1
|
Zhang K, Zhu J, Sun W, Zhang Y, Li W, Wang Y, Zhou C, He Y, Qin J. Antibacterial betaine modified chitosan-based hydrogel with angiogenic property for photothermal enhanced diabetic wound repairing. Carbohydr Polym 2025; 349:123033. [PMID: 39638511 DOI: 10.1016/j.carbpol.2024.123033] [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: 09/08/2024] [Revised: 11/03/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024]
Abstract
Chronic diabetes wound repairing is still a huge challenge in clinical practice. High concentration of reactive oxygen species and vascular disfunction are the main factors hindering the recovery of diabetes wounds. This research grafted betaine onto chitosan (CSBT) to enhance the antibacterial property and the CSBT was cross-linked with PEO90 dialdehyde (PEO DA) to prepare hydrogel with Ca2+ loading to promote the coagulation. The polydopamine nanoparticles (PDA NPs) with photothermal property and antioxidant property was composited to the hydrogel and deferoxamine (DFO) was loaded to fabricate the multifunctional CBPCa/PDA/DFO hydrogel to promote vascular regeneration in combination with photothermal antibacterial performance for the diabetes wounds treatment. The CBPCa/PDA/DFO hydrogel showed good mechanical strength, injectability, anti-inflammatory property and coagulation performance. Furthermore, the antibacterial effect of chitosan based hydrogel was enhanced with near infrared (NIR) stimulated photothermal treatment. Combined with the photothermal effect and the angiogenic drug DFO release, the CBPCa/PDA/DFO hydrogel significantly enhanced vascular regeneration and reduced the inflammation in the in vivo wound repairing experiment. As a result, the CBPCa/PDA/DFO hydrogel may provide a promising therapeutic platform for diabetic trauma repairing.
Collapse
Affiliation(s)
- Kaiyue Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Jingjing Zhu
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Weichen Sun
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Yu Zhang
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China
| | - Wenjuan Li
- Key Laboratory of Pathogenesis mechanism and control of inflammatory-autoimmune diseases in Hebei Province, Hebei University, Baoding, Hebei Province, China.
| | - Yong Wang
- Key Laboratory of Pathogenesis mechanism and control of inflammatory-autoimmune diseases in Hebei Province, Hebei University, Baoding, Hebei Province, China.
| | - Chengyan Zhou
- College of Pharmaceutical Sciences, Hebei University, Baoding, Hebei Province 071002, China
| | - Yingna He
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang City, Hebei Province 050200, China
| | - Jianglei Qin
- College of Chemistry and Materials Science, Hebei University, Baoding City, Hebei Province 071002, China; Key Laboratory of Pathogenesis mechanism and control of inflammatory-autoimmune diseases in Hebei Province, Hebei University, Baoding, Hebei Province, China.
| |
Collapse
|
2
|
Che Z, Sheng X, Sun Q, Wu Y, Song K, Chen A, Chen J, Chen Q, Cai M. Deferoxamine functionalized alginate-based collagen composite material enhances the integration of metal implant and bone interface. Carbohydr Polym 2025; 349:122944. [PMID: 39643405 DOI: 10.1016/j.carbpol.2024.122944] [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: 08/12/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 12/09/2024]
Abstract
Poor osseointegration markedly compromises the longevity of prostheses. To enhance the stability of titanium implants, surface functionalization is a proven strategy to promote prosthesis-bone integration. This study developed a hydrogel coating capable of simultaneous osteoangiogenesis and vascularization by incorporating deferoxamine (DFO) into a sodium alginate mineralized collagen composite hydrogel. The physicochemical properties of this hydrogel were thoroughly analyzed. In vivo and in vitro experiments confirmed the hydrogel scaffold's osteogenic and angiogenic capabilities. Results indicated that sodium alginate notably enhanced the mechanical characteristics of the mineralized collagen, allowing it to fully infiltrate the interstices of the 3D-printed titanium scaffold. Furthermore, as the hydrogel degraded, collagen, calcium ion, phosphate ion, and DFO were gradually released around the scaffolds, altering the local osteogenic microenvironment and strongly inducing new bone tissue growth. These findings offer novel perspectives for the creation and utilization of functionalized bone implant materials.
Collapse
Affiliation(s)
- Zhenjia Che
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| | - Xiao Sheng
- Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, Wuxing, Huzhou, Zhejiang 313000, People's Republic of China
| | - Qi Sun
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Yanglin Wu
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Kaihang Song
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Aopan Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Jing Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Qiyun Chen
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Middle Yanchang Road, Shanghai 200072, People's Republic of China.
| |
Collapse
|
3
|
Hu J, Yang B, Tao Z, Chen J, Zhang X, Wang S, Xing G, Ngeng NA, Malik A, Appiah-Kubi K, Farina M, Skalny AV, Tinkov AA, Aschner M, Lu R. The role of HIF-1α/BNIP3/mitophagy in acrylonitrile-induced neuronal death in HT22 cells and mice: A potential neuroprotection target. Chem Biol Interact 2025; 406:111327. [PMID: 39615733 DOI: 10.1016/j.cbi.2024.111327] [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: 03/08/2024] [Revised: 05/07/2024] [Accepted: 11/28/2024] [Indexed: 12/13/2024]
Abstract
Acrylonitrile (AN) is a widely utilized organic compound in the production of diverse industrial synthetic materials. While acute exposure to AN can cause neurotoxicity, the precise mechanism remains unclear. Hypoxia-inducible factor 1 alpha (HIF-1α) is a pivotal transcription factor that coordinates and orchestrates multiple physiological processes to adapt to hypoxic conditions, ensuring cellular survival and homeostasis. In this study, we used in vitro (cultured mouse hippocampal neuronal cell line, HT22) and in vivo (AN exposed mice) approaches to investigate the potential modulator effects of HIF-1α in AN-induced neurotoxicity. In vitro, AN exposure caused concentration-dependent toxicity in HT22 cells, which was paralleled by increased Bax levels while decreasing Bcl-2. Exposure to AN resulted in reduced protein levels of HIF-1α, Bcl-2 19-kDa interacting protein 3 (BNIP3), microtubule-associated protein 1 light chain 3 beta (LC3B) and Beclin1, while increased the protein levels of the translocase of outer mitochondrial membrane 20 (TOM20). Furthermore, mitochondrial morphology and function were compromised, suggesting that AN impaired HIF-1α/BNIP3-mediated mitochondrial autophagy and promoted apoptosis. Treatment with a HIF-1α activator, cobalt chloride (CoCl2), reversed these effects, while pretreatment with a HIF-1α inhibitor, 2-methoxyestradiol (2-MeOE2), augmented them. In BNIP3 overexpressing HT22 cells, enhanced cell viability and reduced apoptosis rates were observed. Furthermore, the HIF-1α/BNIP3 pathway was activated by the prolyl hydroxylase (PHD2) inhibitor, deferoxamine (DFO), which increased HT22 cell viability. Similarly, the activation of HIF-1α by administering 20 mg/kg of CoCl2 was found to alleviate neurotoxicity in mice. This treatment enhanced elevations of autophagy protein expression and co-localization of BNIP3 and LC3B. In summary, under normoxia, AN induced neurotoxicity by promoting PHD2-mediated HIF-1α degradation, disrupted the BNIP3-mediated mitophagy pathway, and enhanced apoptosis. Our findings underscore the effect of the HIF-1α/BNIP3-mediated mitochondrial autophagy in AN-induced neurotoxicity and suggest potential therapeutic strategies involving HIF-1α activation or BNIP3 overexpression for AN poisoning treatment.
