels. This article reviews chronic wound characteristics and the properties and classification of antibacterial hydrogels, as well as their potential application in chronic wound management.
Collapse
Affiliation(s)
- Yungang Hu
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Lu Yu
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Qiang Dai
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Xiaohua Hu
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| | - Yuming Shen
- Department of Burns Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China.
- Clinical Center for Wounds, Capital Medical University, Beijing, 100035, China
| |
Collapse
|
43
|
Sun L, Ouyang J, She Z, Li R, Zeng F, Yao Z, Wu S. Injectable-Hydrogel-Based Tissue Sealant for Hemostasis, Bacteria Inhibition, and Pro-Angiogenesis in Organ Bleeding Wounds and Therapeutic Outcome Monitoring Via NIR-II Optical Imaging. Adv Healthc Mater 2024; 13:e2303997. [PMID: 38281086 DOI: 10.1002/adhm.202303997] [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: 11/14/2023] [Revised: 01/20/2024] [Indexed: 01/29/2024]
Abstract
Sudden hemorrhage stemming from internal organ wounds poses a grave and potentially fatal risk if left untreated. Injectable-hydrogel-based tissue sealants featuring multiple actions, including fit-to-shape in situ gelation, rapid hemostasis, pro-angiogenic, anti-bacterial and outcome tracking, are ideal for the management of organ trauma wounds. Herein, an injectable-hydrogel tissue sealant AN@CD-PEG&TQ which consists of four-arm 4-arm poly(ethylene glycol) (PEG-SC) succinimidyl carbonate), AN@CD nanoprobe, and two bioactive peptides (anti-microbial peptide Tet213 and pro-angiogenic peptide QK) is developed. Among them, AN@CD nanoparticles form through host/guest complexation of amino-group-containing β-cyclodextrin and adamantyl group, enabling in situ biomarker (NO)-activatable optoacoustic/NIR-II: Near-infrared second biological window fluorescent imaging. The ample ─NH2 groups on the surface of AN@CD readily engage in rapid cross-linking with succinimidyl ester groups located at the ends of four-arm PEG-SC. This cross-linking expedites the gelation process without necessitating additional initiators or cross-linking agents; thus, significantly enhancing both hydrogel's application convenience and biocompatibility. Bioactive peptides (Tet213 and QK) safeguard against possible bacterial infections, facilitate angiogenesis, and eventually, improve organ wounds healing. This hydrogel-based tissue sealant demonstrates superior therapeutic and bioimaging performance in various mouse models including liver hemorrhage, gastric perforation, and bacterial-infected skin wound mouse models, highlighting its potential as a high-performance wound sealant for organ bleeding wound management.
Collapse
Affiliation(s)
- Lihe Sun
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Juan Ouyang
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Zunpan She
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Rong Li
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Fang Zeng
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| | - Zhicheng Yao
- Department of Hepatobiliary & Pancreatic Surgery, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, 510630, China
| | - Shuizhu Wu
- Biomedical Division, State Key Laboratory of Luminescent Materials and Devices, Guangdong Provincial Key Laboratory of Luminescence from Molecular Aggregates, College of Materials Science and Engineering, South China University of Technology, Guangzhou, 510640, China
| |
Collapse
|
44
|
Gafar MA, Omolo CA, Elhassan E, Ibrahim UH, Govender T. Applications of peptides in nanosystems for diagnosing and managing bacterial sepsis. J Biomed Sci 2024; 31:40. [PMID: 38637839 PMCID: PMC11027418 DOI: 10.1186/s12929-024-01029-2] [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: 02/25/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024] Open
Abstract
Sepsis represents a critical medical condition stemming from an imbalanced host immune response to infections, which is linked to a significant burden of disease. Despite substantial efforts in laboratory and clinical research, sepsis remains a prominent contributor to mortality worldwide. Nanotechnology presents innovative opportunities for the advancement of sepsis diagnosis and treatment. Due to their unique properties, including diversity, ease of synthesis, biocompatibility, high specificity, and excellent pharmacological efficacy, peptides hold great potential as part of nanotechnology approaches against sepsis. Herein, we present a comprehensive and up-to-date review of the applications of peptides in nanosystems for combating sepsis, with the potential to expedite diagnosis and enhance management outcomes. Firstly, sepsis pathophysiology, antisepsis drug targets, current modalities in management and diagnosis with their limitations, and the potential of peptides to advance the diagnosis and management of sepsis have been adequately addressed. The applications have been organized into diagnostic or managing applications, with the last one being further sub-organized into nano-delivered bioactive peptides with antimicrobial or anti-inflammatory activity, peptides as targeting moieties on the surface of nanosystems against sepsis, and peptides as nanocarriers for antisepsis agents. The studies have been grouped thematically and discussed, emphasizing the constructed nanosystem, physicochemical properties, and peptide-imparted enhancement in diagnostic and therapeutic efficacy. The strengths, limitations, and research gaps in each section have been elaborated. Finally, current challenges and potential future paths to enhance the use of peptides in nanosystems for combating sepsis have been deliberately spotlighted. This review reaffirms peptides' potential as promising biomaterials within nanotechnology strategies aimed at improving sepsis diagnosis and management.
