1
|
Zarro PR, De Felice S, Sabbieti MG, Agas D. The Inflamed Bone Marrow Scenery Amongst the Symplegades of Ageing and Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40279001 DOI: 10.1007/5584_2025_860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Upon inflammation, the bone marrow (BM) landscape undergoes significant architectural and functional modifications. Stimulation of the hematopoietic niche triggers a series of lightning events, which begin with stem/progenitor blood elements mobilization and culminates with the activation of immune responses. Ageing partially mirrors this process, albeit with a propensity towards chronic inflammation and immune dysfunction. Age-related chronic inflammation disrupts bone homeostasis and accompanies impaired tissue regeneration. Thus, focusing on the bone marrow's dynamics during inflammatory bone diseases could lay the way for the development of novel therapeutic platforms aimed at niche reprogramming. Herein, we summarize inflammatory and age-induced processes in multiple BM compartments, with particular reference to hematopoietic, stromal stem/progenitor cells, and mature immunocytes. Finally, we focus on autophagy and its potential to clinically re-modulate the pathological "flogistic" bias, possibly by restoring functional phenotypes within the bone marrow niche elements.
Collapse
Affiliation(s)
- Pier Raffaele Zarro
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | - Simona De Felice
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy
| | | | - Dimitrios Agas
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, MC, Italy.
| |
Collapse
|
2
|
Tang Y, Guo T, Wang X, Li C, Zhang X, Zhang J. Cyclodextrin-Derived Macromolecular Therapies for Inflammatory Diseases. Macromol Biosci 2025:e2400637. [PMID: 40271896 DOI: 10.1002/mabi.202400637] [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: 12/21/2024] [Revised: 04/02/2025] [Indexed: 04/25/2025]
Abstract
Inflammation is an essential physiological defense mechanism against harmful stimuli, yet dysregulated inflammatory responses are closely associated with the pathogenesis of numerous acute and chronic diseases. Recent advances highlight the remarkable anti-inflammatory potential of bioactive macromolecules, particularly cyclodextrins (CDs) and their engineered derivatives, which are emerging as promising therapeutic agents. This review systematically introduces different CDs and CD-derived macromolecules that demonstrate anti-inflammatory properties, with emphasis on their molecular mechanisms of action. Native CDs exhibit direct therapeutic effects through host-guest interactions, enabling selective sequestration of pathogenic components such as cholesterol crystals and proteins that drive inflammatory cascades. Moreover, chemically modified CD derivatives incorporating functional groups demonstrate enhanced capabilities in neutralizing inflammatory mediators and modulating immune cell responses. This work further discusses the expanding therapeutic applications of these macromolecules across diverse inflammatory conditions, ranging from acute tissue injuries to chronic autoimmune disorders. Finally, this work critically analyzes the crucial challenges and emerging opportunities in translating CD-based macromolecular therapies into clinical practice, addressing key considerations in biocompatibility, targeted delivery, and therapeutic efficacy optimization.
Collapse
Affiliation(s)
- Yige Tang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
- International Medical College, Chongqing Medical University, Chongqing, 400016, China
| | - Tao Guo
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Xuanran Wang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Chenwen Li
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xiangjun Zhang
- College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, 313 Gaoteng Avenue, Jiulongpo District, Chongqing, 400039, China
| |
Collapse
|
3
|
Brigi C, Aghila Rani K, Selvakumar B, Hamad M, Abou Neel EA, Samsudin A. Decoding biomaterial-associated molecular patterns (BAMPs): influential players in bone graft-related foreign body reactions. PeerJ 2025; 13:e19299. [PMID: 40292103 PMCID: PMC12024449 DOI: 10.7717/peerj.19299] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/19/2025] [Indexed: 04/30/2025] Open
Abstract
Bone grafts frequently induce immune-mediated foreign body reactions (FBR), which hinder their clinical performance and result in failure. Understanding biomaterial-associated molecular patterns (BAMPs), including physicochemical properties of biomaterial, adsorbed serum proteins, and danger signals, is crucial for improving bone graft outcomes. Recent studies have investigated the role of BAMPs in the induction and maintenance of FBR, thereby advancing the understanding of FBR kinetics, triggers, stages, and key contributors. This review outlines the stages of FBR, the components of BAMPs, and their roles in immune activation. It also discusses various bone grafting biomaterials, their physicochemical properties influencing protein adsorption and macrophage modulation, and the key mechanisms of protein adsorption on biomaterial surfaces. Recent advancements in surface modifications and immunomodulatory strategies to mitigate FBR are also discussed. Furthermore, the authors look forward to future studies that will focus on a comprehensive proteomic analysis of adsorbed serum proteins, a crucial component of BAMPs, to identify proteins that promote or limit inflammation. This understanding could facilitate the design of biomaterials that selectively adsorb beneficial proteins, thereby reducing the risk of FBR and enhancing bone regeneration.
Collapse
Affiliation(s)
- Carel Brigi
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - K.G. Aghila Rani
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Balachandar Selvakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
| | - Mawieh Hamad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Medical Laboratory Sciences, University of Sharjah, Sharjah, United Arab Emirates
| | - Ensanya Ali Abou Neel
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Department of Preventive and Restorative Dentistry, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - A.R. Samsudin
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, University City, United Arab Emirates
- Oral and Craniofacial Health Sciences Department, College of Dental Medicine, University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
4
|
She Z, Zeng F, Wu S. A zwitterionic chromophore as both a biomarker-activatable optical imaging probe and a therapeutic agent for the detection and treatment of acute lung injury with bacterial infection. Biomater Sci 2025. [PMID: 40243112 DOI: 10.1039/d5bm00419e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025]
Abstract
Acute lung injury (ALI), often complicated by bacterial infection, poses significant challenges in diagnosis and treatment. Nitric oxide (NO) plays a key role in the pathophysiology of ALI, making it an ideal biomarker for early detection. In this study, we developed a zwitterionic chromophore, ZW-N, designed as both a biomarker-activatable imaging probe and a therapeutic agent for ALI with bacterial infection. The chromophore ZW-N integrates quaternary ammonium groups for antimicrobial activity and zwitterionic sulfonate groups to enhance biocompatibility and water solubility. Built on a flexible propanyl linker that couples two heptamethine cyanine dyes, ZW-N enables biomarker-responsive dual-modal imaging via optoacoustic (OA) imaging and near-infrared second-window (NIR-II) fluorescence imaging. Moreover, the chromophore ZW-N demonstrates therapeutic efficacy when combined with the clinically used antioxidant N-acetylcysteine (NAC) to treat ALI with bacterial infection. This dual-functional chromophore offers a promising platform for non-invasive, real-time monitoring of ALI, providing significant potential for improved detection and a more effective treatment strategy.
Collapse
Affiliation(s)
- 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.
| | - 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.
| | - 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
|
5
|
Verma M, Randhawa S, Bathla M, Teji N, Acharya A. Strategic use of nanomaterials as double-edged therapeutics to control carcinogenesis via regulation of dysbiosis and bacterial infection: current status and future prospects. J Mater Chem B 2025; 13:4770-4790. [PMID: 40192037 DOI: 10.1039/d4tb02409e] [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: 04/17/2025]
Abstract
The human microbiome plays a crucial role in modulating health and disease susceptibility through a complex network of interactions with the host. When the delicate balance of this microbial ecosystem is disrupted, it often correlates with the onset of systemic diseases. An over-abundance of pathogenic microorganisms within the microbiome has been implicated as a driving factor in the development of disease conditions such as diabetes, obesity, and chronic infections. It has been observed that microbiome dysbiosis perturbs metabolic, inflammatory, and immunological pathways, potentially facilitating carcinogenesis. Furthermore, the metabolites associated with microbial dysbiosis exert multifaceted effects, including metabolic interference, host DNA damage, and tumor promotion, further underscoring the microbiome's significance in several of the cancers. This new exploration of microbiome involvement in carcinogenesis needs additional patient sample analysis, which could provide new insights into cancer diagnosis and treatment. However, treating these diseases using drugs, traditional methods, etc. has resulted in multi-drug resistance, and this has eventually made the situation worrisome. This review highlights the importance of nanotechnology, which may tackle these pathogenic conditions simultaneously by targeting common receptors present in bacteria and cancer. Herein, we have explained how nanotechnology may come to the forefront for these treatments. It explores the potential of non-antibiotic disinfectants, i.e., nanoparticles (NPs) with dual targeting capabilities against microbes and cancer cells, using mechanisms such as ROS generation and DNA damage while minimizing the chances of drug resistance.
Collapse
Affiliation(s)
- Mohini Verma
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Shiwani Randhawa
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Manik Bathla
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Nandini Teji
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Amitabha Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur (H.P.) 176061, India.
- Academy of Scientific & Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
6
|
Park J, Wu Y, Le QV, Kim JS, Xu E, Lee J, Oh YK. Self-disassembling nanoparticles as oral nanotherapeutics targeting intestinal microenvironment. Nat Commun 2025; 16:3365. [PMID: 40204740 PMCID: PMC11982569 DOI: 10.1038/s41467-025-58513-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] [Received: 07/09/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Inspired by the survival strategies of pyomelanin-producing microbes, we synthesize pyomelanin nanoparticles (PMNPs) from homogentisic acid- γ-lactone via auto-oxidation and investigate their biomedical potential. PMNPs possess distinct physicochemical properties, including reactive oxygen species scavenging and microenvironment-responsive self-disassembly. Under intestinal conditions, PMNPs self-disassemble and penetrate the nanoscale pores of the mucin layer. In an inflammatory bowel disease model, orally administered PMNPs withstand gastric acidity and, in their solubilized form, interact with macrophages and epithelial cells. They significantly reduce reactive oxygen species levels, exert anti-inflammatory effects, and restore gut microbiota composition. Compared to conventional nanoparticles and 5-aminosalicylic acid, PMNPs exhibit greater therapeutic efficacy. Clinical symptoms and intestinal inflammation are alleviated, and the gut microbiota is restored to near-normal levels. These findings underscore the therapeutic potential of PMNPs for inflammatory bowel disease treatment and suggest broader biomedical applications.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, Sejong, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
7
|
Yang X, Zhang L, Ran H, Peng F, Tu Y. Micro/nanomotors for active inflammatory disease therapy. Biomater Sci 2025. [PMID: 40181756 DOI: 10.1039/d5bm00052a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Inflammation is a carefully orchestrated response of the immune system to repair injured tissues and clear various damage factors. However, dysregulated inflammation can eventually contribute to the development and progression of various inflammatory diseases. Although anti-inflammatory drugs have demonstrated certain therapeutic efficacy in clinical settings, significant limitations still persist, highlighting the necessity for the development of improved approaches to address complex inflammatory conditions. Micro/nanomotors (MNMs) have shown significant promise for applications in the biomedical field due to their micro/nano-scale sizes and autonomous movement. Unlike traditional nanoparticles, which exhibit passive diffusion in biological fluids, MNMs can convert external energy into a driving force for self-propulsion. This capability not only enhances the tissue penetration depth and retention rates but also facilitates interaction with inflammatory lesions. Recent efforts have suggested that MNMs for inflammatory disease therapy could provide an efficient therapeutic effect. Herein, we mainly introduce the recent advances in inflammatory disease therapy based on MNMs. We conclude by discussing both the obstacles and potential opportunities for MNMs innovations in addressing inflammation.
Collapse
Affiliation(s)
- Xue Yang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Lishan Zhang
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Hui Ran
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Fei Peng
- School of Materials Science and Engineering, Sun Yat-Sen University, Guangzhou 510275, China.
| | - Yingfeng Tu
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China.
| |
Collapse
|
8
|
Peng Y, Xiong RP, Wang B, Chen X, Ning YL, Zhao Y, Yang N, Zhang J, Li CH, Zhou YG, Li P. c-Ski is a novel repressor of NF-κB through interaction with p65 and HDAC1 in U937 cells. Cell Commun Signal 2025; 23:165. [PMID: 40176138 PMCID: PMC11967118 DOI: 10.1186/s12964-025-02178-z] [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: 10/15/2024] [Accepted: 03/26/2025] [Indexed: 04/04/2025] Open
Abstract
The nuclear factor kappa B (NF-κB) signalling pathway plays a crucial role in the regulation of inflammation, and previous research from our lab and others suggests that c-Ski has potential anti-inflammatory effects. However, the role and mechanism of c-Ski, which are related to the regulation of the NF-κB pathway, are still unclear. Here, U937 cells were used, and increasing c-Ski protein levels inhibited inflammatory factor production, invasion, and phagocytosis. The anti-inflammatory effect of c-Ski was similar to that of hormones. Subsequently, immunoprecipitation (IP), Western blot (WB), electrophoretic mobility shift assays (EMSAs), and dual-luciferase reporter assays were used to determine whether increasing c-Ski protein levels could increase c-Ski binding to NF-κB p65 (p65), leading to a decrease in the acetylation level and transcriptional activity of p65. Conversely, decreased p65 expression through targeted small interfering RNA (siRNA) caused the loss of the anti-inflammatory effects of c-Ski. Furthermore, immunoprecipitation confirmed the mutual interaction of c-Ski with HDAC1 and p65, and WB revealed that the anti-inflammatory effect of c-Ski was achieved through the deacetylation of p65 by HDAC1 combined with HDAC1 siRNA and inhibitors. Additionally, through quantitative proteomic analysis, we determined that increasing c-Ski levels had inhibitory effects on the NF-κB pathway. Finally, similar results were also obtained using primary bone marrow-derived macrophages (BMDMs). These findings not only confirm the anti-inflammatory effect of c-Ski but also reveal novel molecular pathways and regulatory molecules of c-Ski, which may be promising targets for direct intervention in the inflammatory response through regulation of c-Ski.