Collapse
Affiliation(s)
- Jing Hu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Zehua Tao
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Jian Chen
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Xinyu Zhang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Ngwa Adeline Ngeng
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Abdul Malik
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China; Department of General Surgery, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu Province, 212001, China
| | - Kwaku Appiah-Kubi
- Department of Applied Biology, C. K. Tedam University of Technology and Applied Sciences, Navrongo, UK-0215-5321, Ghana
| | - Marcelo Farina
- Department of Biochemistry, Federal University of Santa Catarina, Florianópolis, Santa Catarina, 88040-900, Brazil
| | - Anatoly V Skalny
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia; Orenburg State University, Pobedy Ave.13, Orenburg, 460018, Russia
| | - Alexey A Tinkov
- Laboratory of Molecular Dietetics, IM Sechenov First Moscow State Medical University (Sechenov University), Bolshaya Pirogovskaya St., 2-4, Moscow, 119146, Russia; Orenburg State University, Pobedy Ave.13, Orenburg, 460018, Russia; Laboratory of Ecobiomonitoring and Quality Control, Yaroslavl State University, Yaroslavl, 150000, Russia
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Sciences, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China; Experimental Research Center, Affiliated Kunshan Hospital, Jiangsu University, Kunshan, Jiangsu, 215300, China.
| |
Collapse
|
4
|
Pan X, Zong Q, Fu B, Wang Y, Feng X, Sun W, Zhai Y. The preparation of methacrylated oxidized konjac glucomannan hydrogel system and its treatment for diabetic wounds. Int J Biol Macromol 2025; 298:140005. [PMID: 39828163 DOI: 10.1016/j.ijbiomac.2025.140005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/24/2024] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
The management of diabetic wounds has become an important task for the public health system. Hydrogels are highly anticipated as modern wound dressings for the treatment of diabetic wounds, hence we have prepared a MOK-Gel using methacrylated oxidized konjac glucomannan (MOK) crosslinked with acrylamide (AM). On this basis, we have incorporated drugs such as UiO-66 loaded with sodium ferulate (SF) and deferoxamine (DFO) to develop the hydrogel wound dressing DUS@MOK-Gel (a hydrogel composed of methacrylated oxidized konjac glucomannan, loaded with DFO and UiO-66 loaded with sodium ferulate). It not only has excellent physical properties, including swelling capacity, moisture retention, and water vapor permeability; but also possesses bioactivity functions such as antioxidant, anti-inflammatory, macrophage polarization regulation, promotion of anti-inflammatory factor release, and angiogenesis to accelerate the healing of diabetic wounds. Therefore, DUS@MOK-Gel has great development prospects and market value in the field of diabetic wound treatment.
Collapse
Affiliation(s)
- Xi Pan
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Qida Zong
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Bo Fu
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Ye Wang
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xinke Feng
- Department of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Wei Sun
- Department of Biomedical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China.
| | - Yinglei Zhai
- Department of Biomedical Engineering, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
5
|
Shi J, Tang J, Xu J, Jiang N, Yang Y, Chen H, Han Y, Fu X. Applications of hydrogels and nanoparticles in the treatment of traumatic brain injury. Front Bioeng Biotechnol 2025; 12:1515164. [PMID: 39834632 PMCID: PMC11743581 DOI: 10.3389/fbioe.2024.1515164] [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: 10/22/2024] [Accepted: 12/12/2024] [Indexed: 01/22/2025] Open
Abstract
Traumatic brain injury (TBI) represents a significant global public health issue, with effective management posing numerous challenges. The pathophysiology of TBI is typically categorized into two phases: primary and secondary injuries. Secondary injury involves pathophysiological mechanisms such as blood-brain barrier (BBB) disruption, mitochondrial dysfunction, oxidative stress, and inflammatory responses. Current pharmacological strategies often encounter obstacles in treating TBI effectively, primarily due to challenges in BBB penetration, inadequate target site accumulation, and off-target toxicity. Versatile hydrogels and nanoparticles offer potential solutions to these limitations. This review discusses recent progress in utilizing hydrogels and nanoparticles for TBI treatment over the past 5 years, highlighting their relevance to the underlying injury pathophysiology. Hydrogels and nanoparticles demonstrate substantial promise in addressing secondary brain injury, providing a broad spectrum of future therapeutic opportunities.
Collapse
Affiliation(s)
- Jiaying Shi
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jiajia Tang
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Jin Xu
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Ning Jiang
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Yuanwei Yang
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Honglin Chen
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| | - Yuhan Han
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
- Department of Neurosurgery, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xianhua Fu
- Department of Neurosurgery, The Affiliated Suqian First People’s Hospital of Nanjing Medical University, Suqian, China
| |
Collapse
|
6
|
Zhu Y, Shi Z, Pang Y, Zhou Y. Deferoxamine-loaded gelatin methacryloyl hydrogel endue 3D-printed PGCL-hydroxyapatite scaffold with angiogenesis, anti-oxidative and immunoregulatory capacities for facilitating bone healing. Int J Biol Macromol 2025; 295:139509. [PMID: 39756750 DOI: 10.1016/j.ijbiomac.2025.139509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/07/2025]
Abstract
Promoting angiogenesis, alleviating oxidative stress injury and inflammation response are crucial for bone healing. Herein, the deferoxamine (DFO)-loaded gelatin methacryloyl (GelMA) hydrogel coating (GelMA-DFO) was constructed on the 3D-printed poly(Glycolide-Co-Caprolactone)-hydroxyapatite (PGCL-HAP) scaffold. After the hydrogel coating was established, Fourier transform infrared spectroscopy (FT-IR), X-ray diffraction (XRD), scanning electron microscope (SEM) and water contact angle measurement were employed to evaluate the characteristic and the biological properties were assessed. The modification of GelMA-DFO hydrogel endows the PGCL-HAP scaffold with angiogenic, anti-oxidative and immunoregulatory properties, overcoming the limitation of single-functionality in traditional bone tissue engineering (BTE) scaffolds. The improvement of hydrophilicity via GelMA-DFO hydrogel coating promoted cell adhesion onto the scaffold. Due to the load of DFO, the osteogenic and angiogenic effect of the scaffold in vitro were significantly enhanced. Importantly, the PGCL-HAP-DFO scaffold could effectively scavenge reactive oxygen species (ROS) and further polarized macrophage from pro-inflammation phenotype M1 to anti-inflammation phenotype M2. Experimental results in vivo further confirmed that the GelMA-DFO hydrogel coating promoted the osseointegration of PGCL-HAP scaffold via reducing inflammation, further enhancing new bone formation and tissue vascularization. Above, these results demonstrated that GelMA-DFO hydrogel endows PGCL-HAP scaffold with multiple bio-functions, thus accelerating the process of bone regeneration.
Collapse
Affiliation(s)
- Yanlin Zhu
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China
| | - Zuosen Shi
- State Key Lab of Supramolecular Structure and Materials, College of Chemistry, Jilin University, China
| | - Yuxuan Pang
- Department of Prosthodontics, School and Hospital of Stomatology, Jilin University, Changchun, China.
| | - Yanmin Zhou
- Department of Oral Implantology, School and Hospital of Stomatology, Jilin University, Changchun, China.