Collapse
Affiliation(s)
- Mohammed A Gafar
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
- Department of Pharmaceutics, Faculty of Pharmacy, University of Khartoum, P.O. Box 1996, Khartoum, Sudan
| | - Calvin A Omolo
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
- Department of Pharmaceutics and Pharmacy Practice, School of Pharmacy and Health Sciences, United States International University-Africa, P. O. Box 14634-00800, Nairobi, Kenya.
| | - Eman Elhassan
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa
| | - Usri H Ibrahim
- Discipline of Human Physiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Thirumala Govender
- Discipline of Pharmaceutical Sciences, College of Health Sciences, University of KwaZulu-Natal, Private Bag X54001, Durban, South Africa.
| |
Collapse
|
45
|
Zhang M, Yang B, Shi J, Wang Z, Liu Y. Host defense peptides mitigate the spread of antibiotic resistance in physiologically relevant condition. Antimicrob Agents Chemother 2024; 68:e0126123. [PMID: 38415983 PMCID: PMC10994823 DOI: 10.1128/aac.01261-23] [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: 09/30/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
Antibiotic resistance represents a significant challenge to public health and human safety. The primary driver behind the dissemination of antibiotic resistance is the horizontal transfer of plasmids. Current conjugative transfer assay is generally performed in a standardized manner, ignoring the effect of the host environment. Host defense peptides (HDPs) possess a wide range of biological targets and play an essential role in the innate immune system. Herein, we reveal that sub-minimum inhibitory concentrations of HDPs facilitate the conjugative transfer of RP4-7 plasmid in the Luria Broth medium, and this observation is reversed in the RPMI medium, designed to simulate the host environment. Out of these HDPs, indolicidin (Ind), a cationic tridecapeptide from bovine neutrophils, significantly inhibits the conjugation of multidrug resistance plasmids in a dose-dependent manner, including blaNDM- and tet(X4)-bearing plasmids. We demonstrate that the addition of Ind to RPMI medium as the incubation substrate downregulates the expression of conjugation-related genes. In addition, Ind weakens the tricarboxylic acid cycle, impedes the electron transport chain, and disrupts the proton motive force, consequently diminishing the synthesis of adenosine triphosphate and limiting the energy supply. Our findings highlight the importance of the host-like environments for the development of horizontal transfer inhibitors and demonstrate the potential of HDPs in preventing the spread of resistance plasmids.
Collapse
Affiliation(s)
- Miao Zhang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Bingqing Yang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Jingru Shi
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
46
|
Peng Z, Wei C, Cai J, Zou Z, Chen J. Characterization of an antimicrobial peptide family from the venom gland of Heteropoda venatoria. Toxicon 2024; 241:107657. [PMID: 38428753 DOI: 10.1016/j.toxicon.2024.107657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/15/2024] [Accepted: 02/20/2024] [Indexed: 03/03/2024]
Abstract
Spider venom boasts extensive peptide diversity, constituting a natural biochemical arsenal for defense and predation. The new family HvAMPs, including 9 homologous members, were identified from the unnormalized cDNA library of Heteropoda venatoria venom gland by Sanger sequencing. The putative mature peptide is composed of 22 aliphatic amino acid residues. The mature peptides of HvAMP1 and HvAMP5, with 3 different amino acids, were synthesized and both were shown to adopt an amphipathic α-helical structure and amphipathicity in SDS buffer by CD spectroscopy. In comparison to HvAMP1, HvAMP5 exhibits higher antibacterial activity, particularly against Gram-positive bacteria, coupled with reduced hemolytic activity and cytotoxicity. Results from SYTO 9/PI staining indicate that HvAMP5 acts by disrupting bacterial cell membranes. Analysis of the relationships between structures and functions suggests that HvAMP5 enhances antibacterial activity and reduces mammalian cell toxicity by increasing positive charge and proline substitution. The three residues variation can augment the electrostatic attraction of antibacterial peptides to the bacterial phospholipid bilayer. The present study suggests that the HvAMPs may exert lytic action against cells of different origins to increase cellular and tissue barrier permeability to facilitate spider's defense or predation. Moreover, HvAMP5 holds promise as a novel antibacterial agent for treating Gram-positive bacterial infections. Simultaneously, the numerous diverse amino acid residue substitutions within the HvAMP family offer a template for future study.