Collapse
Affiliation(s)
- Yan Peng
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Ren-Ping Xiong
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Bo Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Xing Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Ya-Lie Ning
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Yan Zhao
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Nan Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Jing Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Chang-Hong Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Yuan-Guo Zhou
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China
| | - Ping Li
- State Key Laboratory of Trauma, Burn and Combined Injury, Department of Army Occupational Disease, Daping Hospital, Army Medical University (Third Military Medical University), Chongqing, People's Republic of China.
| |
Collapse
|
9
|
Wei J, Zhang X, Sui B, Ding X, Li Y, Liu B, Wang J, Lv X, Zhang Y, Jiang X, Yang Y, Lai H, Liu X, Shi J. Potassium-Doped MnO 2 Nanoparticles Reprogram Neutrophil Calcium Signaling to Accelerate Healing of Methicillin-Resistant Staphylococcus aureus-Infected Diabetic Wounds. ACS NANO 2025; 19:11807-11822. [PMID: 40100101 PMCID: PMC11966767 DOI: 10.1021/acsnano.4c14057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 03/08/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
Neutrophils, as first-line immune cells, typically lose their edge within the diabetic wounds accompanied by methicillin-resistant Staphylococcus aureus (MRSA) infections (the D/M setting), playing the role of "more foe than friend" during the healing process. Specifically, reduced influx of calcium ions (Ca2+) and impaired calcium homeostasis yield the dysfunction of neutrophil sequential behaviors in pathogen killing and wound healing, manifesting as suppressed chemotaxis, decreased intracellular reactive oxygen species (ROS) generation, prolonged apoptosis, and retention of neutrophil extracellular traps (NETs). To address this challenge, this study fabricated potassium (K)-doped manganese dioxide nanoparticles (MnO2 NPs), which activated transmembrane Ca2+ channels by inducing neutrophil depolarization via electron transfer. Subsequently, this contributed to the initial Ca2+ influx and reprogrammed Ca2+-dependent behaviors of impaired neutrophils. Also, the potential antimicrobial capacity of K-MnO2 NPs created a favorable extracellular environment that restored calcium homeostasis, enabling apoptotic neutrophils to be removed timely. Therefore, the wounds treated with K-MnO2 NPs in the D/M setting exhibited potent resistance to MRSA and rapid healing, which could be attributed to the synergistic effects of K-MnO2 NPs in leveraging Ca2+ influx and maintaining calcium homeostasis. In brief, K-MnO2 NPs constitute an effective strategy to resist MRSA and rapid wound healing in the D/M setting.
Collapse
Affiliation(s)
- Jianxu Wei
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Xiaomeng Zhang
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Baiyan Sui
- Department
of Dental Materials, Shanghai Biomaterials Research & Testing
Center, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University 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 200011, China
| | - Xinxin Ding
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Yuan Li
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Beilei Liu
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Jiale Wang
- College
of Physics, Donghua University, Shanghai 201620, China
- Shanghai
Institute of Intelligent Electronics and Systems, Donghua University, Shanghai 201620, China
| | - Xiaolei Lv
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Yi Zhang
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Xue Jiang
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Yijie Yang
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Hongchang Lai
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| | - Xin Liu
- Department
of Dental Materials, Shanghai Biomaterials Research & Testing
Center, Shanghai Ninth People’s Hospital,
Shanghai Jiao Tong University 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 200011, China
| | - Junyu Shi
- Department
of Oral and Maxillofacial Implantology, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University
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, Shanghai 200011, China
| |
Collapse
|
10
|
Zhao X, Wang L, Fu YJ, Yu F, Li K, Wang YQ, Guo Y, Zhou S, Yang W. Inflammatory Microenvironment-Responsive Microsphere Vehicles Modulating Gut Microbiota and Intestinal Inflammation for Intestinal Stem Cell Niche Remodeling in Inflammatory Bowel Disease. ACS NANO 2025; 19:12063-12079. [PMID: 40125581 DOI: 10.1021/acsnano.4c17999] [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: 03/25/2025]
Abstract
Intestinal stem cells (ISCs) engage in proliferation to maintain a stable stem cell population and differentiate into functional epithelial subpopulations. This intricate process is upheld by various signals derived from the host and gut microbiota, establishing an ISC niche. However, during inflammatory bowel disease (IBD), this signaling niche undergoes dramatic changes, leading to impaired ISC and hindered restoration of the damaged intestinal epithelial barrier. This study introduces intestinal inflammatory microenvironment-responsive microsphere vehicles designed to remodel the ISC niche, offering an approach to treat IBD. Using an advanced emulsion technique, these microsphere vehicles specifically target colonic inflammation sites, delivering a responsive release of MXene and l-arginine. This delivery system is formulated to modulate intestinal flora and immune responses effectively. l-arginine is converted into nitric oxide to regulate the gut microbiome, while MXene serves as a nanoimmunomodulator to stabilize immune homeostasis. Our findings demonstrate that the anti-inflammatory properties of the microspheres are key to promoting epithelial repair and remodeling of the ISC niche. This study highlights the role of antioxidant microspheres as anti-inflammatory agents that indirectly support ISC function and gut regeneration.
Collapse
Affiliation(s)
- Xing Zhao
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Liya Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ya-Jun Fu
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Fei Yu
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610032, China
| | - Kai Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041 , China
| | - Yu-Qiang Wang
- Department of Cardiovascular Surgery and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Wei Yang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
11
|
Chen X, Yang Y, Chen J, He Y, Huang Y, Huang Q, Deng W, Zhu R, Huang X, Li T. Dual-driven selenium Janus single-atom nanomotors for autonomous regulating mitochondrial oxygen imbalance to catalytic therapy of rheumatoid arthritis. Redox Biol 2025; 81:103574. [PMID: 40043450 PMCID: PMC11926693 DOI: 10.1016/j.redox.2025.103574] [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: 11/19/2024] [Revised: 02/20/2025] [Accepted: 02/26/2025] [Indexed: 03/22/2025] Open
Abstract
O2 deficiency and excessive reactive oxygen and nitrogen species (RONS) in macrophage mitochondria is a key factor causing oxygen imbalance in rheumatoid arthritis microenvironment (RAM). Although nanocatalytic therapy that simultaneously produce O2 and eliminate RONS offer a novel strategy for RA therapy, the therapeutic efficacy of nanozymes is limited by the lack of autonomous targeting into mitochondria. Herein, we constructed a Janus-structured nanomotor (Pd@MSe) with autonomous targeting ability by embedding Pd single-atom nanozymes into mesoporous selenium (MSe) nanozymes, and obtained a composite nanomotor (Pd@MSe-TPP) with dual-driven forces by modifying with triphenylphosphine (TPP) in MSe hemisphere. In RAM, Pd@MSe-TPP nanomotor achieved autonomously target into macrophages mitochondria with the driven of generation O2 and TPP targeting effect, moreover under the single-atom effect of the Pd nanozymes enhanced electronic transfer between nanozymes, which significantly boosted GPx catalytic activity further effectively enhanced the diffusion of Pd@MSe-TPP nanomotor, thus quickly resorted the oxygen balance. Additionally, while regulating oxygen imbalance, Pd@MSe-TPP nanomotor enable rapidly blocked the inflammatory cascade, restored mitochondrial function and alleviated inflammation, further prevented cartilage degradation and effectively inhibited RA progression. Therefore, the exquisitely designed nanoplatform to regulation arthritic microenvironment provides a new direction for the RA therapy and the clinical translation of nanomedicine.
Collapse
Affiliation(s)
- Xu Chen
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China.
| | - Yang Yang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Jiajun Chen
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, PR China
| | - Yuebing He
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Yukai Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Qidang Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Weiming Deng
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Ruiqi Zhu
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Xuechan Huang
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China
| | - Tianwang Li
- Department of Rheumatology and Immunology, Guangdong Second Provincial General Hospital, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, 510317, PR China; Department of Rheumatology and Immunology, Zhaoqing Central People's Hospital, Zhaoqing, 526000, PR China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, PR China
| |
Collapse
|
12
|
Xu C, Liu M, Xie X, Li Z, Zhu Y, Ye Y, Du M, Hu S, Liu T, Guo Y, Wen W, Liu H, Tu Z. Multifunctional Boron-based 2D Nanoplatforms Ameliorate Severe Respiratory Inflammation by Targeting Multiple Inflammatory Mediators. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412626. [PMID: 39950864 PMCID: PMC11967860 DOI: 10.1002/advs.202412626] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/04/2025] [Indexed: 04/05/2025]
Abstract
Effective management of serious respiratory diseases, such as asthma and recalcitrant rhinitis, remains a global challenge. Here, it is shown that induced sputum supernatants (ISS) from patients with asthma contain higher levels of cell-free DNA (cfDNA) compared to that of healthy volunteers. Although cfDNA scavenging strategies have been developed for inflammation modulation in previous studies, this fall short in clinical settings due to the excessive neutrophil extracellular trap (NET) formation, reactive oxygen and nitrogen species (RONS) and bacterial infections in injured airway tissues. Based on this, a multifunctional boron-based 2D nanoplatform B-PM is designed by coating boron nanosheets (B-NS) with polyamidoamine generation 1 (PG1) dendrimer, which can simultaneously target cfDNA, NETs, RONS, and bacteria. The effects of B-PM in promoting mucosal repair, reducing airway inflammation, and mucus production have been demonstrated in model mice, and the therapeutic effect is superior to dexamethasone. Furthermore, flow cytometry with clustering analysis and transcriptome analysis with RNA-sequencing are adopted to comprehensively evaluate the in vivo anti-inflammation therapeutic effects. These findings emphasize the significance of a multi-targeting strategy to modulate dysregulated inflammation and highlight multifunctional boron-based 2D nanoplatforms for the amelioration of respiratory inflammatory diseases.
Collapse
Affiliation(s)
- Changyi Xu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Department of Clinical LaboratoryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Ming Liu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Xinran Xie
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Zhixin Li
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Yuefei Zhu
- Department of Biomedical EngineeringColumbia UniversityNew York10027USA
| | - Yang Ye
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Mengya Du
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Suhua Hu
- Department of Clinical LaboratoryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Tianrun Liu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Yubiao Guo
- Department of Pulmonary and Critical Care MedicineThe First Affiliated HospitalSun Yat‐Sen UniversityGuangzhouGuangdong510655China
| | - Weiping Wen
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Department of OtolaryngologyThe First Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Huanliang Liu
- Department of Clinical LaboratoryThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| | - Zhaoxu Tu
- Department of OtolaryngologyThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
- Biomedical Innovation CenterThe Sixth Affiliated HospitalSun Yat‐sen UniversityGuangzhouGuangdong510655China
| |
Collapse
|
13
|
Adhikari A, Chen IA. Antibody-Nanoparticle Conjugates in Therapy: Combining the Best of Two Worlds. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2409635. [PMID: 40051146 PMCID: PMC12001320 DOI: 10.1002/smll.202409635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 01/02/2025] [Indexed: 04/17/2025]
Abstract
Monoclonal antibodies (mAbs) and antibody fragments have revolutionized medicine as highly specific binding agents and inhibitors. At the same time, several types of nanomaterials, including liposomes, lipid nanoparticles (NPs), polymersomes, metal and metal oxide NPs, and protein nanostructures, are increasingly utilized and explored for therapeutic potential due to their versatility, chemical and physical properties, and tunability. However, nanomaterials alone often lack specificity, leading to relatively low efficacy and/or high toxicity. To address this problem, a rapidly emerging area is antibody-nanomaterial conjugates (ANCs), which combine the precise targeting specificity of antibodies with the effector functionality of the nanomaterial. In this review, we give a brief introduction to mAbs and major conjugation techniques, describe major classes of nanomaterials being studied for therapeutic potential, and review the literature on ANCs of each class. Special focus is given to emerging applications including ANCs addressing the blood-brain barrier, ANCs delivering nucleic acids, and light-activated ANCs. While many disease targets are related to cancer, ANCs are also under development to address autoimmune, neurological, and infectious diseases. While important challenges remain, ANCs are poised to become a next-generation therapeutic technology.
Collapse
Affiliation(s)
- Aniruddha Adhikari
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| | - Irene A. Chen
- Department of Chemical and Biomolecular EngineeringDepartment of Chemistry and BiochemistryUniversity of CaliforniaLos AngelesCA90049USA
| |
Collapse
|
14
|
Chen Z, Zheng X, Mu Z, Lu W, Zhang H, Yan J. Intelligent Nanomaterials Design for Osteoarthritis Managements. SMALL METHODS 2025:e2402263. [PMID: 40159773 DOI: 10.1002/smtd.202402263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 03/12/2025] [Indexed: 04/02/2025]
Abstract
Osteoarthritis (OA) is the most prevalent degenerative joint disorder, characterized by progressive joint degradation, pain, and diminished mobility, all of which collectively impair patients' quality of life and escalate healthcare expenditures. Current treatment options are often inadequate due to limited efficacy, adverse side effects, and temporary symptom relief, underscoring the urgent need for more effective therapeutic strategies. Recent advancements in nanomaterials and nanomedicines offer promising solutions by improving drug bioavailability, reducing side effects and providing targeted therapeutic benefits. This review critically examines the pathogenesis of OA, highlights the limitations of existing treatments, and explores the latest innovations in intelligent nanomaterials design for OA therapy, with an emphasis on their engineered properties, therapeutic mechanisms, and translational potential in clinical application. By compiling recent findings, this work aims to inspire further exploration and innovation in nanomedicine, ultimately advancing the development of more effective and personalized OA therapies.
Collapse
Affiliation(s)
- Zhihao Chen
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Xuan Zheng
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Zhengzhi Mu
- Key Laboratory of Bionic Engineering, Ministry of Education, Jilin University, Changchun, 130022, China
| | - Weijie Lu
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- Department of Orthopedics, Yanjiang Hospital, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
| | - Haiyuan Zhang
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| | - Jiao Yan
- The Key Laboratory of Advanced Interdisciplinary Studies, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 511436, China
- School of Biomedical Engineering, Guangzhou Medical University, Guangzhou, 511436, China
| |
Collapse
|
15
|
Wu Y, Wang X, Song L, Zhao Z, Xia Y, Tang K, Wang H, Liu J, Wang Z. Tuning macrophage phenotype for enhancing patency rate and tissue regeneration of vascular grafts. Acta Biomater 2025:S1742-7061(25)00239-9. [PMID: 40158766 DOI: 10.1016/j.actbio.2025.03.053] [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: 11/09/2024] [Revised: 03/14/2025] [Accepted: 03/27/2025] [Indexed: 04/02/2025]
Abstract
Macrophages are primary immune cells that play a crucial role in tissue regeneration during the early stages of biomaterial implantation. They create a microenvironment that facilitates cell infiltration, angiogenesis, and tissue remodeling. In the field of vascular tissue engineering, numerous studies have been conducted to modulate the macrophage phenotype by designing various biomaterials, which in turn enhances the regenerative capacity and long-term patency of vascular grafts. However, the mechanism underlying the different phenotypes of macrophages involved in the tissue regeneration of vascular grafts remains unclear. In this study, vascular grafts loaded with various macrophage phenotypes were developed, and their effects were evaluated both in vivo and in vitro. The RAW 264.7 macrophages (M0) were initially treated with LPS or IL-4/IL-10 and polarized into M1 and M2 phenotypes. Subsequently, M0, M1, and M2 macrophages were seeded onto electrospun PCL scaffolds to obtain macrophage-loaded vascular grafts (PCL-M0, PCL-M1, and PCL-M2). As prepared vascular grafts were implanted into the mouse carotid artery for up to one month. The results indicate that the loading of M2 macrophages effectively enhances the patency rate and neotissue formation of vascular grafts. This is achieved through the development of a well-defined endothelium and smooth muscle layer. RNA sequencing was used to investigate the mechanisms of action of different macrophages on tissue regeneration. The study found that M1 macrophages inhibited tissue regeneration by mediating angiogenesis and chronic inflammation through upregulation of VEGFa, IL-1β, and IL-6 expression. In contrast, M2 macrophages regulate the immune microenvironment by upregulating the expression of IL-4 and TGF-β, thereby promoting tissue regeneration. In conclusion, our study demonstrates how different macrophage phenotypes contribute to the initial inflammatory microenvironment surrounding vascular grafts, thereby modulating the biological process of vascular remodeling. STATEMENT OF SIGNIFICANCE: Regulating the biophysical and biochemical characteristics of biomaterials can induce macrophage polarization and enhance vascular remodeling. In previous work, we fabricated a vascular graft with a macroporous structure that promoted macrophage infiltration and polarization into a pro-regenerative phenotype. To illustrate the mechanism, we established a new mouse model and evaluated the effects of different macrophages on vascular regeneration. The study revealed that tuning macrophage phenotype can impact the initial inflammatory microenvironment by secreting cytokines, which can increase the patency rate and regenerative capacity of vascular grafts. These findings provide essential theoretical support for the development of immunoregulatory scaffolds for vascular and other tissue regeneration.