| |
Collapse
|
7
|
Hu D, Li Y, Yuan W, Ye X, Wang K, Xiao Y, Peng Z, Wu Q, Zeng C, Liu J, Zhou L. Bioactive cationic polymer-based hydrogel with Engrailed-1 gene silencing and microenvironment modulation for enhanced scarless diabetic wound healing. CHEMICAL ENGINEERING JOURNAL 2025; 504:158713. [DOI: 10.1016/j.cej.2024.158713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
8
|
Leng S, Cong R, Xia Y, Kang F. Deferoxamine Accelerates Mandibular Condylar Neck Fracture Early Bone Healing by Promoting Type H Vessel Proliferation. J Oral Rehabil 2025; 52:17-26. [PMID: 39363428 DOI: 10.1111/joor.13869] [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: 06/14/2024] [Revised: 09/05/2024] [Accepted: 09/15/2024] [Indexed: 10/05/2024]
Abstract
BACKGROUND Condylar fractures (CFs) are a common type of maxillofacial trauma, especially in adolescents. Conservative treatment of CF avoids the possible complications of surgical intervention, but prolongs the patient's suffering because of the requirement for extended intermaxillary fixation. Therefore, the development of a new strategy to accelerate the rate of fracture healing to shorten the period of conservative treatment is of great clinical importance. OBJECTIVE To investigate the potential of deferoxamine (DFO) in promoting the healing process of CF in adolescent mice. METHODS Thirty-two 4-week-old male C57BL/6J mice were randomly assigned to four groups: vehicle + sham group, vehicle + CF group, DFO + sham group and DFO + CF group. After constructing the mandibular CF model, mandibular tissue samples were collected respectively at 1, 2 and 4 weeks postoperatively. Radiographic and histomorphometric analyses were employed to assess bone tissue healing and vascular formation. RESULTS Deferoxamine was observed to promote the early bone healing of fracture, both radiologically and histomorphometrically. Furthermore, this enhancement of condylar neck fracture healing was attributed to the upregulation of the hypoxia-inducible factor-1α (HIF-1α) signalling pathway while facilitating the formation of type H vessels. In addition, DFO did not produce significant effects on the condylar neck between vehicle + sham and DFO + sham group. CONCLUSION The application of the HIF-1α inducer DFO can enhance type H vessels expansion thereby accelerating condylar neck fracture healing.
Collapse
Affiliation(s)
- Sijia Leng
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Rong Cong
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Yuxing Xia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | - Feiwu Kang
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital and Dental School of Tongji University, Shanghai, China
- Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| |
Collapse
|
9
|
Teng H, Cho M, Ma Z, Ji X, Jin S, Zhou Y, Xuan M, Seong HA, Li X, Choi SH, Li Q. Preclinical Assessment of Safety and Efficacy of Deferoxamine (DFO)-pretreated Feline Adipose Mesenchymal Stem Cells. In Vivo 2025; 39:267-279. [PMID: 39740892 PMCID: PMC11705137 DOI: 10.21873/invivo.13825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/13/2024] [Accepted: 11/15/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND/AIM This study aimed to investigate the safety and efficacy of deferoxamine (DFO) pretreated feline adipose tissue derived mesenchymal stem cells (fATMSCs) for the treatment of inflammatory disorders. MATERIALS AND METHODS fATMSCs were isolated from feline adipose tissue and characterized using flow cytometry for surface marker expression and differentiation assays for adipogenic, osteogenic, and chondrogenic lineages. Different concentrations of DFO were used to evaluate its impact on fATMSC activity. The therapeutic potential of these preconditioned cells was validated using a mouse model of acute lung injury (ALI) by LPS injection. Comprehensive evaluations, including clinical, hematological, and radiological assessments, were conducted before and after intravenous injection of preconditioned cells in three feline subjects. RESULTS 25 μM DFO pretreatment significantly up-regulated immunomodulatory genes (Tgfb, Hgf, and Tsg-6) in fATMSCs. In the mouse ALI model, DFO-pretreated fATMSCs exhibited enhanced anti-inflammatory effects, reducing inflammatory cytokines (Tnfa, Il1b, Il6). Clinical safety assessment in felines showed no immediate adverse effects, structural alterations, or tumorigenesis. CONCLUSION Utilizing a mouse model of acute lung injury, we demonstrated the potential of DFO-pretreated fATMSCs as a safe and effective therapeutic approach for inflammatory disorders.
Collapse
Affiliation(s)
- Hailong Teng
- Department of Veterinary Medicine, Yanbian University, Yanji, P.R. China
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji, P.R. China
| | - Minkyung Cho
- Department of Biological Sciences and Biotechnology, Chungbuk National University College of Natural Science, Cheongju, Republic of Korea
| | | | - Xinpeng Ji
- Department of Veterinary Medicine, Yanbian University, Yanji, P.R. China
| | - Shiyu Jin
- Department of Veterinary Medicine, Yanbian University, Yanji, P.R. China
| | - Yuze Zhou
- Department of Veterinary Medicine, Yanbian University, Yanji, P.R. China
| | - Meifu Xuan
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji, P.R. China
| | - Hyun-A Seong
- Department of Biological Sciences and Biotechnology, Chungbuk National University College of Natural Science, Cheongju, Republic of Korea
| | - Xiangzi Li
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji, P.R. China
| | - Seong-Ho Choi
- Department of Biological Sciences and Biotechnology, Chungbuk National University College of Natural Science, Cheongju, Republic of Korea;
| | - Qiang Li
- Department of Veterinary Medicine, Yanbian University, Yanji, P.R. China;
- Engineering Research Center of North-East Cold Region Beef Cattle Science & Technology Innovation, Ministry of Education, Yanbian University, Yanji, P.R. China
| |
Collapse
|
10
|
Phan CM, Luu CH, Murugesan M, Nguyen TNQ, Ha NYN, Ngo HL, Nguyen NDH, Pan Z, Phan VHG, Li Y, Thambi T. Injectable gelatin-pectin hydrogel for dental tissue engineering: Enhanced angiogenesis and antibacterial efficacy for pulpitis therapy. Int J Biol Macromol 2025; 284:137939. [PMID: 39592046 DOI: 10.1016/j.ijbiomac.2024.137939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 11/28/2024]
Abstract
Pulpitis is inflammation of the dental pulp, often caused by bacterial infection from untreated cavities, leading to pain. The main challenge in treatment is eliminating infection while preserving tooth vitality. This study aims to address this challenge by developing a hydrogel for convenient insertion into the root canal system, securely attaching to dentin walls. An injectable hydrogel system is developed by chemically cross-linking natural polysaccharide pectin with gelatin (GPG) through reversible Schiff base reaction. The GPG system was then used to encapsulate and release drugs, such as ciprofloxacin (CIP) for infection prevention and deferoxamine (DFO) for promoting blood vessel proliferation and reducing inflammatory reactions. The GPGs absorbed significant amounts of CIP and DFO, enabling sustained release over a nearly ten-day period. When subcutaneously implanted, the GPGs formed stable gel depots, with only 50 % of the gels degrading after 3 weeks, indicating a sustained biodegradation pattern. Additionally, the GPG system demonstrated excellent antibacterial activity against both gram-negative and gram-positive bacteria. Results from in vitro scratch healing tests and in ovo chorioallantoic membrane chick model tests showed promising biocompatibility and promotion of vascular proliferation by the GPG. This study heralds a novel frontier in endodontic therapeutics, poised to potentially enable dental pulp regeneration.