Collapse
Affiliation(s)
- Zhihao Peng
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410000, China
| | - Chao Wei
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410000, China
| | - Jisen Cai
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410000, China
| | - Zhaoxia Zou
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410000, China; School of Public Health & Laboratory Medicine, Hunan University of Medicine, Huaihua, 418000, China.
| | - Jinjun Chen
- College of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, 410000, China; Hunan Provincial Engineering Technology Research Center for Cell Mechanics and Function Analysis, Changsha, 418000, China.
| |
Collapse
|
47
|
Kordi M, Talkhounche PG, Vahedi H, Farrokhi N, Tabarzad M. Heterologous Production of Antimicrobial Peptides: Notes to Consider. Protein J 2024; 43:129-158. [PMID: 38180586 DOI: 10.1007/s10930-023-10174-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/28/2023] [Indexed: 01/06/2024]
Abstract
Heavy and irresponsible use of antibiotics in the last century has put selection pressure on the microbes to evolve even faster and develop more resilient strains. In the confrontation with such sometimes called "superbugs", the search for new sources of biochemical antibiotics seems to have reached the limit. In the last two decades, bioactive antimicrobial peptides (AMPs), which are polypeptide chains with less than 100 amino acids, have attracted the attention of many in the control of microbial pathogens, more than the other types of antibiotics. AMPs are groups of components involved in the immune response of many living organisms, and have come to light as new frontiers in fighting with microbes. AMPs are generally produced in minute amounts within organisms; therefore, to address the market, they have to be either produced on a large scale through recombinant DNA technology or to be synthesized via chemical methods. Here, heterologous expression of AMPs within bacterial, fungal, yeast, plants, and insect cells, and points that need to be considered towards their industrialization will be reviewed.
Collapse
Affiliation(s)
- Masoumeh Kordi
- Department of Cell & Molecular Biology, Faculty of Life Sciences & Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Parnian Ghaedi Talkhounche
- Department of Cell & Molecular Biology, Faculty of Life Sciences & Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Helia Vahedi
- Department of Cell & Molecular Biology, Faculty of Life Sciences & Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Naser Farrokhi
- Department of Cell & Molecular Biology, Faculty of Life Sciences & Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Maryam Tabarzad
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
48
|
Pereira AJ, Xing H, de Campos LJ, Seleem MA, de Oliveira KMP, Obaro SK, Conda-Sheridan M. Structure-Activity Relationship Study to Develop Peptide Amphiphiles as Species-Specific Antimicrobials. Chemistry 2024; 30:e202303986. [PMID: 38221408 PMCID: PMC10939825 DOI: 10.1002/chem.202303986] [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: 11/29/2023] [Revised: 01/05/2024] [Accepted: 01/09/2024] [Indexed: 01/16/2024]
Abstract
Antimicrobial peptide amphiphiles (PAs) are a promising class of molecules that can disrupt the bacterial membrane or act as drug nanocarriers. In this study, we prepared 33 PAs to establish supramolecular structure-activity relationships. We studied the morphology and activity of the nanostructures against different Gram-positive and Gram-negative bacterial strains (such as Staphylococcus aureus, Escherichia coli, Pseudomonas aeruginosa and Acinetobacter baumannii). Next, we used principal component analysis (PCA) to determine the key contributors to activity. We found that for S. aureus, the zeta potential was the major contributor to the activity while Gram-negative bacteria were more influenced by the partition coefficient (LogP) with the following order P. aeruginosa>E. coli>A. baumannii. We also performed a study of the mechanism of action of selected PAs on the bacterial membrane assessing the membrane permeability and depolarization, changes in zeta potential and overall integrity. We studied the toxicity of the nanostructures against mammalian cells. Finally, we performed an in vivo study using the wax moth larvae to determine the therapeutic efficacy of the active PAs. This study shows cationic PA nanostructures can be an intriguing platform for the development of nanoantibacterials.