Collapse
Affiliation(s)
- Yifan Wu
- College of Life Sciences, Tiangong University, Tianjin 300387, China; State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Xixi Wang
- College of Life Sciences, Tiangong University, Tianjin 300387, China; Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Lili Song
- College of Life Sciences, Tiangong University, Tianjin 300387, China; Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Zhe Zhao
- College of Life Sciences, Tiangong University, Tianjin 300387, China
| | - Ying Xia
- College of Life Sciences, Tiangong University, Tianjin 300387, China
| | - Kai Tang
- Department of Cardiovascular Surgery, State Key Laboratory of Cardiovascular Disease, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Fuwai Hospital, Beijing 100037, China
| | - Huiquan Wang
- College of Life Sciences, Tiangong University, Tianjin 300387, China
| | - Jing Liu
- Tianjin Key Laboratory of Biomaterial Research, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Zhihong Wang
- Institute of Transplant Medicine, School of Medicine, Key Laboratory of Bioactive Materials of Ministry of Education, Nankai University, Tianjin 300071, China.
| |
Collapse
|
16
|
Saleh MHA, Roh A, Martin K, Mianecki M, Tariq Sheikh A, Singh P, Akorede K, Saunders MN, Shea LD, Decker A, Decker JT. Cargo-less Nanoparticles Prevent Bone Loss in Periodontitis and Peri-implantitis. J Dent Res 2025:220345251319256. [PMID: 40077863 DOI: 10.1177/00220345251319256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025] Open
Abstract
Periodontal and peri-implant diseases are a significant public health problem worldwide, resulting in the destruction of the supporting bone. These bone defects can cause esthetic problems, increased relapse rate, and eventually tooth loss. The etiology of periodontal disease involves an influx of innate immune cells (neutrophils and monocytes) and upregulation of local inflammatory cytokines in the gingiva. Biodegradable polymeric nanoparticles are an inexpensive, safe, and effective means of preventing innate immune activation by bacterial biofilms. We therefore hypothesize that this technology is a potential means of managing periodontal disease. Polylactic acid (PLA) particles were fabricated using an oil-in-water emulsion and used as a therapy in ligature-induced periodontitis and peri-implantitis. Mice were treated daily with nanoparticles or saline control through intravenous injection for 5 or 7 d. Bone loss and quality were characterized using micro-computed tomography and histology, and immune cell infiltrate was characterized by flow cytometry and enzyme-linked immunosorbent assay. PLA particle therapy prevented bone loss in both periodontitis and peri-implantitis. Particle treatment was associated with decreased osteoclast activation. Flow cytometry showed particles were mainly taken up by macrophages and limited inflammatory monocyte recruitment to the ligature site. In vitro evaluation of particle therapy demonstrated the inhibition of toll-like receptor activation during particle treatment. These results extended to monocytes that had been presensitized by titania nanoparticles. Taken together, the results of these experiments demonstrated that cargo-less PLA particle therapy may be a safe, cost-effective therapy to manage inflammatory bone loss in periodontal disease.
Collapse
Affiliation(s)
- M H A Saleh
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - A Roh
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - K Martin
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - M Mianecki
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - A Tariq Sheikh
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - P Singh
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - K Akorede
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - M N Saunders
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - L D Shea
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - A Decker
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
| | - J T Decker
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
17
|
Kou M, Xu Z, Guo Y, Zhang X, Wu M, Chen P, Liu Y, Tang X, Tang Y, Liu W. Development of a Mitochondria-Targeted Ruthenium(II)-Based Phosphorescent Probe for Hypochlorite Detection in Acute Inflammatory Model. Anal Chem 2025; 97:4987-4997. [PMID: 39993269 DOI: 10.1021/acs.analchem.4c05524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The uncontrolled acute inflammatory response triggers dysregulation of the immunoinflammatory system, contributing to the development and progression of various acute inflammatory diseases (AIDs). Hypochlorite (ClO-), as a crucial oxidative mediator in AIDs, accumulates in the inflammatory environment, leading to direct cytotoxicity, secondary injury, and tissue dysfunction. However, achieving rapid detection, accurate tracking, in situ monitoring, and real-time imaging of ClO- in vivo remains a significant challenge. To address these issues, we developed a mitochondria-targeted phosphorescent probe (RuDM), which introduces a ligand containing a C═N bond as a ClO- recognition site to precisely identify ClO- in AIDs. It responds rapidly (6 s) and exhibits long-lived luminescence (471 ns), with a 190-fold luminescence enhancement in monitoring ClO-. Meanwhile, density functional theory (DFT) indicates that the luminescence enhancement of RuCOOH is attributed to the removal of an electron-withdrawing group (diaminomaleonitrile) from RuDM, which leads to an increase in the intersystem crossing rate and a greater probability of radiative transition from the T1 state. Finally, RuDM is used to monitor the levels of exogenous and endogenous ClO- in cells using confocal microscopy imaging and to evaluate its capability for ClO- detection over time in an acute inflammatory model. The above results suggest that RuDM, as a novel molecular platform to detect ClO-, has potential as a practical tool for research on the pathogenesis of acute inflammatory diseases.
Collapse
Affiliation(s)
- Manchang Kou
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Zhongsheng Xu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Yanan Guo
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Xinfeng Zhang
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Meilin Wu
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Peng Chen
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yun Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Xiaoliang Tang
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- Academy of Plateau Science and Sustainability, People's Government of Qinghai Province & Beijing Normal University, College of Chemistry and Chemical Engineering, Qinghai Normal University, Xining 810016, China
| | - Yu Tang
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
| | - Weisheng Liu
- MOE Frontiers Science Center for Rare Isotopes, Key Laboratory of Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Engineering Research Center of Rare Earth Functional Materials, Ministry of Education, State Key Laboratory of Applied Organic Chemistry, College of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou 730000, China
- Academy of Plateau Science and Sustainability, People's Government of Qinghai Province & Beijing Normal University, College of Chemistry and Chemical Engineering, Qinghai Normal University, Xining 810016, China
| |
Collapse
|
18
|
Azadmanesh M, Noorbakhsh MF, Nazifi S, Faraji M. Green Synthesis and Characterization of Silver and Gold Nanoparticles Using Echinophora platyloba Extract and Evaluation of Their Anti-Inflammatory and Antioxidant Properties. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2025; 2025:4421985. [PMID: 40225415 PMCID: PMC11986947 DOI: 10.1155/omcl/4421985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/30/2025] [Indexed: 04/15/2025]
Abstract
This study intends to investigate the green synthesis of silver (Ag) and gold (Au) nanoparticles (NPs) using Echinophora platyloba extract and to evaluate the antioxidant and anti-inflammatory effects of the synthesized NPs and the extract. In this study, aqueous and hydroalcoholic extracts of E. platyloba were prepared, which were used for the biosynthesis of Ag and Au NPs. Dynamic light scattering (DLS), zeta potential analysis, transmission electron microscopy (TEM), Fourier transform infrared (FT-IR) spectroscopy, UV-Vis spectroscopy, and X-ray diffraction (XRD) methods were used to characterize the green NPs. The antioxidant effect of the NPs was estimated using in vitro methods, including reducing power (RP), ferric reducing/antioxidant power (FRAP), and 2,2-diphenyl-1-picrylhydrazyl (DPPH). To evaluate the anti-inflammatory and antioxidant activity of E. platyloba extract and Ag and Au NPs, we used the carrageenan method. In our experiment, the extract and the synthesized NPs were administered orally to the mice 2 h before the carrageenan injection. The subsequent inhibition of inflammation and reduction of paw thickness were quantified. To evaluate their antioxidant effect, malondialdehyde (MDA), and total antioxidant capacity (TAC) levels were measured. Levels of pro-inflammatory cytokines, interleukin-6 (IL-6) and tumor necrosis factor-α (TNF-α), were also quantified. In this study, the results indicate that the synthesized Ag and Au NPs have antioxidant and anti-inflammatory effects. The most promising results were observed in the groups that received the Ag NPs.
Collapse
Affiliation(s)
- Maryam Azadmanesh
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | | | - Saeed Nazifi
- Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Milad Faraji
- Department of Basic Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| |
Collapse
|
19
|
Zhang K, Wang T, Huang X, Wu P, Shen L, Yang Y, Wan W, Sun S, Zhang Z. Ultrasound-mediated nanomaterials for the treatment of inflammatory diseases. ULTRASONICS SONOCHEMISTRY 2025; 114:107270. [PMID: 39961217 PMCID: PMC11875835 DOI: 10.1016/j.ultsonch.2025.107270] [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: 12/12/2024] [Revised: 02/01/2025] [Accepted: 02/11/2025] [Indexed: 03/03/2025]
Abstract
Sterile and infection-associated inflammatory diseases are becoming increasingly prevalent worldwide. Conventional drug therapies often entail significant drawbacks, such as the risk of drug overdose, the development of drug resistance in pathogens, and systemic adverse reactions, all of which can undermine the effectiveness of treatments for these conditions. Nanomaterials (NMs) have emerged as a promising tool in the treatment of inflammatory diseases due to their precise targeting capabilities, tunable characteristics, and responsiveness to external stimuli. Ultrasound (US), a non-invasive and effective treatment method, has been explored in combination with NMs to achieve enhanced therapeutic outcomes. This review provides a comprehensive overview of the recent advances in the use of US-mediated NMs for treating inflammatory diseases. A comprehensive introduction to the application and classification of US was first presented, emphasizing the advantages of US-mediated NMs and the mechanisms through which US and NMs interact to enhance anti-inflammatory therapy. Subsequently, specific applications of US-mediated NMs in sterile and infection-associated inflammation were summarized. Finally, the challenges and prospects of US-mediated NMs in clinical translation were discussed, along with an outline of future research directions. This review aims to provide insights to guide the development and improvement of US-mediated NMs for more effective therapeutic interventions in inflammatory diseases.
Collapse
Affiliation(s)
- Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Xingyong Huang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China
| | - Wenyu Wan
- Key Laboratory of Immunodermatology, Ministry of Education, Department of Dermatology, The First Hospital of China Medical University, PR China; Key Laboratory of Immunodermatology, National Health Commission of the People's Republic of China, The First Hospital of China Medical University, PR China; National and Local Joint Engineering Research Center of Immunodermatological Theranostics, The First Hospital of China Medical University, PR China.
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Shengjing Hospital of China Medical University, PR China; Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, PR China.
| | - Zhan Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, PR China; Cancer Stem Cell and Translational Medicine Laboratory, Shengjing Hospital of China Medical University, Shenyang, PR China.
| |
Collapse
|
20
|
Xu F, Zhao G, Gong Y, Liang X, Yu M, Cui H, Xie L, Zhu N, Zhu X, Shao X, Qi K, Lu B, Tu J, Na S. Enhancement of Osseointegration via Endogenous Electric Field by Regulating the Charge Microenvironments around Implants. Adv Healthc Mater 2025; 14:e2403388. [PMID: 39757756 PMCID: PMC11874649 DOI: 10.1002/adhm.202403388] [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: 09/08/2024] [Revised: 11/19/2024] [Indexed: 01/07/2025]
Abstract
The regulation of the charged microenvironment around implants is an effective way to promote osseointegration. Although homeostasis of the charged microenvironment plays an integral role in tissues, current research is externally invasive and unsuitable for clinical applications. In this study, functional materials with different surface potential differences are prepared by changing the spatial layout of Ta and Ag on the surface of a Ti-6Al-4V alloy (TC4). This naturally formed an endogenous electric field (EEF) with a negatively charged cell membrane after in vivo implantation and promoted osseointegration at the interface between the bone and implant through the upregulation of Ca2+ concentration and activation of subsequent pathways. Interestingly, the promotion of stem cell differentiation, regulation of the direction of immune cell polarization, and antibacterial efficacy are determined by the free charge contained in the implant, rather than by the magnitude of the surface potential difference. This functional implant represents a unique strategy for regulating the charged microenvironment around the implant and enhancing osseointegration, thereby providing ideas and technical approaches for the clinical development of novel implant materials.