Collapse
Affiliation(s)
- Chau My Phan
- College of Materials and Textile Engineering & Nanotechnology Research Institute, Jiaxing University, Jiaxing 314001, Zhejiang Province, PR China; Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Cuong Hung Luu
- School of Environment and Science, Griffith University, Nathan, QLD 4111, Australia; Queensland Micro- and Nanotechnology Centre, Griffith University, Nathan, QLD 4111, Australia
| | - Mohanapriya Murugesan
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin si, Gyeonggi do 17104, Republic of Korea
| | - Thi-Nhu-Quynh Nguyen
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Nhu-Y Ngoc Ha
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Huong Lan Ngo
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Ngoc-Dan Ho Nguyen
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Zhouyi Pan
- College of Materials and Textile Engineering & Nanotechnology Research Institute, Jiaxing University, Jiaxing 314001, Zhejiang Province, PR China
| | - V H Giang Phan
- Biomaterials and Nanotechnology Research Group, Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh City, Vietnam.
| | - Yi Li
- College of Materials and Textile Engineering & Nanotechnology Research Institute, Jiaxing University, Jiaxing 314001, Zhejiang Province, PR China.
| | - Thavasyappan Thambi
- Graduate School of Biotechnology, College of Life Sciences, Kyung Hee University, Yongin si, Gyeonggi do 17104, Republic of Korea.
| |
Collapse
|
11
|
Zhang J, Zhao S, Zhou Y, Liang H, Zhao L, Tan H. Carboxymethyl Chitosan-Based Antioxidant Hydrogel Accelerates Diabetic Wound Healing. Adv Healthc Mater 2024:e2403198. [PMID: 39716833 DOI: 10.1002/adhm.202403198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/14/2024] [Indexed: 12/25/2024]
Abstract
Diabetic wound healing is hampered due to oxidative stress, exacerbated inflammation, and impaired angiogenesis in the wounds. A pH-sensitive antioxidant hydrogel based on carboxymethyl chitosan (CMCS), oligoprocyanidins (OPC), and oxide dextran (Oxd) is prepared to accelerate diabetic wound healing. The hydrogel network is formed via imine and hydrogen bonding interactions in the presence of hydroxyl, amino, and aldehyde groups, and deferoxamine (DFO) is incorporated into the hydrogel. The hydrogel shows pH-triggered drug release, a good antioxidant, and anti-inflammatory properties, and can promote tube formation and cell migration in vitro. Moreover, the hydrogel can accelerate wound healing in streptozotocin (STZ)-induced diabetic mice by regulating the inflammation environment with up-regulation of anti-inflammatory factors (IL-4 and IL-10) and down-regulation of pro-inflammatory factors (TNF-α and IL-6), and promoting angiogenesis with up-regulation of HIF-1α, VEGF, and CD31. Thus, the pH-sensitive antioxidant hydrogel provides a promising therapeutic strategy for diabetic wound healing.
Collapse
Affiliation(s)
- Jiaying Zhang
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 311215, China
| | - Shifang Zhao
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 311215, China
| | - Yifan Zhou
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 311215, China
| | - Hongze Liang
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 311215, China
| | - Lingling Zhao
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, 311215, China
| | - Hui Tan
- Infectious Diseases Department, Shenzhen Children's Hospital, Shenzhen, 518038, China
| |
Collapse
|
12
|
Saber S, Abdelhady R, Elhemely MA, Elmorsy EA, Hamad RS, Abdel-Reheim MA, El-kott AF, AlShehri MA, Morsy K, Negm S, Kira AY. Nanoscale Systems for Local Activation of Hypoxia-Inducible Factor-1 Alpha: A New Approach in Diabetic Wound Management. Int J Nanomedicine 2024; 19:13735-13762. [PMID: 39723173 PMCID: PMC11669355 DOI: 10.2147/ijn.s497041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 11/03/2024] [Indexed: 12/28/2024] Open
Abstract
Chronic wounds in diabetic patients experience significant clinical challenges due to compromised healing processes. Hypoxia-inducible factor-1 alpha (HIF-1α) is a critical regulator in the cellular response to hypoxia, enhancing angiogenesis and tissue restoration. Nevertheless, the cellular response to the developed chronic hypoxia within diabetes is impaired, likely due to the destabilization of HIF-1α via degradation by prolyl hydroxylase domain (PHD) enzymes. Researchers have extensively explored HIF-1α activation as a potential pathway for diabetic wound management, focusing mainly on deferoxamine (DFO) as a potent agent to stabilize HIF-1α. This review provides an update of the other recent pharmacological agents managing HIF-1α activation, including novel PHD inhibitors (roxadustat and daprodustat) and Von Hippel-Lindau protein (VHL) antagonists, which could be potential alternatives for the local treatment of diabetic wounds. Furthermore, it highlights how localized delivery via advanced nanostructures can enhance the efficacy of these novel therapies. Importantly, by addressing these points, the current review can offer a promising area for research. Given that, these novel drugs have minimal applications in diabetic wound healing, particularly in the context of local application through nanomaterials. This gap presents an exciting opportunity for further investigation, as combining these drugs with localized nanotechnology could avoid undesired systemic side effects and sustain drug release within wound site, offering a transformative platform for diabetes wound treatment.
Collapse
Affiliation(s)
- Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| | - Rasha Abdelhady
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Fayoum University, Fayoum, 63514, Egypt
| | - Mai A Elhemely
- School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M20 4BX, UK
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
| | - Elsayed A Elmorsy
- Department of Pharmacology and Therapeutics, College of Medicine, Qassim University, Buraidah, 51452, Saudi Arabia
| | - Rabab S Hamad
- Biological Sciences Department, College of Science, King Faisal University, Al Ahsa, 31982, Saudi Arabia
| | - Mustafa Ahmed Abdel-Reheim
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Beni-Suef University, Beni Suef, 62521, Egypt
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, 11961, Saudi Arabia
| | - Attalla F El-kott
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Mohammed A AlShehri
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Kareem Morsy
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia
| | - Sally Negm
- Department of Life Sciences, College of Science and Art Mahyel Aseer, King Khalid University, Abha, 62529, Saudi Arabia
| | - Ahmed Y Kira
- Department of Pharmaceutics, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, 11152, Egypt
| |
Collapse
|
13
|
Chen L, Jiang Z, Zhou H, Zhang H, Huang C, Wen Q, Liu X, He Y, Shi P, Liu K, Yang L. Effect of hyaluronic acid on the formation of acellular dermal matrix-based interpenetrating network sponge scaffolds for accelerating diabetic wound healing through photothermal warm bath. Int J Biol Macromol 2024; 283:137268. [PMID: 39505193 DOI: 10.1016/j.ijbiomac.2024.137268] [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: 07/26/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/08/2024]
Abstract
Adequate vascularization essential for delivering nutrients critical to wound healing, yet impaired angiogenesis is a major barrier in diabetic wound repair. This study investigates the impact of hyaluronic acid on interpenetrating network sponge scaffolds derived from an acellular dermal matrix, with the aim of enhancing vascularization and healing of diabetic wounds via photothermal warm bath therapy. We prepared three-dimensional porous sponges (H1P4D2@DFO) using molecular interpenetration and ion crosslinking of porcine acellular dermal matrix (PADM), hyaluronic acid, and polydopamine nanoparticles loaded with deferoxamine mesylate (PDA@DFO). This resulting extracellular matrix-based sponge demonstrated properties suitable for wound repair, including high cell adhesion, biocompatibility, bioactivity, porosity (85 %), and water absorption (4500 %). The near-infrared (NIR) photothermal effect of PDA@DFO and the sustained release of deferoxamine mesylate (DFO) enhanced wound vascularization within the wound site. These findings suggest that our sponge scaffold can simulate skin-like structures and establish a supportive microenvironment conducive to microvascular reconstruction. By combining the photothermal warm bath approach with the scaffold's tailored 3D structure, we observed enhanced angiogenesis and accelerated diabetic wound healing, indicating potential clinical applications of these scaffolds in chronic wound management.
Collapse
Affiliation(s)
- Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China
| | - Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China
| | - Hai Zhou
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China
| | - Huihui Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China
| | - Chaoyang Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China
| | - Qiulan Wen
- Department of Orthopaedic Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xiaoyang Liu
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China
| | - Yufang He
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China
| | - Pengwei Shi
- Emergency Department, Nanfang Hospital, Southern Medical University, Guangzhou 50515, China.
| | - Kun Liu
- Experimental Education/Administration Centre, National Demonstration Centre for Experimental Education of Basic Medical Sciences, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China.