Collapse
Affiliation(s)
- Aramis J. Pereira
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| | - Huihua Xing
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| | - Luana J. de Campos
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| | - Mohamed A. Seleem
- Dr. M.A. Seleem, Department of Pharmaceutical Organic Chemistry, Al-Azhar University, Cairo, 4434003 (Egypt)
| | - Kelly M. P. de Oliveira
- Prof. Dr. K. M. P. de Oliveira, Department of Biological and Environmental Science, Federal University of Grande Dourados (UFGD), Dourados, MS 79804-970 (Brazil)
| | - Stephen K. Obaro
- Prof. Dr. S. K. Obaro, Division of Pediatric Infectious Diseases, University of Alabama at Birmingham (UAB), Birmingham, AL 35233 (USA), International Foundation against Infectious Diseases in Nigeria (IFAIN), Abuja, 900108 (Nigeria)
| | - Martin Conda-Sheridan
- A. J. Pereira, Dr. H. Xing, L. J. de Campos, Prof. Dr. M. Conda-Sheridan, Department of Pharmaceutical Sciences, University of Nebraska Medical Center (UNMC), Omaha, NE 68198 (USA)
| |
Collapse
|
49
|
Liu L, Fan X, Lu Q, Wang P, Wang X, Han Y, Wang R, Zhang C, Han S, Tsuboi T, Dai H, Yeow J, Geng H. Antimicrobial research of carbohydrate polymer- and protein-based hydrogels as reservoirs for the generation of reactive oxygen species: A review. Int J Biol Macromol 2024; 260:129251. [PMID: 38211908 DOI: 10.1016/j.ijbiomac.2024.129251] [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: 09/12/2023] [Revised: 12/23/2023] [Accepted: 01/03/2024] [Indexed: 01/13/2024]
Abstract
Reactive oxygen species (ROS) play an important role in biological milieu. Recently, the rapid growth in our understanding of ROS and their promise in antibacterial applications has generated tremendous interest in the combination of ROS generators with bulk hydrogels. Hydrogels represent promising supporters for ROS generators and can locally confine the nanoscale distribution of ROS generators whilst also promoting cellular integration via biomaterial-cell interactions. This review highlights recent efforts and progress in developing hydrogels derived from biological macromolecules with embedded ROS generators with a focus on antimicrobial applications. Initially, an overview of passive and active antibacterial hydrogels is provided to show the significance of proper hydrogel selection and design. These are followed by an in-depth discussion of the various approaches for ROS generation in hydrogels. The structural engineering and fabrication of ROS-laden hydrogels are given with a focus on their biomedical applications in therapeutics and diagnosis. Additionally, we discuss how a compromise needs to be sought between ROS generation and removal for maximizing the efficacy of therapeutic treatment. Finally, the current challenges and potential routes toward commercialization in this rapidly evolving field are discussed, focusing on the potential translation of laboratory research outcomes to real-world clinical outcomes.
Collapse
Affiliation(s)
- Lan Liu
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212000, China; Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China
| | - Xin Fan
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| | - Qianyun Lu
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212000, China; Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China
| | - Pengxu Wang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| | - Xingang Wang
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212000, China.
| | - Yuxing Han
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| | - Runming Wang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| | - Canyang Zhang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| | - Sanyang Han
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| | - Tatsuhisa Tsuboi
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| | - Hongliang Dai
- School of Environmental and Chemical Engineering, Jiangsu University of Science and Technology, Zhenjiang 212000, China.
| | - Jonathan Yeow
- Graduate School of Biomedical Engineering, The University of New South Wales Sydney, Sydney, NSW 2052, Australia.
| | - Hongya Geng
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518075, China.
| |
Collapse
|
50
|
Colomina-Alfaro L, Sist P, Marchesan S, Urbani R, Stamboulis A, Bandiera A. A Versatile Elastin-Like Carrier for Bioactive Antimicrobial Peptide Production and Delivery. Macromol Biosci 2024; 24:e2300236. [PMID: 37698188 DOI: 10.1002/mabi.202300236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/07/2023] [Indexed: 09/13/2023]
Abstract
Elastin-like polypeptides are biotechnological protein and peptide carriers that offer a vast scope of applicability. This work aims to build a model for the expression of antimicrobial peptides (AMPs) by genetically engineering the Human Elastin-like Polypeptide platform developed in the lab. The well-characterized AMP indolicidin is selected as an example of an antimicrobial domain for the recombinant fusion at the C-terminus of the carrier. The fusion construct has been designed to allow the release of the antimicrobial domain. The expression product has been purified and its physicochemical and antimicrobial properties has been characterized. Taking advantage of the self-assembling and matrix-forming properties of the recombinant biopolymer, the materials that are obtained have been evaluated for antimicrobial activity toward bacterial-strain models. This approach represents a cost-effective strategy for the production of smart components and materials endowed with antimicrobial capacity triggered by external stimuli.
Collapse
Affiliation(s)
- Laura Colomina-Alfaro
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Paola Sist
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Silvia Marchesan
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Ranieri Urbani
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, via L. Giorgieri, 1, Trieste, 34127, Italy
| | - Artemis Stamboulis
- School of Metallurgy and Materials, Biomaterials Research Group, University of Birmingham, Edgbaston, Birmingham, B15 2TT, UK
| | - Antonella Bandiera
- Department of Life Sciences, University of Trieste, via L. Giorgieri, 1, Trieste, 34127, Italy
| |
Collapse
|