Collapse
Affiliation(s)
- Fangfang Xu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
- Department of Oral and Maxillofacial SurgeryCollege of StomatologyXi'an Jiaotong UniversityXi'an710004China
| | - Guangbin Zhao
- State Key Laboratory for Manufacturing System EngineeringSchool of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
| | - Yuxin Gong
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
| | - Xiang Liang
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
- Department of Oral and Maxillofacial SurgeryCollege of StomatologyXi'an Jiaotong UniversityXi'an710004China
| | - Ming Yu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
| | - Hao Cui
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
- Department of Oral and Maxillofacial SurgeryCollege of StomatologyXi'an Jiaotong UniversityXi'an710004China
| | - Linyang Xie
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
- Department of Oral and Maxillofacial SurgeryCollege of StomatologyXi'an Jiaotong UniversityXi'an710004China
| | - Nan Zhu
- State Key Laboratory for Manufacturing System EngineeringSchool of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
| | - Xuan Zhu
- State Key Laboratory for Manufacturing System EngineeringSchool of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
| | - Xiaoxi Shao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and RegenerationNational Clinical Research Center for Oral DiseasesShaanxi Clinical Research Center for Oral DiseasesDepartment of Oral and Maxillofacial SurgerySchool of StomatologyThe Fourth Military Medical University145 West Changle RoadXi'an710032China
| | - Kun Qi
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
- Department of OrthodonticsCollege of StomatologyXi'an Jiaotong UniversityXi'an710004China
| | - Bingheng Lu
- State Key Laboratory for Manufacturing System EngineeringSchool of Mechanical EngineeringXi'an Jiaotong UniversityXi'an710054China
| | - Junbo Tu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
- Department of Oral and Maxillofacial SurgeryCollege of StomatologyXi'an Jiaotong UniversityXi'an710004China
| | - Sijia Na
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine ResearchCollege of StomatologyXi’ an Jiaotong UniversityXi’ an710004China
- Clinical Research Center of Shaanxi Province for Dental and Maxillofacial DiseasesCollege of StomatologyXi'an Jiaotong UniversityXi’ an710004China
- Department of Oral and Maxillofacial SurgeryCollege of StomatologyXi'an Jiaotong UniversityXi'an710004China
| |
Collapse
|
21
|
Xie L, Wu H, Li Y, Shi L, Liu Y. Recent Development of Nanozymes for Combating Bacterial Drug Resistance: A Review. Adv Healthc Mater 2025; 14:e2402659. [PMID: 39388414 DOI: 10.1002/adhm.202402659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 08/31/2024] [Indexed: 10/12/2024]
Abstract
The World Health Organization has warned that without effective action, deaths from drug-resistant bacteria can exceed 10 million annually, making it the leading cause of death. Conventional antibiotics are becoming less effective due to rapid bacterial drug resistance and slowed new antibiotic development, necessitating new strategies. Recently, materials with catalytic/enzymatic properties, known as nanozymes, have been developed, inspired by natural enzymes essential for bacterial eradication. Unlike recent literature reviews that broadly cover nanozyme design and biomedical applications, this review focuses on the latest advancements in nanozymes for combating bacterial drug resistance, emphasizing their design, structural characteristics, applications in combination therapy, and future prospects. This approach aims to promote nanozyme development for combating bacterial drug resistance, especially towards clinical translation.
Collapse
Affiliation(s)
- Lingping Xie
- The People's Hospital of Yuhuan, Taizhou, Zhejiang, 317600, China
| | - Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin, 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, China
| | - Yuanfeng Li
- Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| | - Yong Liu
- The People's Hospital of Yuhuan, Taizhou, Zhejiang, 317600, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300071, China
| |
Collapse
|
22
|
Gao S, Li H, Li Z, Wang H, Li X, Yang S, Huang L, Zhang B, Zhang K, Tsoi JKH, He J, Dissanayaka WL. Multifunctional Injectable Bioadhesive with Toll-like Receptor 4 and Myeloid Differentiation Factor 2 Antagonistic Anti-inflammatory Potential for Periodontal Regeneration. ACS NANO 2025; 19:7098-7116. [PMID: 39951685 PMCID: PMC11867008 DOI: 10.1021/acsnano.4c15922] [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: 11/07/2024] [Revised: 01/28/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025]
Abstract
Effectively addressing inflammation in periodontitis is challenging as conventional injectable hydrogels typically require the addition of drugs to provide sufficient anti-inflammatory effects. To overcome this limitation, we developed a multifunctional injectable hydrogel with inherent properties that antagonize the Toll-like receptor 4 and myeloid differentiation factor 2 complex (TLR4-MD2). This hydrogel allows for direct inhibition of inflammatory pathways without the need for additional drugs. We identified xylitol, caffeic acid, and citric acid as natural materials that effectively meet biological needs for anti-inflammatory and antibacterial effects as well as support bone regeneration. With this in mind, we developed a caffeic-acid-modified poly(xylitol succinate) (PXS)-based iCPC@MgO composite hydrogel and tested its potential application for periodontal regeneration. The iCPC@MgO hydrogel demonstrated rapid wet tissue adhesion and injectability, which are ascribed to incorporating catechol groups derived from caffeic acid. Intriguingly, the PXS polymer used for synthesizing the hydrogel was found to possess anti-inflammatory properties and act as an antagonist for the TLR4-MD2 complex. This hydrogel also exhibited outstanding antibacterial efficiency against Porphyromonas gingivalis and Aggregatibacter actinomycetemcomitans by stimulating antibiotic synthesis within bacteria and disrupting bacterial cell walls. In a periodontitis mouse model, the iCPC@MgO hydrogel demonstrated the therapeutic potential of reducing inflammatory factors, inhibiting dominant periodontitis-associated bacteria, and maintaining subgingival microbiota balance in addition to the regenerative effects. These properties, combined with their ecofriendly nature, firmly established the iCPC@MgO hydrogel as a highly promising option for use in periodontitis therapy as well as in tissue healing, repair, and regeneration in various other inflammatory conditions.
Collapse
Affiliation(s)
- Shuting Gao
- Applied
Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Huihua Li
- Applied
Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Zekun Li
- Department
of Chemistry, Faculty of Science, The University
of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Hong Wang
- Applied
Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Xinyue Li
- State
Key Laboratory of Applied Organic Chemistry and Key Laboratory of
Nonferrous Metal Chemistry and Resources Utilization of Gansu Province, Lanzhou University, Lanzhou 730000, China
| | - Shengyan Yang
- Applied
Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Lin Huang
- Applied
Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Baoping Zhang
- Department
of Stomatology Lanzhou University, Lanzhou University, Lanzhou 730000, China
| | - Kailiang Zhang
- Department
of Stomatology Lanzhou University, Lanzhou University, Lanzhou 730000, China
| | - James Kit Hon Tsoi
- Applied
Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Jian He
- Department
of Chemistry, Faculty of Science, The University
of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| | - Waruna Lakmal Dissanayaka
- Applied
Oral Sciences & Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong, Hong Kong SAR 999077, China
| |
Collapse
|
23
|
Xavier-de-Britto I, Gomes-da-Silva NC, Gomes Soares MA, Follmer C, Dabkiewicz D, Alencar LMR, Sant’Anna C, Ferreira TPT, Martins PRES, Ricci-Junior E, Fechine PBA, Santos-Oliveira R. Therapeutic Potential of Arimoclomol Nanomicelles: In Vitro Impact on Alzheimer's and Parkinson's Pathology and Correlation with In Vivo Inflammatory Response. ACS Chem Neurosci 2025; 16:699-710. [PMID: 39907698 PMCID: PMC11843614 DOI: 10.1021/acschemneuro.4c00734] [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: 10/29/2024] [Revised: 01/23/2025] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
This study investigates the potential of arimoclomol-loaded nanomicelles for the treatment of neurodegenerative diseases like Alzheimer's and Parkinson's, as well as their anti-inflammatory properties. Arimoclomol, a coinducer of heat shock proteins (HSPs), has shown clinical promise in mitigating protein misfolding, a hallmark of these diseases. In this work, arimoclomol nanomicelles significantly reduced the aggregation of β-amyloid (Aβ1-42) and α-synuclein (α-syn), key pathological proteins in Alzheimer's and Parkinson's. Additionally, the nanomicelles demonstrated potent anti-inflammatory effects, reducing leukocyte and neutrophil counts in an acute inflammation model. These results suggest that arimoclomol nanomicelles could enhance clinical outcomes by targeting both neurodegenerative and inflammatory processes, offering a promising therapeutic strategy for long-term disease management.
Collapse
Affiliation(s)
- Isabelle Xavier-de-Britto
- Brazilian
Nuclear Energy Commission, Nuclear Engineering
Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro, Rio de Janeiro 21941906, Brazil
| | - Natália Cristina Gomes-da-Silva
- Brazilian
Nuclear Energy Commission, Nuclear Engineering
Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro, Rio de Janeiro 21941906, Brazil
| | - Marilia Amável Gomes Soares
- Brazilian
Nuclear Energy Commission, Nuclear Engineering
Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro, Rio de Janeiro 21941906, Brazil
| | - Cristian Follmer
- Laboratory
of Biological Chemistry of Neurodegenerative Disorders, Department
of Physical Chemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - David Dabkiewicz
- Laboratory
of Biological Chemistry of Neurodegenerative Disorders, Department
of Physical Chemistry, Institute of Chemistry, Federal University of Rio de Janeiro, Rio de Janeiro 21941-909, Brazil
| | - Luciana Magalhães Rebelo Alencar
- Biophysics
and Nanosystems Laboratory, Federal University of Maranhão, Department of Physics, São Luis, Maranhão 65065690, Brazil
| | - Celso Sant’Anna
- Laboratory
of Microscopy Applied to Life Science–Lamav, National Institute of Metrology, Quality and Technology, Duque de Caxias, Rio de
Janeiro 25250-020, Brazil
| | | | | | - Eduardo Ricci-Junior
- Federal
University of Rio de Janeiro, School of
Pharmacy, Rio de Janeiro, Rio de Janeiro 21941900, Brazil
| | - Pierre Basílio Almeida Fechine
- Group
of Chemistry of Advanced Materials (GQMat)–Department of Analytical
Chemistry and Physical-Chemistry, Federal
University of Ceará, Fortaleza, Ceará 451-970, Brazil
| | - Ralph Santos-Oliveira
- Brazilian
Nuclear Energy Commission, Nuclear Engineering
Institute, Laboratory of Nanoradiopharmacy and Synthesis of New Radiopharmaceuticals, Rio de Janeiro, Rio de Janeiro 21941906, Brazil
- Rio
de Janeiro State University, Laboratory
of Radiopharmacy and Nanoradiopharmaceuticals, Rio de Janeiro 23070200, Rio de Janeiro, Brazil
| |
Collapse
|
24
|
Zhang L, Fu X, Li J, Xiao W, Xiong X, Lv H, Zhang Z, Ju J. Treatment of Acute Ulcerative Colitis with Zinc Hyaluronate in Mice. J Microbiol Biotechnol 2025; 35:e2408050. [PMID: 39947703 DOI: 10.4014/jmb.2408.08050] [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: 08/28/2024] [Revised: 12/06/2024] [Accepted: 12/06/2024] [Indexed: 03/06/2025]
Abstract
Ulcerative colitis (UC) is a type of inflammatory bowel disease arising from numerous factors, while UC patients face insufficient treatment options and a high incidence of adverse reactions to the current therapies. As a functional food additive, hyaluronic acid plays a certain role in intestinal repair. In this study, we constructed a mouse model of dextran sulfate sodium (DSS)-induced UC to examine the effects and underlying mechanisms of action of zinc hyaluronate (ZnHA) on the pathogenesis of UC. ZnHA effectively alleviated key clinical UC symptoms, such as weight loss, loose stools, and bloody stools. Mechanistically, ZnHA attenuated the expression of inflammatory factors, such as tumor necrosis factor-α, interleukin (IL)-6, and myeloperoxidase while upregulating the expression of IL-10. Furthermore, through intestinal flora and short-chain fatty acid analyses, ZnHA was found to promote propionic acid production by enriching beneficial bacteria. ZnHA simultaneously enhanced the expression of tight junction proteins, specifically ZO-1 and occludin, thereby restoring intestinal barrier function. Overall, our findings elucidate the therapeutic potential of ZnHA in treating acute UC by inhibiting intestinal inflammation and regulating flora, while also providing further theoretical support for development of hyaluronic acid to treat this disease.
Collapse
Affiliation(s)
- Lan Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, P.R. China
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
| | - Xuedan Fu
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, P.R. China
| | - Jiazheng Li
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, P.R. China
| | - Wan Xiao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, P.R. China
| | - Xi Xiong
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, P.R. China
| | - Huixia Lv
- Department of Pharmaceutics, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, P.R. China
| | - Zhenhai Zhang
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, P.R. China
| | - Jianming Ju
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, P.R. China
- Jiangsu Province Academy of Traditional Chinese Medicine, Nanjing 210023, P.R. China
| |
Collapse
|
25
|
Morsy HM, Zaky MY, Yassin NYS, Khalifa AYZ. Nanoparticle-based flavonoid therapeutics: Pioneering biomedical applications in antioxidants, cancer treatment, cardiovascular health, neuroprotection, and cosmeceuticals. Int J Pharm 2025; 670:125135. [PMID: 39732216 DOI: 10.1016/j.ijpharm.2024.125135] [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: 09/05/2024] [Revised: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Flavonoids, a type of natural polyphenolic molecule, have garnered significant research interest due to their ubiquitous nature and diverse biological activities, including antioxidant, anti-inflammatory, and anticancer effects, making them appealing to various scientific disciplines. In this regard, the use of a flavonoid nanoparticle delivery system is to overcome low bioavailability, bioactivity, poor aqueous solubility, systemic absorption, and intensive metabolism. Therefore, this review summarizes the classification of nanoparticles (liposomes, polymeric, and solid lipid nanoparticles) and the advantages of using nanoparticle-flavonoid formulations to boost flavonoid bioavailability. Moreover, this review illustrated the pioneering biomedical applications of nanoparticle-based flavonoid therapeutics, as well as safety and toxicity considerations of using a flavonoid nanoparticle delivery system.
Collapse
Affiliation(s)
- Hadeer M Morsy
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Mohamed Y Zaky
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt.
| | - Nour Y S Yassin
- Molecular Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, P.O.Box 62521, Beni-Suef, Egypt
| | - Ashraf Y Z Khalifa
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, Saudi Arabia.
| |
Collapse
|
26
|
Qin W, Ma Z, Bai G, Qin W, Li L, Hao D, Wang Y, Yan J, Han X, Niu W, Niu L, Jiao K. Neurovascularization inhibiting dual responsive hydrogel for alleviating the progression of osteoarthritis. Nat Commun 2025; 16:1390. [PMID: 39910066 PMCID: PMC11799281 DOI: 10.1038/s41467-025-56727-8] [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: 05/22/2024] [Accepted: 01/28/2025] [Indexed: 02/07/2025] Open
Abstract
Treating osteoarthritis (OA) associated pain is a challenge with the potential to significantly improve patients lives. Here, we report on a hydrogel for extracellular RNA scavenging and releasing bevacizumab to block neurovascularization at the osteochondral interface, thereby mitigating OA pain and disease progression. The hydrogel is formed by cross-linking aldehyde-phenylboronic acid-modified sodium alginate/polyethyleneimine-grafted protocatechuic acid (OSAP/PPCA) and bevacizumab sustained-release nanoparticles (BGN@Be), termed OSPPB. The dynamic Schiff base bonds and boronic ester bonds allow for injectability, self-healing, and pH/reactive oxygen species dual responsiveness. The OSPPB hydrogel can significantly inhibit angiogenesis and neurogenesis in vitro. In an in vivo OA model, intraarticular injection of OSPPB accelerates the healing process of condyles and alleviates chronic pain by inhibiting neurovascularization at the osteochondral interface. The injectable hydrogel represents a promising technique to treat OA and OA associated pain.