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Jingxi Street, Baiyun District, Guangdong 510515, China.
| |
Collapse
|
14
|
Sheppard NT, Daniel MC, Nelson NS, Donneys A, Buchman SR. Optimizing immunofluorescent staining of H vessels within an irradiated fracture callus in paraffin-embedded tissue samples. J Histotechnol 2024; 47:173-179. [PMID: 38957981 DOI: 10.1080/01478885.2024.2371060] [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: 09/22/2023] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
H vessels are an essential link in angiogenic-osteogenic coupling and orchestrate the process of bone healing. H vessels are critically deficient in the setting of radiation-induced fractures, which have been reported to occur in up to 25% of patients undergoing radiotherapy. By increasing H-vessel proliferation, Deferoxamine (DFO) revitalizes the physiologic response to skeletal injury and accelerates irradiated fracture repair. H-vessel quantification is therefore an important outcome measure in histologic analysis of bone healing. However, an optimized protocol for staining H vessels in formalin-fixed paraffin-embedded (FFPE) tissue sections has not been reported. With this protocol, we describe a method of staining FFPE bone samples with minimal background fluorescence and high signal-to-noise ratio. We examined mandibular specimens in a rat model of bone healing from a range of fracture conditions, including healthy bone (Fx), irradiated bone (XFx), and irradiated bone with DFO treatment (XFx-DFO). Quantitative analysis revealed a significant increase of H vessels in the XFxDFO group compared to both the Fx and XFx groups. By optimizing immunofluorescent staining of H vessels in FFPE samples across a range of fracture conditions, we offer investigators an efficacious means of producing reliable imaging for quantitative analysis of H vessels in an irradiated fracture callus.
Collapse
Affiliation(s)
- Nathan T Sheppard
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Melissa C Daniel
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Noah S Nelson
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Alexis Donneys
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Steven R Buchman
- Department of Surgery, Section of Plastic and Reconstructive Surgery, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
15
|
Dai F, Zhang J, Chen F, Chen X, Lee CJ, Liang H, Zhao L, Tan H. A Multi-Responsive Hydrogel Combined With Mild Heat Stimulation Promotes Diabetic Wound Healing by Regulating Inflammatory and Enhancing Angiogenesis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2408783. [PMID: 39435670 PMCID: PMC11633493 DOI: 10.1002/advs.202408783] [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: 09/27/2024] [Indexed: 10/23/2024]
Abstract
The repair of diabetic wound still encounters huge challenges, such as disordered inflammatory regulation and impaired neovascularization. Here, a pH/ROS/glucose responsive and photothermal hydrogel is developed for diabetic wound healing. The hydrogel is formed through cross-linkage between phenylboronic acid-modified carboxymethyl chitosan (CMCS-PBA) and oxide dextran (OXD), utilizing Schiff base and phenylboronate ester bonds. Additionally, insulin-like growth factor 1 C domain (IGF-1C) and deferoxamine-loaded polydopamine nanoparticles (D@P) are incorporated into the hydrogel. The hydrogel demonstrates sustained drug release, excellent photo thermal effect, prominent antioxidant, antibacterial and anti-inflammatory activities, desirable mechanical and tissue adhesive properties, enhanced tube formation, and cell migration. Furthermore, the hydrogel combined with mild heat treatment can regulate chronic inflammation by promoting the transformation of macrophages from M1 phenotype to M2 phenotype and enhance angiogenesis by up-regulating the expression levels of angiogenesis-related factors such as hypoxia-inducible factor-1 alpha (HIF-1α), vascular endothelial growth factor (VEGF), CD31, and α-SMA, thus greatly accelerates the wound healing in streptozotocin (STZ)-induced diabetic mice. Therefore, this multi-responsive and multifunctional hydrogel holds potential as a therapeutic strategy for diabetic wounds.
Collapse
Affiliation(s)
- Fanjia Dai
- School of Material Science and Chemical EngineeringNingbo UniversityNingbo315211China
- Center for Child Care and Mental HealthShenzhen Children's Hospital Affiliated to Shantou University Medical CollegeShenzhen518026China
| | - Jiaying Zhang
- School of Material Science and Chemical EngineeringNingbo UniversityNingbo315211China
| | - Fengjiao Chen
- School of Material Science and Chemical EngineeringNingbo UniversityNingbo315211China
| | - Xianwu Chen
- The Affiliated Hospital of Medical SchoolNingbo UniversityNingbo315211China
| | | | - Hongze Liang
- School of Material Science and Chemical EngineeringNingbo UniversityNingbo315211China
| | - Lingling Zhao
- School of Material Science and Chemical EngineeringNingbo UniversityNingbo315211China
| | - Hui Tan
- Center for Child Care and Mental HealthShenzhen Children's Hospital Affiliated to Shantou University Medical CollegeShenzhen518026China
| |
Collapse
|
16
|
Tang X, Zhou F, Wang S, Wang G, Bai L, Su J. Bioinspired injectable hydrogels for bone regeneration. J Adv Res 2024:S2090-1232(24)00486-7. [PMID: 39505143 DOI: 10.1016/j.jare.2024.10.032] [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: 01/07/2024] [Revised: 09/28/2024] [Accepted: 10/27/2024] [Indexed: 11/08/2024] Open
Abstract
The effective regeneration of bone/cartilage defects remains a significant clinical challenge, causing irreversible damage to millions annually.Conventional therapies such as autologous or artificial bone grafting often yield unsatisfactory outcomes, emphasizing the urgent need for innovative treatment methods. Biomaterial-based strategies, including hydrogels and active scaffolds, have shown potential in promoting bone/cartilage regeneration. Among them, injectable hydrogels have garnered substantial attention in recent years on account of their minimal invasiveness, shape adaptation, and controlled spatiotemporal release. This review systematically discusses the synthesis of injectable hydrogels, bioinspired approaches-covering microenvironment, structural, compositional, and bioactive component-inspired strategies-and their applications in various bone/cartilage disease models, highlighting bone/cartilage regeneration from an innovative perspective of bioinspired design. Taken together, bioinspired injectable hydrogels offer promising and feasible solutions for promoting bone/cartilage regeneration, ultimately laying the foundations for clinical applications. Furthermore, insights into further prospective directions for AI in injectable hydrogels screening and organoid construction are provided.
Collapse
Affiliation(s)
- Xuan Tang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Fengjin Zhou
- Department of Orthopaedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an 710000, China
| | - Sicheng Wang
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Department of Orthopedics Trauma, Shanghai Zhongye Hospital, Shanghai 201900, China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China; Wenzhou Institute of Shanghai University, Wenzhou 325000, China.
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
17
|
Zhao Y, Chen J, Zhou M, Zhang G, Wu W, Wang Z, Sun J, Zhong A. Desferrioxamine-Laden Nanofibrous Scaffolds with Efficient Angiogenesis for Accelerating Diabetic Wound Healing. Int J Nanomedicine 2024; 19:10551-10568. [PMID: 39435042 PMCID: PMC11492907 DOI: 10.2147/ijn.s477109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/06/2024] [Indexed: 10/23/2024] Open
Abstract
Background Delayed diabetic wound healing is one of the clinical difficulties, the main reason is the limited angiogenesis ability. Deferriamine (DFO) is an iron chelating agent that can induce angiogenesis, but its application is limited due to its short half-life. Increasing the load and slow release performance of desferriamine is beneficial to accelerate diabetic wound healing. Materials and Methods In this study, we developed collagen (Col)-graphene oxide (GO) and (1% w/w) DFO-loaded nanofiber electrospinning scaffolds (DCG) using the electrospinning technique. We tested the physicochemical properties, drug release performance, and vascularization biological function of the scaffolds, and finally evaluated the promotion of full-thickness wound healing in the diabetic rat models. Results The results showed that DCG scaffolds have good mechanical properties and water-holding capacity and can release DFO continuously for 14 days. In vitro, the novel DCG scaffold exhibited good biocompatibility, with the up-regulation at the gene level of VEGF and its regulator HIF-1α, promoters of angiogenesis. This was verified in vivo, as the scaffold enhanced granulation tissue formation and improved neovascularization, thereby accelerating wound healing when applied to full-thickness defects on the back of diabetic rats. Conclusion The DCG nanofiber scaffold prepared in this study has good biocompatibility and vascularization ability, and improves the microenvironment in vivo, and has a good application prospect in diabetic wound repair.