Collapse
Affiliation(s)
- Wenpin Qin
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Zhangyu Ma
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Guo Bai
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- College of Stomatology, Shanghai Jiao Tong University, Shanghai, 200011, China
- 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
| | - Wen Qin
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Ling Li
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Dongxiao Hao
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Yuzhu Wang
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Jianfei Yan
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Xiaoxiao Han
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China
| | - Wen Niu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
| | - Lina Niu
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
| | - Kai Jiao
- Department of Stomatology, Tangdu Hospital, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
- State Key Laboratory of Oral and Maxillofacial Reconstruction and Regeneration, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
- National Clinical Research Center for Oral Diseases, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
- Shaanxi Key Laboratory of Stomatology, School of Stomatology, The Fourth Military Medical University, Xi'an, 710000, Shaanxi, China.
| |
Collapse
|
27
|
Zeng J, Zhao Q, Xiong Z, Zhang S, Deng S, Liu D, Zhang X. Surface functionalization of two-dimensional nanomaterials beyond graphene: Applications and ecotoxicity. Adv Colloid Interface Sci 2025; 336:103357. [PMID: 39612722 DOI: 10.1016/j.cis.2024.103357] [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: 05/27/2024] [Revised: 10/22/2024] [Accepted: 11/18/2024] [Indexed: 12/01/2024]
Abstract
Two dimensional (2D) nanomaterials have emerged as promising candidates in nanotechnology due to their excellent physical, chemical, and electronic properties. However, they also pose challenges such as environmental instability and low biosafety. To address these issues, researchers have been exploring various surface functionalization methods to enhance the performance of 2D nanomaterials in practical applications. Moreover, when released into the environment, these 2D nanomaterials may interact with natural organic matter (NOM). Both intentional surface modification and unintentional environmental corona formation can alter the structure and physicochemical properties of 2D nanomaterials, potentially affecting their ecological toxicity. This review provides a comprehensive overview of covalent functionalization strategies and non-covalent interactions of 2D nanomaterials beyond graphene with organic substances, examining the resultant changes in material properties after modification. Covalent functionalization methods discussed include nucleophilic substitution reactions, addition reactions, condensation, and coordination. Non-covalent interactions are classified by substance type, covering interactions with NOM, in vivo biomolecules, and synthetic compounds. In addition, the review delves into the effects of surface functionalization on the toxicity of 2D nanomaterials to bacteria and algae. This discussion contributes to a foundational understanding for assessing the potential ecological risks associated with 2D nanomaterials.
Collapse
Affiliation(s)
- Jin Zeng
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qing Zhao
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Zhiqiang Xiong
- State Key Laboratory of Radiation Medicine and Protection, School of Radiological and Interdisciplinary Sciences (RAD-X), Suzhou Medical College, Soochow University, Suzhou 215123, China
| | - Siyu Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China
| | - Shuo Deng
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Daxu Liu
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xuejiao Zhang
- Key Laboratory of Pollution Ecology and Environmental Engineering, Institute of Applied Ecology, Chinese Academy of Sciences, Shenyang 110016, China; National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, China.
| |
Collapse
|
28
|
Álvarez-López A, Tabraue-Rubio R, Daza R, Colchero L, Guinea GV, Cohen-Solal M, Pérez-Rigueiro J, González-Nieto D. Osteoblastic Differentiation and Mitigation of the Inflammatory Response in Titanium Alloys Decorated with Oligopeptides. Biomimetics (Basel) 2025; 10:58. [PMID: 39851774 PMCID: PMC11760451 DOI: 10.3390/biomimetics10010058] [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: 12/19/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Under benign conditions, bone tissue can regenerate itself without external intervention. However, this regenerative capacity can be compromised by various factors, most importantly related with the extent of the injury. Critical-sized defects, exceeding the body's natural healing ability, demand the use of temporary or permanent devices like artificial joints or bone substitutes. While titanium is a widely used material for bone replacement, its integration into the body remains limited. This often leads to the progressive loosening of the implant and the need for revision surgeries, which are technically challenging, are commonly associated with high complication rates, and impose a significant economic burden. To enhance implant osseointegration, numerous studies have focused on the development of surface functionalization techniques to improve the response of the body to the implant. Yet, the challenge of achieving reliable and long-lasting prostheses persists. In this work, we address this challenge by applying a robust and versatile biofunctionalization process followed by the decoration of the material with oligopeptides. We immobilize four different peptides (RGD, CS-1, IKVAV, PHSRN) on R-THAB® functionalized surfaces and find them to be highly stable in the long term. We also find that RGD is the best-performing peptide in in vitro cell cultures, enhancing adhesion, proliferation, and osteogenic differentiation of mesenchymal stem cells. To assess the in vivo effect of RGD-decorated Ti-6Al-4V implants, we develop a calvarial model in murine hosts. We find that the RGD-decoration remains stable for 1 week after the surgical procedure and reduces post-implantation macrophage-related inflammation. These results highlight the potential of peptide decoration on R-THAB® functionalized surfaces to expedite the development of novel metallic biomaterials with enhanced biocompatibility properties, thereby advancing the field of regenerative medicine.
Collapse
Affiliation(s)
- Aroa Álvarez-López
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (A.Á.-L.); (R.D.); (L.C.); (G.V.G.)
- Departamento de Ciencia de Materiales, Escuela Técnica Superior de Ingenieros Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Raquel Tabraue-Rubio
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (A.Á.-L.); (R.D.); (L.C.); (G.V.G.)
- Departamento de Ciencia de Materiales, Escuela Técnica Superior de Ingenieros Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Rafael Daza
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (A.Á.-L.); (R.D.); (L.C.); (G.V.G.)
- Departamento de Ciencia de Materiales, Escuela Técnica Superior de Ingenieros Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Luis Colchero
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (A.Á.-L.); (R.D.); (L.C.); (G.V.G.)
- Departamento de Ciencia de Materiales, Escuela Técnica Superior de Ingenieros Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| | - Gustavo V. Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (A.Á.-L.); (R.D.); (L.C.); (G.V.G.)
- Departamento de Ciencia de Materiales, Escuela Técnica Superior de Ingenieros Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Profesor Martín Lagos s/n, 28040 Madrid, Spain
| | - Martine Cohen-Solal
- INSERM U1132 Bioscar, Université de Paris Cité, 75006 Paris, France;
- Department of Rheumatology, APHP, Lariboisière Hospital, 75010 Paris, France
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (A.Á.-L.); (R.D.); (L.C.); (G.V.G.)
- Departamento de Ciencia de Materiales, Escuela Técnica Superior de Ingenieros Caminos, Canales y Puertos, Universidad Politécnica de Madrid, 28040 Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Profesor Martín Lagos s/n, 28040 Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Pozuelo de Alarcón, 28223 Madrid, Spain; (A.Á.-L.); (R.D.); (L.C.); (G.V.G.)
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), 28029 Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería, ETSI Telecomunicaciones, Universidad Politécnica de Madrid, 28040 Madrid, Spain
| |
Collapse
|
29
|
Soleimani Z, Baharifar H, Najmoddin N, Khoshnevisan K. Evaluation of Carboxymethyl Cellulose/Gelatin Hydrogel-Based Dressing Containing Cefdinir for Wound Healing Promotion in Animal Model. Gels 2025; 11:38. [PMID: 39852009 PMCID: PMC11765047 DOI: 10.3390/gels11010038] [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: 12/04/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/26/2025] Open
Abstract
The skin serves as a critical barrier against external pathogens, and its wound healing is a complex biological process that requires careful management to ensure optimal tissue regeneration. Hydrogels, a class of hydrophilic polymers, have emerged as promising materials for wound dressings due to their biocompatibility, biodegradability, and ability to create a moist wound environment conducive to cell proliferation and migration. In this research, a hydrogel dressing containing cefdinir (Cef) was made from a combination of carboxymethyl cellulose (CMC) and gelatin (Gel) by a physical crosslinking method, and their physicochemical, mechanical, and biological properties were investigated. Results show that the addition of Cef does not cause a significant change in the morphology or the tensile strength of the wound dressing. The swelling and degradation rate of the hydrogel slightly increased in the presence of Cef. The presence of Cef enhanced antibacterial effects up to 2.5-fold against P. aeruginosa (35 mm), S. aureus (36 mm), and S. pyogenes (35 mm). The results of the cytotoxicity test showed the absence of cytotoxicity in both drug-containing and drug-free wound dressings, as well as a survival rate of over 75% in cells after 48 h. The drug-containing wound dressing accelerates the formation of the epidermis layer and the production of fibroblast cells, and as a result, accelerates the wound healing process. The percentage of wound healing on the ninth day of treatment for an untreated wound was 30%, while this percentage was 40% with a wound dressing without medicine and 60% with a wound dressing containing medicine, and on the fifteenth day of treatment, the wound treated with both wound dressings had more than 85% healing. As a result, it is possible to use CMC/Gel hydrogel polymeric wound dressing containing Cef as a wound dressing for wound healing, according to the desired physicochemical properties and biocompatibility.
Collapse
Affiliation(s)
- Zahra Soleimani
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Hadi Baharifar
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
- Applied Biophotonics Research Center, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Najmeh Najmoddin
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | - Kamyar Khoshnevisan
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1983963113, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1983963113, Iran
- Research and Development Team, Evolution Wound Dressing (EWD) Startup Co., Tehran 1983963113, Iran
| |
Collapse
|
30
|
Qian Y, Ding J, Zhao R, Song Y, Yoo J, Moon H, Koo S, Kim JS, Shen J. Intrinsic immunomodulatory hydrogels for chronic inflammation. Chem Soc Rev 2025; 54:33-61. [PMID: 39499495 DOI: 10.1039/d4cs00450g] [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: 11/07/2024]
Abstract
The immune system plays a pivotal role in maintaining physiological homeostasis and influencing disease processes. Dysregulated immune responses drive chronic inflammation, which in turn results in a range of diseases that are among the leading causes of death globally. Traditional immune interventions, which aim to regulate either insufficient or excessive inflammation, frequently entail lifelong comorbidities and the risk of severe side effects. In this context, intrinsic immunomodulatory hydrogels, designed to precisely control the local immune microenvironment, have recently attracted increasing attention. In particular, these advanced hydrogels not only function as delivery mechanisms but also actively engage in immune modulation, optimizing interactions with the immune system for enhanced tissue repair, thereby providing a sophisticated strategy for managing chronic inflammation. In this tutorial review, we outline key elements of chronic inflammation and subsequently explore the strategic design principles of intrinsic immunomodulatory hydrogels based on these elements. Finally, we examine the challenges and prospects of such immunomodulatory hydrogels, which are expected to inspire further preclinical research and clinical translation in addressing chronic inflammation.
Collapse
Affiliation(s)
- Yuna Qian
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| | - Jiayi Ding
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Institute of Imaging Diagnosis and Minimally Invasive Intervention, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Rui Zhao
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| | - Yang Song
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610065, China
| | - Jiyoung Yoo
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Huiyeon Moon
- Department of Chemistry, Korea University, Seoul 02841, Korea
| | - Seyoung Koo
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jong Seung Kim
- Department of Chemical and Molecular Engineering, Hanyang University ERICA, Ansan, Gyeonggi-do 15588, Korea.
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China.
| |
Collapse
|
31
|
Zhang Y, Yang N, Wang L, Zheng Y, Dong Z, Wu J, Zhang G, Zhang Y, Qiu J, Wang W, Wang X, Liang P. Anti-inflammatory and heat shock protein-inhibiting nanoplatform for synergetic cancer chemo/photothermal therapy. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2025; 63:102801. [PMID: 39667417 DOI: 10.1016/j.nano.2024.102801] [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: 04/28/2024] [Revised: 08/20/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024]
Abstract
Photothermal therapy is a novel and promising method for cancer treatment due to its controllable property, noninvasive nature, and high selectivity. Nevertheless, tumor recurrence of inflammatory response and tumor tolerance of heat shock protein over-expression remain serious challenges in current photothermal therapy. Additionally, the high dosage requirement of nanomaterial for optimal imaging and therapeutic effect would result in various side effects, organ excretion burdens, and long-term accumulation in the body. In this work, RD/Qu nanoplatform is designed and prepared with near-infrared (NIR) absorbance, high photothermal conversion efficiency, and great chemotherapy effect for synergetic cancer chemo/photothermal therapy at an ultralow-dose. More importantly, both in vitro and in vivo studies demonstrate that it could decrease the expression of HSP70 to fight hyperthermia tumor tolerance and inhibit inflammatory factor COX-2 to suppress tumor recurrence. Therefore, the RD/Qu nanoparticles show excellent outcome in tumor ablation at a quite low dosage, providing a promising avenue for cancer treatment.
Collapse
Affiliation(s)
- Yuanying Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Nan Yang
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), Nanjing 211816, China
| | - Lingling Wang
- Department of Vascular Surgery of the First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230002, China
| | - Yi Zheng
- Department of Vascular Surgery of the First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230002, China
| | - Ziyi Dong
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Jiahui Wu
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Gege Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Yanling Zhang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China
| | - Jianda Qiu
- Department of Vascular Surgery of the First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230002, China
| | - Wenbin Wang
- Department of Vascular Surgery of the First Affiliated Hospital of Anhui Medical University, Anhui Public Health Clinical Center, Hefei 230002, China.
| | - Xianwen Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Medical University, Hefei, Anhui 230032, China.
| | - Pingping Liang
- School of Life Sciences, Anhui Medical University, Hefei, Anhui 230032, China.
| |
Collapse
|
32
|
Liu X, Zhu J, Zeng J, Qiao M, Wang H, Shen M, Meng D. Phenolic Acids Derivatives from Meehania Fargesii with Anti-inflammation Effects. Chem Biodivers 2025; 22:e202401414. [PMID: 39261298 DOI: 10.1002/cbdv.202401414] [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: 06/07/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/13/2024]
Abstract
Utilizing systematic plant chemistry separation techniques, two previously unreported phenolic acids (1-2) and eighteen (3-20) phenolic acids were isolated from 70 % ethanol extract of Meehania fargesii var. radicans (Vaniot) C.Y.Wu. The structures of the unreported compounds were determined using spectroscopic methods, including 1D and 2D NMR, ECD, and HR-ESI-MS. The activity of all isolated phenolic acids was evaluated for their ability to inhibit nitric oxide (NO) production in RAW264.7 cells activated by lipopolysaccharide (LPS). Among the isolated compounds, compounds 1 and 19 exhibited significant inhibitory activity against NO production in RAW264.7 cells, with higher concentrations being more active than the indomethacin, without displaying cytotoxicity. This study provides a basis for the application and development of M. fargesii and the discovery of natural anti-inflammatory drugs.
Collapse
Affiliation(s)
- Xingpeng Liu
- Guizhou Institute for Food and Drug Control, Guiyang, 550081, China
| | - Jialiang Zhu
- National Institutes for Food and Drug Control, Beijing, 100050, China
| | - Jun Zeng
- NMPA Key Laboratory for Quality Monitoring of Narcotic Drugs and Psychotropic substances, Chongging institute for Food and Drug Control, Chongqing, 401121, China
| | - Mingming Qiao
- NMPA Key Laboratory for Quality Monitoring of Narcotic Drugs and Psychotropic substances, Chongging institute for Food and Drug Control, Chongqing, 401121, China
| | - Hanchuan Wang
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, Shenyang, 110016, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Mingyi Shen
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, Shenyang, 110016, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| | - Dali Meng
- Key Laboratory of Ethnomedicine Material Basis & Pharmacological Mechanisms, Shenyang, 110016, China
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
33
|
Kong J, Yao Z, Stelzel JL, Yang Y, Chen J, Feng H, Schmidt C, Zhang C, Krishnan K, Chen L, Pan J, Ding K, Zhu Y, Li X, Doloff JC, Mao H, Reddy SK. Granular Nanofiber-Hydrogel Composite-Programmed Regenerative Inflammation and Adipose Tissue Formation. Adv Healthc Mater 2025; 14:e2403094. [PMID: 39580666 PMCID: PMC11773126 DOI: 10.1002/adhm.202403094] [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: 08/17/2024] [Revised: 11/04/2024] [Indexed: 11/26/2024]
Abstract
The interplay between biomaterials and host immune responses critically determines outcomes in tissue restoration. Recent studies suggest that physicochemical properties of materials can dictate pro-regenerative versus pro-fibrotic responses and have begun to define the key immune cell types and signals governing these divergent effects. This emerging understanding enables the engineering of regenerative biomaterials capable of functional restoration in situ. An injectable nanofiber-hydrogel composite (NHC) microparticles are designed and constructed from cross-linked electrospun collagen nanofiber fragments surface-bonded to the hyaluronic acid hydrogel network via covalent conjugation during the cross-linking process. The collagen nanofiber fragments, acting as the structural reinforcement component, increased the overall storage modulus of the NHC to a level comparable to native soft tissues while maintaining a sufficiently high degree of porosity of the hydrogel phase to allow host cell infiltration following subcutaneous injection of the NHC microparticles. More importantly, the NHC promoted macrophage/monocyte infiltration, migration, and spreading, sustained cell recruitment over time, and enhanced the proangiogenic effect and recruitment of PDGFRα+ perivascular progenitor cells, leading to extensive adipose tissue remodeling. This study demonstrates the regenerative potential of the injectable NHC microgels as an off-the-shelf solution for devastating soft tissue losses.
Collapse
|
34
|
Wang G, Zhang C, Huang Z, Chen J, Chen H, Lin T, Zhou Z, Gu N, Huang P. Transcytosable and Ultrasound-Activated Liposome Enables Deep Penetration of Biofilm for Surgical Site Infection Management. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2411092. [PMID: 39463041 DOI: 10.1002/adma.202411092] [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: 07/29/2024] [Revised: 09/22/2024] [Indexed: 10/29/2024]
Abstract
Biofilm-associated surgical site infection (BSSI) is a common and grievous postoperative complication lacking effective remedies, mainly due to the poor drug accumulation and penetration in the biofilms featured by dense extracellular polymeric substances (EPSs). Here, it is found that the vascular cell adhesion molecule-1 (VCAM1) is highly overexpressed in the vascular cells of BSSI. It is proposed that the combination of VCAM1-mediated transcytosis and ultrasonic cavitation can consecutively overcome the biological barriers of vascular endothelial cells and EPS for biofilm eradication. To demonstrate the feasibility, a VCAM1-targeted and ultrasound (US)-activated liposome (LPCOTML) loaded with a reactive-oxygen-species (ROS)-responsive lipoid prodrug of oleoyl meropenem, sonosensitizer of lipoid Ce6, and perfluoropentane is developed. LPCOTML can recognize the receptors on vascular cells, and initiate receptor-mediated transcytosis for transendothelial transport into the BSSI periphery. LPCOTML subsequently transforms from nanoparticle into microbubble via liquid-gas phase transition under US irradiation, triggering strong ultrasonic cavitation to blow up the EPS and deeply penetrate the biofilms. The sonosensitizer Ce6 induces ROS production under US irradiation and triggers the release of meropenem to induce potent antibacterial effect in a BSSI model. This study presents an effective strategy to tackle the biological barriers in BSSI via combining receptor-mediated transcytosis and ultrasonic cavitation.
Collapse
Affiliation(s)
- Guowei Wang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Chengyue Zhang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zixuan Huang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310030, China
| | - Jifan Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Hongjian Chen
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| | - Tao Lin
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhuxian Zhou
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Zhejiang Key Laboratory of Smart Biomaterials and Key Laboratory of Biomass Chemical Engineering of Ministry of Education, College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Ning Gu
- Medical School of Nanjing University, Nanjing University, Nanjing, 210093, China
| | - Pintong Huang
- Department of Ultrasound in Medicine, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310009, China
| |
Collapse
|
35
|
Zhu Y, Guo S, Ravichandran D, Ramanathan A, Sobczak MT, Sacco AF, Patil D, Thummalapalli SV, Pulido TV, Lancaster JN, Yi J, Cornella JL, Lott DG, Chen X, Mei X, Zhang YS, Wang L, Wang X, Zhao Y, Hassan MK, Chambers LB, Theobald TG, Yang S, Liang L, Song K. 3D-Printed Polymeric Biomaterials for Health Applications. Adv Healthc Mater 2025; 14:e2402571. [PMID: 39498750 PMCID: PMC11694096 DOI: 10.1002/adhm.202402571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/17/2024] [Indexed: 11/07/2024]
Abstract
3D printing, also known as additive manufacturing, holds immense potential for rapid prototyping and customized production of functional health-related devices. With advancements in polymer chemistry and biomedical engineering, polymeric biomaterials have become integral to 3D-printed biomedical applications. However, there still exists a bottleneck in the compatibility of polymeric biomaterials with different 3D printing methods, as well as intrinsic challenges such as limited printing resolution and rates. Therefore, this review aims to introduce the current state-of-the-art in 3D-printed functional polymeric health-related devices. It begins with an overview of the landscape of 3D printing techniques, followed by an examination of commonly used polymeric biomaterials. Subsequently, examples of 3D-printed biomedical devices are provided and classified into categories such as biosensors, bioactuators, soft robotics, energy storage systems, self-powered devices, and data science in bioplotting. The emphasis is on exploring the current capabilities of 3D printing in manufacturing polymeric biomaterials into desired geometries that facilitate device functionality and studying the reasons for material choice. Finally, an outlook with challenges and possible improvements in the near future is presented, projecting the contribution of general 3D printing and polymeric biomaterials in the field of healthcare.
Collapse
Affiliation(s)
- Yuxiang Zhu
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Shenghan Guo
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Dharneedar Ravichandran
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Arunachalam Ramanathan
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - M. Taylor Sobczak
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Alaina F. Sacco
- School of Chemical, Materials and Biomedical Engineering (CMBE), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Dhanush Patil
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Sri Vaishnavi Thummalapalli
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Tiffany V. Pulido
- Department of ImmunologyMayo Clinic Arizona13400 E Shea BlvdScottsdaleAZ85259USA
| | - Jessica N. Lancaster
- Department of ImmunologyMayo Clinic Arizona13400 E Shea BlvdScottsdaleAZ85259USA
| | - Johnny Yi
- Department of Medical and Surgical GynecologyMayo Clinic Arizona5777 E Mayo BlvdPhoenixAZ85054USA
| | - Jeffrey L. Cornella
- Department of Medical and Surgical GynecologyMayo Clinic Arizona5777 E Mayo BlvdPhoenixAZ85054USA
| | - David G. Lott
- Division of Laryngology, Department of OtolaryngologyMayo Clinic ArizonaPhoenixAZUSA
| | - Xiangfan Chen
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
| | - Xuan Mei
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolCambridgeMA02139USA
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women's HospitalHarvard Medical SchoolCambridgeMA02139USA
| | - Linbing Wang
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Xianqiao Wang
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Yiping Zhao
- Physics, Franklin College of Arts and SciencesUniversity of GeorgiaAthensGA30602USA
| | | | - Lindsay B. Chambers
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Taylor G. Theobald
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| | - Sui Yang
- Materials Science and Engineering, School for Engineering of MatterTransport and Energy (SEMTE) at Arizona State UniversityTempeAZ85287USA
| | | | - Kenan Song
- Manufacturing Engineering, The School of Manufacturing Systems and Networks (MSN), Ira A. Fulton Schools of EngineeringArizona State University (ASU)MesaAZ85212USA
- School of Environmental, Civil, Agricultural, and Mechanical Engineering (ECAM), College of EngineeringUniversity of GeorgiaAthensGA30602USA
| |
Collapse
|
36
|
Yu M, Wang S, Lin D. Mechanism and Application of Biomaterials Targeting Reactive Oxygen Species and Macrophages in Inflammation. Int J Mol Sci 2024; 26:245. [PMID: 39796102 PMCID: PMC11720555 DOI: 10.3390/ijms26010245] [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: 12/03/2024] [Revised: 12/22/2024] [Accepted: 12/26/2024] [Indexed: 01/13/2025] Open
Abstract
Inflammation, an adaptive reaction to harmful stimuli, is a necessary immune system response and can be either acute or chronic. Since acute inflammation tends to eliminate harmful stimuli and restore equilibrium, it is generally advantageous to the organism. Chronic inflammation, however, is caused by either increased inflammatory signaling or decreased pro-anti-inflammatory signaling. According to current studies, inflammation is thought to be a major factor in a number of chronic diseases, including diabetes, cancer, arthritis, inflammatory bowel disease, and obesity. Consequently, reducing inflammation is essential for both preventing and delaying diseases. The application of biomaterials in the treatment of inflammatory illnesses has grown in recent years. A variety of biomaterials can be implanted either by themselves or in conjunction with other bioactive ingredients and therapeutic agents. The mechanisms of action and therapeutic applications of well-known anti-inflammatory biomaterials are the main topics of this article.
Collapse
|
37
|
Wu H, Liu Y, Wang Y, Piao Y, Meng Z, Hu X, Shi L, Shen J, Li Y. Dynamic Covalent Prodrug Nanonetworks via Reaction-Induced Self-Assembly for Periodontitis Treatment. ACS NANO 2024; 18:34884-34901. [PMID: 39663546 DOI: 10.1021/acsnano.4c12580] [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: 12/13/2024]
Abstract
Periodontitis is characterized by dysbiotic biofilms, gingival inflammation, and bone resorption, highlighting the urgent need for a comprehensive approach to drug combination therapy. In this study, we introduce dynamic covalent nanonetworks (dcNNWs) synthesized through a one-pot, four-component reaction-induced self-assembly method using polyamines, 2-formylphenylboronic acid, epigallocatechin gallate, and alendronate. The formation of iminoboronate bonds drives the creation of dcNNWs, allowing controlled release in the periodontitis microenvironment. The inclusion of catechol and bisphosphonate imparts exceptional bioadhesive properties to the dcNNWs, enhancing their efficacy in preventing pathogenic bacterial biofilm formation and eliminating mature biofilms. Moreover, the dcNNWs efficiently absorb pathogen-associated molecular patterns and scavenge excess reactive oxygen species, regulating the local immune response and demonstrating anti-inflammatory effects. Additionally, the released polyphenol and alendronate from the dcNNWs alleviated inflammation and enhanced osteogenesis significantly. The detailed synergistic effects of dcNNWs in biofilm eradication, anti-inflammation, and bone remodeling, with minimal impact on healthy tissues, are confirmed in a rat model of periodontitis. With a facile synthesis process, excellent synergistic effects in periodontitis treatment, and biocompatibility, our dcNNWs present a promising and translational solution for the effective management of periodontitis.
Collapse
Affiliation(s)
- Haoyue Wu
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Yong Liu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yumeng Wang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University institution, Wenzhou, Zhejiang 325035, China
| | - Yinzi Piao
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Zhuojun Meng
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Xiaowen Hu
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, China
| | - Linqi Shi
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Jing Shen
- Department of International VIP Dental Clinic, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, China
| | - Yuanfeng Li
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, Translational Medicine Laboratory, The First Affiliated Hospital of Wenzhou Medical University institution, Wenzhou, Zhejiang 325035, China
| |
Collapse
|
38
|
Liu F, Wang X, Ren M, He P, Li Y, Cui J, Yang S. A shielded cascade of targeted nanocarriers spanning multiple microenvironmental barriers for inflammatory disease therapy. J Nanobiotechnology 2024; 22:789. [PMID: 39710698 DOI: 10.1186/s12951-024-03075-2] [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: 05/27/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
BACKGROUND The multi-biological barriers present in the inflammatory microenvironment severely limit the targeted aggregation of anti-inflammatory drugs in the lesion area. However, conventional responsive drug carriers inevitably come into contact with several pro-responsive stimulatory mediators simultaneously, leading to premature drug release and loss of most therapeutic effects. Breaking through the multi-level barriers of the inflammatory microenvironment is essential to improve the enrichment and bioavailability of drugs. RESULTS In this study, we propose a novel two-stage structural strategy to build shielded cascades of targeted nanocarriers (FA-PTP@Que) through inflammatory mediators, using cascade structures to cross multiple environmental barriers. The cascade structure of FA-PTP@Que is responsive to inflammatory mediators and exhibits ideal pathological microenvironmental response and drug release properties. FA-PTP@Que has shown good macrophage regulation and anti-inflammatory effects by efficiently targeting macrophages, scavenging intracellular reactive oxygen species (ROS), and down-regulating the secretion of pro-inflammatory factors. Significantly, in mice with arthritis and colitis, FA-PTP@Que enriches and targets macrophages at the sites of arthritis and colitis, showing significant anti-inflammatory effects. CONCLUSION FA-PTP@Que combines active chemotaxis of nanocarriers to inflammatory tissues and active targeting of effector cells, acting precisely at each barrier level in different microenvironments by responding to inflammatory mediators and overcoming the multiple barriers in the inflammatory microenvironment. This innovative strategy can effectively break through various inflammatory microenvironments and has the potential application to other inflammatory diseases.