Collapse
Affiliation(s)
- Yang Zhao
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| | - Jialong Chen
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| | - Muran Zhou
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| | - Guo Zhang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| | - Wenhao Wu
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| | - Zhenxing Wang
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| | - Jiaming Sun
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| | - Aimei Zhong
- Department of Plastic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Wuhan Clinical Research Center for Superficial Organ Reconstruction, Wuhan, 430022, People’s Republic of China
| |
Collapse
|
18
|
Chen Z, Zhang J, Lee FY, Kyriakides TR. Bone-derived extracellular matrix hydrogel from thrombospondin-2 knock-out mice for bone repair. Acta Biomater 2024; 186:85-94. [PMID: 39134130 PMCID: PMC11500023 DOI: 10.1016/j.actbio.2024.08.011] [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: 04/10/2024] [Revised: 08/03/2024] [Accepted: 08/07/2024] [Indexed: 08/26/2024]
Abstract
Bone extracellular matrix (ECM) has been shown to mimic aspects of the tissue's complex microenvironment, suggesting its potential role in promoting bone repair. However, current ECM-based therapies suffer from limitations such as inefficient scale-up, lack of mechanical integrity, and sub-optimal efficacy. Here, we fabricated hydrogels from decellularized ECM (dECM) from wild type (WT) and thrombospondin-2 knock-out (TSP2KO) mouse bones. TSP2KO bone ECM hydrogel was found to have distinct mechanical properties and collagen fibril assembly from WT. Furthermore, TSP2KO hydrogel promoted mesenchymal stem cell (MSC) attachment, spreading, and invasion in vitro. Similarly, it promoted formation of tube-like structures by human umbilical vein endothelial cells (HUVECs). When applied to a murine calvarial defect model, TSP2KO hydrogel enhanced repair, in part, due to increased angiogenesis. Our study suggests the pro-angiogenic therapeutic potential of TSP2KO bone ECM hydrogel in bone repair. STATEMENT OF SIGNIFICANCE: The study describes the first successful preparation of a novel hydrogel made from decellularized bones from wild-type mice and mice lacking thrombospondin-2 (TSP2). Hydrogels from TSP2 knock-out (TSP2KO) bones have unique characteristics in structure and biomechanics. These gels interacted well with cells in vitro and helped repair damaged bone in a mouse model. Therefore, TSP2KO bone-derived hydrogel has translational potential for accelerating repair of bone defects that are otherwise difficult to heal. This study not only creates a new material with promise for accelerated healing, but also validates tunability of native biomaterials by genetic engineering.
Collapse
Affiliation(s)
- Zhuoyue Chen
- Department of Pathology, Yale University, New Haven, CT 06519, USA; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA
| | - Junqi Zhang
- Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA
| | - Francis Y Lee
- Department of Orthopaedics and Rehabilitation, Yale University, New Haven, CT 06519, USA
| | - Themis R Kyriakides
- Department of Pathology, Yale University, New Haven, CT 06519, USA; Vascular Biology and Therapeutics Program, Yale University, New Haven, CT 06519, USA; Department of Biomedical Engineering, Yale University, New Haven, CT 06519, USA.
| |
Collapse
|
19
|
Lou S, Liu YX, Xia C, Zhang Q, Deng L, Tang JJ. Novel meroterpene-like compounds inhibit ferroptosis through Fe 2+ chelation. Int J Biochem Cell Biol 2024; 173:106610. [PMID: 38879152 DOI: 10.1016/j.biocel.2024.106610] [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: 03/29/2024] [Revised: 05/30/2024] [Accepted: 06/09/2024] [Indexed: 07/22/2024]
Abstract
Colorectal cancer (CRC) is the third most common type of cancer in the world. It is characterized by complex crosstalk between various signaling pathways, as a result of which it is highly challenging to identify optimal therapeutic targets and design treatment strategies. In this study, we tested the effect of 700 compounds on the CRC cell line HT-29 by using the sulforhodamine B assay and screened out 17 compounds that exhibited high toxicity (indicated by an inhibition rate of ≥75 % when applied at a concentration of 10 µM) against the HT-29 cell line. Next, we investigated the mechanisms underlying the effects of these 17 highly toxic compounds. The results of ferroptosis analysis and electron microscopy showed that compounds 575 and 578 were able to significantly reverse RSL3-induced increase in ferroptosis, while compound 580 had a less pronounced ferroptosis-regulating effect. In subsequent experiments, western blotting showed that compounds 575, 578, and 580, which belong to a class of meroterpene-like compounds that affect ferroptosis, do not induce autophagy or apoptosis in the CRC cell line. Instead, Fe2+ chelation experiments showed that these three compounds can serve as iron chelators by chelating Fe2+ at a 1:1 (chelator: Fe2+) ratio. Specifically, the aldehyde and hydroxyl groups of the benzene ring in these compounds may chelate Fe2+, thus reducing Fe2+ levels in cells and inhibiting ferroptosis. These results indicate that these novel meroterpene-like compounds are potential therapeutic small-molecule candidates for targeting ferroptosis in tumors.
Collapse
Affiliation(s)
- Shiyang Lou
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Yan-Xiang Liu
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Chao Xia
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Qiang Zhang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China
| | - Lu Deng
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shanxi 712100, China; Shenzhen Research Institute, Northwest A&F University, Shenzhen, Guangdong 518000, China.
| | - Jiang-Jiang Tang
- College of Chemistry & Pharmacy, Northwest A&F University, Yangling, Shanxi 712100, China.
| |
Collapse
|
20
|
Lu C, Chen M, Zhao Y, Zhan Y, Wei X, Lu L, Yang M, Gong X. A Co-MOF encapsulated microneedle patch activates hypoxia induction factor-1 to pre-protect transplanted flaps from distal ischemic necrosis. Acta Biomater 2024; 184:171-185. [PMID: 38871202 DOI: 10.1016/j.actbio.2024.06.008] [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: 03/17/2024] [Revised: 05/25/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024]
Abstract
Avoiding ischemic necrosis after flap transplantation remains a significant clinical challenge. Developing an effective pretreatment method to promote flap survival postoperatively is crucial. Cobalt chloride (CoCl2) can increase cell tolerance to ischemia and hypoxia condition by stimulating hypoxia-inducible factor-1 (HIF-1) expression. However, the considerable toxic effects severely limit the clinical application of CoCl2. In this study, cobalt-based metal-organic frameworks (Co-MOF) encapsulated in a microneedle patch (Co-MOF@MN) was developed to facilitate the transdermal sustained release of Co2+ for rapid, minimally invasive rapid pretreatment of flap transplantation. The MN patch was composed of a fully methanol-based two-component cross-linked polymer formula, with a pyramid structure and high mechanical strength, which satisfied the purpose of penetrating the skin stratum corneum of rat back to achieve subcutaneous vascular area administration. Benefiting from the water-triggered disintegration of Co-MOF and the transdermal delivery via the MN patch, preoperative damage and side effects were effectively mitigated. Moreover, in both the oxygen-glucose deprivation/recovery (OGD/R) cell model and the rat dorsal perforator flap model, Co-MOF@MN activated the HIF-1α pathway and its associated downstream proteins, which reduced reperfusion oxidative damage, improved blood supply in choke areas, and increased flap survival rates post-transplantation. This preprotection strategy, combining MOF nanoparticles and the MN patch, meets the clinical demands for trauma minimization and uniform administration in flap transplantation. STATEMENT OF SIGNIFICANCE: Cobalt chloride (CoCl2) can stimulate the expression of hypoxia-inducible factor (HIF-1) and improve the tolerance of cells to ischemia and hypoxia conditions. However, the toxicity and narrow therapeutic window of CoCl2 severely limit its clinical application. Herein, we explored the role of Co-MOF as a biocompatible nanocage for sustained release of Co2+, showing the protective effect on vascular endothelial cells in the stress model of oxygen-glucose deprivation. To fit the clinical needs of minimal trauma in flap transplantation, a Co-MOF@MN system was developed to achieve local transdermal delivery at the choke area, significantly improving blood supply opening and flap survival rate. This strategy of two-step delivery of Co2+ realized the enhancement of biological functions while ensuring the biosafety.