Collapse
Affiliation(s)
- Fengyi Liu
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Xu Wang
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
| | - Mingxing Ren
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
| | - Ping He
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Yuzhou Li
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China
| | - Jing Cui
- Chongqing Medical University, Chongqing, China
| | - Sheng Yang
- College of Stomatology, Chongqing Medical University, 426#Songshibei Road, Yubei District, Chongqing, 401147, China.
- Chongqing Key Laboratory of Oral Diseases, Chongqing, China.
- Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Chongqing, China.
| |
Collapse
|
39
|
Brettner FEB, Gier S, Haessler A, Schreiner J, Vogel-Kindgen S, Windbergs M. Anti-inflammatory effects of cyclodextrin nanoparticles enable macrophage repolarization and reduce inflammation. DISCOVER NANO 2024; 19:211. [PMID: 39707045 DOI: 10.1186/s11671-024-04175-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
Inflammation plays a critical role in the pathophysiology of many diseases, and dysregulation of the involved signaling cascades often culminates in uncontrollable disease progression and, ultimately, chronic manifestation. Addressing these disorders requires balancing inflammation control while preserving essential immune functions. Cyclodextrins (CDs), particularly β-CD, have gained attention as biocompatible biomaterials with intrinsic anti-inflammatory properties, and chemical modification of their backbone offers a promising strategy to enhance their physicochemical properties, adaptability, and therapeutic potential. This study evaluated and characterized the immunomodulatory effects of amphiphilic CD derivatives, which self-assemble into nanoparticles, compared to soluble parent β-CD. In a human macrophage model, CD nanoparticles demonstrated superior anti-inflammatory activity, with derivative-specific effects tied to their physicochemical properties, surpassing the soluble β-CD control. Alongside the downregulation of key pro-inflammatory markers, significant reductions in inflammasome activation and changes in lipid profiles were observed. The findings of this study underscore the potential of cyclodextrin-based nanoparticles as versatile biomaterials for treating the complex pathophysiology of various acute and chronic inflammation-associated disorders.
Collapse
Affiliation(s)
- Felix E B Brettner
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Stefanie Gier
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Annika Haessler
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Jonas Schreiner
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Sarah Vogel-Kindgen
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany
| | - Maike Windbergs
- Institute of Pharmaceutical Technology, Goethe-University Frankfurt, Max-von-Laue-Straße 9, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
40
|
Kim H, Kim KS, Lee YC, Cho JH. Chloroform Extract from Fermented Viola mandshurica Regulates LPS-Induced Inflammation Response in RAW 264.7 Cells by Inhibiting iNOS and COX-2. J Microbiol Biotechnol 2024; 35:e2408047. [PMID: 39849923 PMCID: PMC11813387 DOI: 10.4014/jmb.2408.08047] [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: 08/27/2024] [Revised: 11/12/2024] [Accepted: 11/30/2024] [Indexed: 01/25/2025]
Abstract
Inflammatory is a crucial part of the immune system of body protect it from harmful invaders, such as bacteria, viruses, and other foreign substances. In this study, the effects of chloroform extract of fermented Viola mandshurica (CEFV) on lipopolysaccharide (LPS)-induced inflammatory response in RAW264.7 macrophages were investigated. The CEFV significantly inhibited NO production and reduced the expression of inducible nitric oxide synthase (iNOS) at both protein and mRNA levels in a dose-dependent manner. Also, CEFV decreased PGE2 production, suppressed COX-2 expression, and inhibited the activation of the ERK and JNK pathways but not the p38 pathway. Taken together, CEFV suppressed NF-κB activation, which is a key regulator in the inflammatory response. The main phenolic compounds identified in CEFV were tectoridin, luteolin, resveratrol, and hesperetin. Therefore, in this study, CEFC exhibits potent anti-inflammatory effects by downregulating the production of pro-inflammatory mediators and inhibiting key inflammatory pathway in RAW264.7 cells.
Collapse
Affiliation(s)
- Hyunju Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A
University, Busan 49315, Republic of Korea
| | - Kyoung-Sook Kim
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A
University, Busan 49315, Republic of Korea
| | - Young-Choon Lee
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A
University, Busan 49315, Republic of Korea
| | - Jong Hyun Cho
- Department of Medicinal Biotechnology, College of Health Sciences, Dong-A
University, Busan 49315, Republic of Korea
| |
Collapse
|
41
|
Cheng X, Sui H, Chen F, Li C, Du M, Zhang S, Chen J, Dou J, Huang Y, Xie X, Cheng C, Yang R, Yang C, Shi B, Shao D, Leong KW, Huang H. Nanomaterial-Mediated Reprogramming of Macrophages to Inhibit Refractory Muscle Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410368. [PMID: 39548911 PMCID: PMC11849413 DOI: 10.1002/adma.202410368] [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: 07/17/2024] [Revised: 11/05/2024] [Indexed: 11/18/2024]
Abstract
Orofacial muscles are particularly prone to refractory fibrosis after injury, leading to a negative effect on the patient's quality of life and limited therapeutic options. Gaining insights into innate inflammatory response-fibrogenesis homeostasis can aid in the development of new therapeutic strategies for muscle fibrosis. In this study, the crucial role of macrophages is identified in the regulation of orofacial muscle fibrogenesis after injury. Hypothesizing that orchestrating macrophage polarization and functions will be beneficial for fibrosis treatment, nanomaterials are engineered with polyethylenimine functionalization to regulate the macrophage phenotype by capturing negatively charged cell-free nucleic acids (cfNAs). This cationic nanomaterial reduces macrophage-related inflammation in vitr and demonstrates excellent efficacy in preventing orofacial muscle fibrosis in vivo. Single-cell RNA sequencing reveals that the cationic nanomaterial reduces the proportion of profibrotic Gal3+ macrophages through the cfNA-mediated TLR7/9-NF-κB signaling pathway, resulting in a shift in profibrotic fibro-adipogenic progenitors (FAPs) from the matrix-producing Fabp4+ subcluster to the matrix-degrading Igf1+ subcluster. The study highlights a strategy to target innate inflammatory response-fibrogenesis homeostasis and suggests that cationic nanomaterials can be exploited for treating refractory fibrosis.
Collapse
Affiliation(s)
- Xu Cheng
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Hao Sui
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Fangman Chen
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Chenghao Li
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Meijun Du
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Shiming Zhang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jiali Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Jinfeng Dou
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yixuan Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Xiaochun Xie
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Chuanxu Cheng
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Renjie Yang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Eastern Clinic, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Chao Yang
- Department of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510630, China
| | - Bing Shi
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Dan Shao
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510006, China
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, Guangdong, 510006, China
| | - Kam W Leong
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
- Department of Systems Biology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Hanyao Huang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases and Department of Oral and Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, 610041, China
- Department of Biomedical Engineering, Columbia University, New York, NY, 10027, USA
| |
Collapse
|
42
|
Xu F, Zhao S, Zhu Y, Zhu J, Kong L, Li H, Ma S, Wang B, Qu Y, Tian Z, Zhao J, Liu L. Phytic acid-based nanomedicine against mTOR represses lipogenesis and immune response for metabolic dysfunction-associated steatohepatitis therapy. LIFE METABOLISM 2024; 3:loae026. [PMID: 39873005 PMCID: PMC11748520 DOI: 10.1093/lifemeta/loae026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 06/03/2024] [Accepted: 06/17/2024] [Indexed: 01/30/2025]
Abstract
Metabolic dysfunction-associated steatohepatitis (MASH) is one of the most common chronic liver diseases and is mainly caused by metabolic disorders and systemic inflammatory responses. Recent studies have indicated that the activation of the mammalian (or mechanistic) target of rapamycin (mTOR) signaling participates in MASH progression by facilitating lipogenesis and regulating the immune microenvironment. Although several molecular medicines have been demonstrated to inhibit the phosphorylation or activation of mTOR, their poor specificity and side effects limit their clinical application in MASH treatment. Phytic acid (PA), as an endogenous and natural antioxidant in the liver, presents significant anti-inflammatory and lipid metabolism-inhibiting functions to alleviate MASH. In this study, considering the unique phosphate-rich structure of PA, we developed a cerium-PA (CePA) nanocomplex by combining PA with cerium ions possessing phosphodiesterase activity. CePA intervened in the S2448 phosphorylation of mTOR through the occupation effect of phosphate groups, thereby inhibiting the inflammatory response and mTOR-sterol regulatory element-binding protein 1 (SREBP1) regulation axis. The in vivo experiments suggested that CePA alleviated MASH progression and fat accumulation in high-fat diet-fed mice. Mechanistic studies validated that CePA exerts a liver-targeted mTOR repressive function, making it a promising candidate for MASH and other mTOR-related disease treatments.
Collapse
Affiliation(s)
- Fenghua Xu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, Shaanxi 710032, China
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi’an, Shaanxi 710062, China
| | - Shoujie Zhao
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi 710038, China
| | - Yejing Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, Shaanxi 710032, China
| | - Jun Zhu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, Shaanxi 710032, China
| | - Lingyang Kong
- Key Laboratory of Modern Teaching Technology, Ministry of Education, Shaanxi Normal University, Xi’an, Shaanxi 710062, China
| | - Huichen Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, Shaanxi 710032, China
| | - Shouzheng Ma
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi 710038, China
| | - Bo Wang
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi 710038, China
| | - Yongquan Qu
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, China
| | - Zhimin Tian
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an, Shaanxi 710072, China
| | - Junlong Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Medical Genetics and Developmental Biology, Air Force Medical University, Xi’an, Shaanxi 710032, China
- Department of Pediatrics, Tangdu Hospital, Air Force Medical University, Xi’an, Shaanxi 710000, China
| | - Lei Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and Xijing Hospital of Digestive Diseases, Air Force Medical University, Xi’an, Shaanxi 710032, China
| |
Collapse
|
43
|
Chen T, Jiang H, Zhang R, He F, Han N, Wang Z, Jia J. Leveraging printability and biocompatibility in materials for printing implantable vessel scaffolds. Mater Today Bio 2024; 29:101366. [PMID: 39698000 PMCID: PMC11652949 DOI: 10.1016/j.mtbio.2024.101366] [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: 07/11/2024] [Revised: 11/10/2024] [Accepted: 11/23/2024] [Indexed: 12/20/2024] Open
Abstract
Vessel scaffolds are crucial for treating cardiovascular diseases (CVDs). It is currently feasible to fabricate vessel scaffolds from a variety of materials using traditional fabrication methods, but the risks of thrombus formation, chronic inflammation, and atherosclerosis associated with these scaffolds have led to significant limitations in the clinical usages. Bioprinting, as an emerging technology, has great potential in constructing implantable vessel scaffolds. During the fabrication of the constructs, the biomaterials used for bioprinting have offered significant contributions for the successful fabrications of the vessel scaffolds. Herein, we review recent advances in biomaterials for bioprinting implantable vessel scaffolds. First, we briefly introduce the requirements for implantable vessel scaffolds and its conventional manufacturing methods. Next, a brief overview of the classic methods for bioprinting vessel scaffolds is presented. Subsequently, we provide an in-depth analysis of the properties of the representative natural, synthetic, composite and hybrid biomaterials that can be used for bioprinting implantable vessel scaffolds. Ultimately, we underscore the necessity of leveraging biocompatibility and printability for biomaterials, and explore the unmet needs and potential applications of these biomaterials in the field of bioprinted implantable vessel scaffolds.
Collapse
Affiliation(s)
- Tianhong Chen
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Haihong Jiang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Ruoxuan Zhang
- School of Life Sciences, Shanghai University, Shanghai, China
| | - Fan He
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| | - Ning Han
- Department of Orthopedic Traumatology, Shanghai East Hospital, Tongji University, China
| | - Zhimin Wang
- Shanghai-MOST Key Laboratory of Health and Disease Genomics, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, 200237, China
| | - Jia Jia
- School of Life Sciences, Shanghai University, Shanghai, China
- Sino-Swiss Institute of Advanced Technology, School of Micro-electronics, Shanghai University, Shanghai, China
| |
Collapse
|
44
|
Nowotnick AG, Xi Z, Jin Z, Khalatbarizamanpoor S, Brauer DS, Löffler B, Jandt KD. Antimicrobial Biomaterials Based on Physical and Physicochemical Action. Adv Healthc Mater 2024; 13:e2402001. [PMID: 39301968 DOI: 10.1002/adhm.202402001] [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: 05/31/2024] [Revised: 08/09/2024] [Indexed: 09/22/2024]
Abstract
Developing effective antimicrobial biomaterials is a relevant and fast-growing field in advanced healthcare materials. Several well-known (e.g., traditional antibiotics, silver, copper etc.) and newer (e.g., nanostructured, chemical, biomimetic etc.) approaches have been researched and developed in recent years and valuable knowledge has been gained. However, biomaterials associated infections (BAIs) remain a largely unsolved problem and breakthroughs in this area are sparse. Hence, novel high risk and potential high gain approaches are needed to address the important challenge of BAIs. Antibiotic free antimicrobial biomaterials that are largely based on physical action are promising, since they reduce the risk of antibiotic resistance and tolerance. Here, selected examples are reviewed such antimicrobial biomaterials, namely switchable, protein-based, carbon-based and bioactive glass, considering microbiological aspects of BAIs. The review shows that antimicrobial biomaterials mainly based on physical action are powerful tools to control microbial growth at biomaterials interfaces. These biomaterials have major clinical and application potential for future antimicrobial healthcare materials without promoting microbial tolerance. It also shows that the antimicrobial action of these materials is based on different complex processes and mechanisms, often on the nanoscale. The review concludes with an outlook and highlights current important research questions in antimicrobial biomaterials.
Collapse
Affiliation(s)
- Adrian G Nowotnick
- Chair of Materials Science (CMS), Otto Schott Institute of Materials Research (OSIM), Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
- Jena School for Microbial Communication (JSMC), 07743, Neugasse 23, Jena, Germany
| | - Zhongqian Xi
- Chair of Materials Science (CMS), Otto Schott Institute of Materials Research (OSIM), Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
- Jena School for Microbial Communication (JSMC), 07743, Neugasse 23, Jena, Germany
| | - Zhaorui Jin
- Bioactive Glasses Group, Otto Schott Institute of Materials Research (OSIM), Friedrich Schiller University Jena, Lessingstraße 12, 07743, Jena, Germany
| | - Sadaf Khalatbarizamanpoor
- Jena School for Microbial Communication (JSMC), 07743, Neugasse 23, Jena, Germany
- Institute of Medical Microbiology, Jena University Hospital, 07747, Am Klinikum 1, Jena, Germany
| | - Delia S Brauer
- Bioactive Glasses Group, Otto Schott Institute of Materials Research (OSIM), Friedrich Schiller University Jena, Lessingstraße 12, 07743, Jena, Germany
| | - Bettina Löffler
- Jena School for Microbial Communication (JSMC), 07743, Neugasse 23, Jena, Germany
- Institute of Medical Microbiology, Jena University Hospital, 07747, Am Klinikum 1, Jena, Germany
| | - Klaus D Jandt
- Chair of Materials Science (CMS), Otto Schott Institute of Materials Research (OSIM), Friedrich Schiller University Jena, Löbdergraben 32, 07743, Jena, Germany
- Jena School for Microbial Communication (JSMC), 07743, Neugasse 23, Jena, Germany
| |
Collapse
|
45
|
Samulevich ML, Carman LE, Aneskievich BJ. Critical Analysis of Cytoplasmic Progression of Inflammatory Signaling Suggests Potential Pharmacologic Targets for Wound Healing and Fibrotic Disorders. Biomedicines 2024; 12:2723. [PMID: 39767629 PMCID: PMC11726985 DOI: 10.3390/biomedicines12122723] [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: 10/30/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025] Open
Abstract
Successful skin wound healing is dependent on an interplay between epidermal keratinocytes and dermal fibroblasts as they react to local extracellular factors (DAMPs, PAMPs, cytokines, etc.) surveyed from that environment by numerous membrane receptors (e.g., TLRs, cytokine receptors, etc.). In turn, those receptors are the start of a cytoplasmic signaling pathway where balance is key to effective healing and, as needed, cell and matrix regeneration. When directed through NF-κB, these signaling routes lead to transient responses to the benefit of initiating immune cell recruitment, cell replication, local chemokine and cytokine production, and matrix protein synthesis. The converse can also occur, where ongoing canonical NF-κB activation leads to chronic, hyper-responsive states. Here, we assess three key players, TAK1, TNFAIP3, and TNIP1, in cytoplasmic regulation of NF-κB activation, which, because of their distinctive and yet inter-related functions, either promote or limit that activation. Their balanced function is integral to successful wound healing, given their significant control over the expression of inflammation-, fibrosis-, and matrix remodeling-associated genes. Intriguingly, these three proteins have also been emphasized in dysregulated NF-κB signaling central to systemic sclerosis (SSc). Notably, diffuse SSc shares some tissue features similar to an excessive inflammatory/fibrotic wound response without eventual resolution. Taking a cue from certain instances of aberrant wound healing and SSc having some shared aspects, e.g., chronic inflammation and fibrosis, this review looks for the first time, to our knowledge, at what those pathologies might have in common regarding the cytoplasmic progression of NF-κB-mediated signaling. Additionally, while TAK1, TNFAIP3, and TNIP1 are often investigated and reported on individually, we propose them here as three proteins whose consequences of function are very highly interconnected at the signaling focus of NF-κB. We thus highlight the emerging promise for the eventual clinical benefit derived from an improved understanding of these integral signal progression modulators. Depending on the protein, its indirect or direct pharmacological regulation has been reported. Current findings support further intensive studies of these points in NF-κB regulation both for their basic function in healthy cells as well as with the goal of targeting them for translational benefit in multiple cutaneous wound healing situations, whether stemming from acute injury or a dysregulated inflammatory/fibrotic response.
Collapse
Affiliation(s)
- Michael L. Samulevich
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (M.L.S.); (L.E.C.)
| | - Liam E. Carman
- Graduate Program in Pharmacology & Toxicology, University of Connecticut, Storrs, CT 06269-3092, USA; (M.L.S.); (L.E.C.)
| | - Brian J. Aneskievich
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Connecticut, Storrs, CT 06269-3092, USA
| |
Collapse
|
46
|
Li Y, Chen W, Koo S, Liu H, Saiding Q, Xie A, Kong N, Cao Y, Abdi R, Serhan CN, Tao W. Innate immunity-modulating nanobiomaterials for controlling inflammation resolution. MATTER 2024; 7:3811-3844. [PMID: 40123651 PMCID: PMC11925551 DOI: 10.1016/j.matt.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The acute inflammatory response is an inherent protective mechanism, its unsuccessful resolution can contribute to disease pathogenesis and potentially lead to death. Innate immune cells are the first line of host defenders and play a substantial role in inflammation initiation, amplification, resolution, or subsequent disease progression. As the resolution of inflammation is an active and highly regulated process, modulating innate immune cells, including neutrophils, monocytes and macrophages, and endothelial cells, and their interactions offer opportunities to control excessive inflammation. Nanobiomaterials have shown superior therapeutic potential in inflammation-related diseases by manipulating inflammatory responses because nanobiomaterials can target and interact with innate immune cells. Versatile nanobiomaterials can be designed for targeted modulation of specific innate immune responses. Nanopro-resolving medicines have been prepared both with pro-resolving lipid mediators and peptides each demonstrated to active resolution of inflammation in animal disease models. Here, we review innovative nanobiomaterials for modulating innate immunity and alleviating inflammation. We summarise the strategies converging the design of nanobiomaterials and the nano-bio interaction in modulating innate immune profiles and propelling the advancement of nanobiomaterials for inflammatory disease treatments. We also propose the future perspectives and translational challenges of nanobiomaterials that need to be overcome in this swiftly rising field.
Collapse
Affiliation(s)
- Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haijun Liu
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angel Xie
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
47
|
Liu X, Chen S, Huang J, Du Y, Luo Z, Zhang Y, Liu L, Chen Y. Synthetic polypeptides inhibit nucleic acid-induced inflammation in autoimmune diseases by disrupting multivalent TLR9 binding to LL37-DNA bundles. NATURE NANOTECHNOLOGY 2024; 19:1745-1756. [PMID: 39160338 DOI: 10.1038/s41565-024-01759-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Accepted: 07/15/2024] [Indexed: 08/21/2024]
Abstract
Complexes of extracellular nucleic acids (NAs) with endogenous proteins or peptides, such as LL37, break immune balance and cause autoimmune diseases, whereas NAs with arginine-enriched peptides do not. Inspired by this, we synthesize a polyarginine nanoparticle PEG-TK-NPArg, which effectively inhibits Toll-like receptor-9 (TLR9) activation, in contrast to LL37. To explore the discrepancy effect of PEG-TK-NPArg and LL37, we evaluate the periodic structure of PEG-TK-NPArg-NA and LL37-NA complexes using small-angle X-ray scattering. LL37-NA complexes have a larger inter-NA spacing that accommodates TLR9, while the inter-NA spacing in PEG-TK-NPArg-NA complexes mismatches with the cavity of TLR9, thus inhibiting an interaction with multiple TLR9s, limiting their clustering and damping immune induction. Subsequently, the inhibitory inflammation effect of PEG-TK-NPArg is proved in an animal model of rheumatoid arthritis. This work on how the scavenger-NA complexes inhibit the immune response may facilitate proof-of-concept research translating to clinical application.
Collapse
Affiliation(s)
- Xingliang Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
- School of Engineering, Westlake University, Hangzhou, China
| | - Shi Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Jing Huang
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
- School of Engineering, Westlake University, Hangzhou, China
| | - Yibo Du
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China
| | - Zhi Luo
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, China
| | - Yue Zhang
- School of Engineering, Westlake University, Hangzhou, China.
| | - Lixin Liu
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China.
| | - Yongming Chen
- School of Materials Science and Engineering, Key Laboratory for Polymeric Composite and Functional Materials of Ministry of Education, Sun Yat-sen University, Guangzhou, China.
- Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- College of Chemistry and Molecular Sciences, Henan University, Kaifeng, China.
- State Key Laboratory of Antiviral Drugs, Henan University, Zhengzhou, China.
| |
Collapse
|
48
|
Wang K, Wang X, Song L. Unraveling the complex role of neutrophils in lymphoma: From pathogenesis to therapeutic approaches (Review). Mol Clin Oncol 2024; 21:85. [PMID: 39347476 PMCID: PMC11428085 DOI: 10.3892/mco.2024.2783] [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: 05/22/2024] [Accepted: 08/21/2024] [Indexed: 10/01/2024] Open
Abstract
Lymphoma, a malignancy of the lymphatic system, which is critical for maintaining the body's immune defenses, has become a focal point in recent research due to its intricate interplay with neutrophils-white blood cells essential for combating infections and inflammation. Unlike prior perceptions associating neutrophils only with tumor support, contemporary studies underscore their intricate and multifaceted involvement in the immune response to lymphoma. Recognizing the nuanced participation of neutrophils in lymphoma is crucial for developing innovative treatments to improve patient outcomes.
Collapse
Affiliation(s)
- Ke Wang
- Department of Cell Engineering, School of Life Sciences and Biotechnology, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Xiao Wang
- Reproduction Medicine Center, Affiliated Hospital of Guangdong Medical University, Guangdong Medical University, Zhanjiang, Guangdong 524002, P.R. China
| | - Li Song
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Sanquan College of Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| |
Collapse
|
49
|
Lee JH, Shin SJ, Lee JH, Knowles JC, Lee HH, Kim HW. Adaptive immunity of materials: Implications for tissue healing and regeneration. Bioact Mater 2024; 41:499-522. [PMID: 39206299 PMCID: PMC11350271 DOI: 10.1016/j.bioactmat.2024.07.027] [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: 05/07/2024] [Revised: 07/16/2024] [Accepted: 07/21/2024] [Indexed: 09/04/2024] Open
Abstract
Recent cumulative findings signify the adaptive immunity of materials as a key agenda in tissue healing that can improve regenerative events and outcomes. Modulating immune responses, mainly the recruitment and functions of T and B cells and their further interplay with innate immune cells (e.g., dendritic cells, macrophages) can be orchestrated by materials. For instance, decellularized matrices have been shown to promote muscle healing by inducing T helper 2 (Th2) cell immunity, while synthetic biopolymers exhibit differential effects on B cell responses and fibrosis compared decellularized matrices. We discuss the recent findings on how implantable materials instruct the adaptive immune events and the subsequent tissue healing process. In particular, we dissect the materials' physicochemical properties (shape, size, topology, degradation, rigidity, and matrix dynamic mechanics) to demonstrate the relations of these parameters with the adaptive immune responses in vitro and the underlying biological mechanisms. Furthermore, we present evidence of recent in vivo phenomena, including tissue healing, cancer progression, and fibrosis, wherein biomaterials potentially shape adaptive immune cell functions and in vivo outcomes. Our discussion will help understand the materials-regulated immunology events more deeply, and offer the design rationale of materials with tunable matrix properties for accelerated tissue repair and regeneration.
Collapse
Affiliation(s)
- Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Seong-Jin Shin
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
| | - Jonathan C. Knowles
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman Dental Institute, University College London, London NW3 2PX, United Kingdom
| | - Hae-Hyoung Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Republic of Korea
- Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan 31116, Republic of Korea
- Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
- Cell & Matter Institute, Dankook University, Cheonan 31116, Republic of Korea
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan 31116, Republic of Korea
- UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan 31116, Republic of Korea
| |
Collapse
|
50
|
Wang L, Jiang Y, Yao Y, Deng Y, Liu Z, Ding J, Wang W, Chen H, Nan K, Li L. Injectable drug-loaded thermosensitive hydrogel delivery system for protecting retina ganglion cells in traumatic optic neuropathy. Regen Biomater 2024; 11:rbae124. [PMID: 39569076 PMCID: PMC11578600 DOI: 10.1093/rb/rbae124] [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: 05/03/2024] [Revised: 09/17/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Currently, generalized therapy for traumatic optic neuropathy (TON) is lacking. Various strategies have been developed to protect and regenerate retinal ganglion cells (RGCs) after TON. Intravitreal injection of supplements has been approved as a promising approach, although serious concerns, such as low delivery efficacy and pain due to frequent injections, remain. In this study, we tested an injectable thermosensitive hydrogel drug delivery system engineered to deliver ciliary neurotrophic factor (CNTF) and triamcinolone acetonide (TA). The results of rheological studies showed that the prepared drug-loaded hydrogel possessed a suitable mechanical modulus of ∼300 Pa, consistent with that of vitreum. The hydrogel exhibited thermosensitive with sustained drug release performance. In vitro co-culture of the CNTF-loaded hydrogel system with primary RGCs also induced significant axon regeneration, with 38.5% increase in neurite length, indicating the regenerative response of the thermosensitive hydrogel drug delivery system. A Sprague-Dawley rat optic nerve crush model was constructed and applied to determine the neuroprotective and regenerative capacities of the system. The results demonstrated that a single intravitreal injection of the drug-loaded hydrogel (PLGA-PEG-PLGA + TA or PLGA-PEG-PLGA + CNTF) significantly increased RGC survival at both 14 and 28 days. The RGC survival rate was 31.05 ± 1.41% for the drug-loaded hydrogel system (the control group was 16.79 ± 1.50%) at Day 28. These findings suggest that the injectable drug-loaded thermosensitive hydrogel delivery system is a promising therapeutic tool for treating optic nerve degeneration.
Collapse
Affiliation(s)
- Lei Wang
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- The Affiliated Xiangshan Hospital, Wenzhou Medical University, Ningbo, Zhejiang 315700, China
| | - Yan Jiang
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Refractive Surgery Center, Chongqing Eye and Vision Care Hospital, Chongqing 40042, China
| | - Yili Yao
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Yudan Deng
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Zhiqiang Liu
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jiangtao Ding
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Wenwen Wang
- Engineering Research Center of Clinical Functional Materials and Diagnosis & Treatment Devices of Zhejiang Province, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| | - Hao Chen
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Kaihui Nan
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Lingli Li
- School of Ophthalmology & Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325000, China
| |
Collapse
|