Collapse
Affiliation(s)
- Cheng Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Miao Chen
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Yuanyuan Zhao
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Yongxin Zhan
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Xin Wei
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China
| | - Laijin Lu
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China; Orthopedics Central Laboratory, Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun 130021, PR China
| | - Mingxi Yang
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China; Orthopedics Central Laboratory, Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun 130021, PR China.
| | - Xu Gong
- Department of Hand and Podiatric Surgery, Orthopedics Center, The First Hospital of Jilin University, Jilin University, Changchun 130031, PR China; Orthopedics Central Laboratory, Institute of Translational Medicine, The First Hospital of Jilin University, Jilin University, Changchun 130021, PR China.
| |
Collapse
|
21
|
Tan Y, Xu C, Liu Y, Bai Y, Li X, Wang X. Sprayable and self-healing chitosan-based hydrogels for promoting healing of infected wound via anti-bacteria, anti-inflammation and angiogenesis. Carbohydr Polym 2024; 337:122147. [PMID: 38710554 DOI: 10.1016/j.carbpol.2024.122147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 05/08/2024]
Abstract
Treatment of infected wound by simultaneously eliminating bacteria and inducing angiogenesis to promote wound tissue regeneration remains a clinical challenge. Dynamic and reversable hydrogels can adapt to irregular wound beds, which have raised great attention as wound dressings. Herein, a sprayable chitosan-based hydrogel (HPC/CCS/ODex-IGF1) was developed using hydroxypropyl chitosan (HPC), caffeic acid functionalized chitosan (CCS), oxidized dextran (ODex) to crosslink through the dynamic imine bond, which was pH-responsive to the acidic microenvironment and could controllably release insulin growth factor-1 (IGF1). The HPC/CCS/ODex-IGF1 hydrogels not only showed self-healing, self-adaptable and sprayable properties, but also exhibited excellent antibacterial ability, antioxidant property, low-cytotoxicity and angiogenetic activity. In vivo experiments demonstrated that hydrogels promoted tissue regeneration and healing of bacteria-infected wound with a rate of approximately 98.4 % on day 11 by eliminating bacteria, reducing inflammatory and facilitating angiogenesis, demonstrating its great potential for wound dressing.
Collapse
Affiliation(s)
- Yujiao Tan
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China
| | - Changliang Xu
- Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine Prevention and Treatment of Tumor, The First Clinical Medical College of Nanjing University of Chinese Medicine, 138 Xianlin Avenue, Nanjing 210023, China
| | - Ying Liu
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China
| | - Yun Bai
- Jiangsu Province People's Hospital (Nanjing Medical University First Affiliated Hospital), 300 Guangzhou Road, Nanjing 210029, China
| | - Xiaoyun Li
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China.
| | - Xiaoying Wang
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou 510640, China.
| |
Collapse
|
22
|
Bieri S, Möller B, Amsler J. Ferroptosis in Arthritis: Driver of the Disease or Therapeutic Option? Int J Mol Sci 2024; 25:8212. [PMID: 39125782 PMCID: PMC11311315 DOI: 10.3390/ijms25158212] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/12/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Ferroptosis is a form of iron-dependent regulated cell death caused by the accumulation of lipid peroxides. In this review, we summarize research on the impact of ferroptosis on disease models and isolated cells in various types of arthritis. While most studies have focused on rheumatoid arthritis (RA) and osteoarthritis (OA), there is limited research on spondylarthritis and crystal arthropathies. The effects of inducing or inhibiting ferroptosis on the disease strongly depend on the studied cell type. In the search for new therapeutic targets, inhibiting ferroptosis in chondrocytes might have promising effects for any type of arthritis. On the other hand, ferroptosis induction may also lead to a desired decrease of synovial fibroblasts in RA. Thus, ferroptosis research must consider the cell-type-specific effects on arthritis. Further investigation is needed to clarify these complexities.
Collapse
Affiliation(s)
- Shania Bieri
- Faculty of Medicine, University of Bern, 3012 Bern, Switzerland
| | - Burkhard Möller
- Department of Rheumatology and Immunology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland;
| | - Jennifer Amsler
- Department of Rheumatology and Immunology, Bern University Hospital, University of Bern, 3010 Bern, Switzerland;
- Department for BioMedical Research DBMR, University of Bern, 3008 Bern, Switzerland
| |
Collapse
|
23
|
Guo Q, Yin T, Huang W, Nan R, Xiang T, Zhou S. Hybrid Hydrogels for Immunoregulation and Proangiogenesis through Mild Heat Stimulation to Accelerate Whole-Process Diabetic Wound Healing. Adv Healthc Mater 2024; 13:e2304536. [PMID: 38519046 DOI: 10.1002/adhm.202304536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/27/2024] [Indexed: 03/24/2024]
Abstract
Intense and persistent oxidative stress, excessive inflammation, and impaired angiogenesis severely hinder diabetic wound healing. Bioactive hydrogel dressings with immunoregulatory and proangiogenic properties have great promise in treating diabetic wounds. However, the therapeutic effects of dressings always depend on drugs with side effects, expensive cytokines, and cell therapies. Herein, a novel dynamic borate-bonds crosslinked hybrid multifunctional hydrogel dressings with photothermal properties are developed to regulate the microenvironment of diabetic wound sites and accelerate the whole process of its healing without additional medication. The hydrogel is composed of phenylboronic acid-modified chitosan and hyaluronic acid (HA) crosslinked by tannic acid (TA) through borate bonds and Prussian blue nanoparticles (PBNPs) with photothermal response characteristics are embedded in the polymer networks. The results indicate hydrogels show inherent broad-spectrum antioxidative activities through the integrated interaction of borate bonds, TA, and PBNPs. Meanwhile, combined with the regulation of macrophage phenotype by HA, the inflammatory microenvironment of diabetic wounds is transformed. Moreover, the angiogenesis is then enhanced by the mild photothermal effect of PBNPs, followed by promoted epithelialization and collagen deposition. In summary, this hybrid hydrogel system accelerates all stages of wound repair through antioxidative stress, immunomodulation, and proangiogenesis, showing great potential applications in diabetic wound management.
Collapse
Affiliation(s)
- Qianru Guo
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Tianyu Yin
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Wei Huang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Rui Nan
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Tao Xiang
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu, 610031, P. R. China
- Key Laboratory of Advanced Technologies of Materials Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu, 610031, P. R. China
| |
Collapse
|
24
|
Zhao X, Zhuang Y, Cao Y, Cai F, Lv Y, Zheng Y, Yang J, Shi X. Electrospun Biomimetic Periosteum Capable of Controlled Release of Multiple Agents for Programmed Promoting Bone Regeneration. Adv Healthc Mater 2024; 13:e2303134. [PMID: 38348511 DOI: 10.1002/adhm.202303134] [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: 09/18/2023] [Revised: 01/29/2024] [Indexed: 05/08/2024]
Abstract
The effective repair of large bone defects remains a major challenge due to its limited self-healing capacity. Inspired by the structure and function of the natural periosteum, an electrospun biomimetic periosteum is constructed to programmatically promote bone regeneration using natural bone healing mechanisms. The biomimetic periosteum is composed of a bilayer with an asymmetric structure in which an aligned electrospun poly(ε-caprolactone)/gelatin/deferoxamine (PCL/GEL/DFO) layer mimics the outer fibrous layer of the periosteum, while a random coaxial electrospun PCL/GEL/aspirin (ASP) shell and PCL/silicon nanoparticles (SiNPs) core layer mimics the inner cambial layer. The bilayer controls the release of ASP, DFO, and SiNPs to precisely regulate the inflammatory, angiogenic, and osteogenic phases of bone repair. The random coaxial inner layer can effectively antioxidize, promoting cell recruitment, proliferation, differentiation, and mineralization, while the aligned outer layer can promote angiogenesis and prevent fibroblast infiltration. In particular, different stages of bone repair are modulated in a rat skull defect model to achieve faster and better bone regeneration. The proposed biomimetic periosteum is expected to be a promising candidate for bone defect healing.
Collapse
Affiliation(s)
- Xingkai Zhao
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yu Zhuang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yongjian Cao
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Fengying Cai
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yicheng Lv
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Yunquan Zheng
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Jianmin Yang
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| | - Xianai Shi
- College of Biological Science and Engineering, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
- Fujian Key Laboratory of Medical Instrument and Pharmaceutical Technology, Fuzhou University, No. 2 Xueyuan Road, Fuzhou, 350108, China
| |
Collapse
|
25
|
Shen H, Ma Y, Qiao Y, Zhang C, Chen J, Zhang R. Application of Deferoxamine in Tissue Regeneration Attributed to Promoted Angiogenesis. Molecules 2024; 29:2050. [PMID: 38731540 PMCID: PMC11085206 DOI: 10.3390/molecules29092050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 04/19/2024] [Accepted: 04/25/2024] [Indexed: 05/13/2024] Open
Abstract
Deferoxamine, an iron chelator used to treat diseases caused by excess iron, has had a Food and Drug Administration-approved status for many years. A large number of studies have confirmed that deferoxamine can reduce inflammatory response and promote angiogenesis. Blood vessels play a crucial role in sustaining vital life by facilitating the delivery of immune cells, oxygen, and nutrients, as well as eliminating waste products generated during cellular metabolism. Dysfunction in blood vessels may contribute significantly to the development of life-threatening diseases. Anti-angiogenesis therapy and pro-angiogenesis/angiogenesis strategies have been frequently recommended for various diseases. Herein, we describe the mechanism by which deferoxamine promotes angiogenesis and summarize its application in chronic wounds, bone repair, and diseases of the respiratory system. Furthermore, we discuss the drug delivery system of deferoxamine for treating various diseases, providing constructive ideas and inspiration for the development of new treatment strategies.
Collapse
Affiliation(s)
- Haijun Shen
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China; (Y.M.); (Y.Q.); (C.Z.); (J.C.)
| | - Yane Ma
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China; (Y.M.); (Y.Q.); (C.Z.); (J.C.)
| | - Yi Qiao
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China; (Y.M.); (Y.Q.); (C.Z.); (J.C.)
| | - Chun Zhang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China; (Y.M.); (Y.Q.); (C.Z.); (J.C.)
| | - Jialing Chen
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212013, China; (Y.M.); (Y.Q.); (C.Z.); (J.C.)
| | - Ran Zhang
- Jiangsu Cancer Hospital, Jiangsu Institute of Cancer Research, The Affiliated Cancer Hospital of Nanjing Medical University, No. 42 Baiziting, Nanjing 210009, China
| |
Collapse
|
26
|
Abbadessa A, Ronca A, Salerno A. Integrating bioprinting, cell therapies and drug delivery towards in vivo regeneration of cartilage, bone and osteochondral tissue. Drug Deliv Transl Res 2024; 14:858-894. [PMID: 37882983 DOI: 10.1007/s13346-023-01437-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
The biological and biomechanical functions of cartilage, bone and osteochondral tissue are naturally orchestrated by a complex crosstalk between zonally dependent cells and extracellular matrix components. In fact, this crosstalk involves biomechanical signals and the release of biochemical cues that direct cell fate and regulate tissue morphogenesis and remodelling in vivo. Three-dimensional bioprinting introduced a paradigm shift in tissue engineering and regenerative medicine, since it allows to mimic native tissue anisotropy introducing compositional and architectural gradients. Moreover, the growing synergy between bioprinting and drug delivery may enable to replicate cell/extracellular matrix reciprocity and dynamics by the careful control of the spatial and temporal patterning of bioactive cues. Although significant advances have been made in this direction, unmet challenges and open research questions persist. These include, among others, the optimization of scaffold zonality and architectural features; the preservation of the bioactivity of loaded active molecules, as well as their spatio-temporal release; the in vitro scaffold maturation prior to implantation; the pros and cons of each animal model and the graft-defect mismatch; and the in vivo non-invasive monitoring of new tissue formation. This work critically reviews these aspects and reveals the state of the art of using three-dimensional bioprinting, and its synergy with drug delivery technologies, to pattern the distribution of cells and/or active molecules in cartilage, bone and osteochondral engineered tissues. Most notably, this work focuses on approaches, technologies and biomaterials that are currently under in vivo investigations, as these give important insights on scaffold performance at the implantation site and its interaction/integration with surrounding tissues.
Collapse
Affiliation(s)
- Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), IDIS Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain.
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125, Naples, Italy.
| | - Aurelio Salerno
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125, Naples, Italy.
| |
Collapse
|
27
|
Xu Y, Saiding Q, Zhou X, Wang J, Cui W, Chen X. Electrospun fiber-based immune engineering in regenerative medicine. SMART MEDICINE 2024; 3:e20230034. [PMID: 39188511 PMCID: PMC11235953 DOI: 10.1002/smmd.20230034] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/26/2024] [Indexed: 08/28/2024]
Abstract
Immune engineering, a burgeoning field within regenerative medicine, involves a spectrum of strategies to optimize the intricate interplay between tissue regenerative biomaterials and the host tissue. These strategies are applied across different types of biomaterials and various disease models, which encompasses finely modulating the immune response at the levels of immune cells and factors, aiming to mitigate adverse effects like fibrosis and persistent inflammation that may arise at the injury site and consequently promote tissue regeneration. With the continuous progress in electrospinning technology, the immunoregulatory capabilities of electrospun fibers have gained substantial attention over the years. Electrospun fibers, with their extracellular matrix-like characteristics, high surface-area-to-volume ratio, and reliable pharmaceutical compound capacity, have emerged as key players among tissue engineering materials. This review specifically focuses on the role of electrospun fiber-based immune engineering, emphasizing their unique design strategies. Notably, electrospinning actively engages in immune engineering by modulating immune responses through four essential strategies: (i) surface modification, (ii) drug loading, (iii) physicochemical parameters, and (iv) biological grafting. This review presents a comprehensive overview of the intricate mechanisms of the immune system in injured tissues while unveiling the key strategies adopted by electrospun fibers to orchestrate immune regulation. Furthermore, the review explores the current developmental trends and limitations concerning the immunoregulatory function of electrospun fibers, aiming to drive the advancements in electrospun fiber-based immune engineering to its full potential.
Collapse
Affiliation(s)
- Yiru Xu
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Qimanguli Saiding
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue Zhou
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xinliang Chen
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| |
Collapse
|
28
|
Huang J, Lu D, Wu C, Pei D, Guo C, Guo H, Yu S, Gao B. Guanidinylated bioactive chitosan-based injectable hydrogels with pro-angiogenic and mechanical properties for accelerated wound closure. Int J Biol Macromol 2024; 258:128943. [PMID: 38143070 DOI: 10.1016/j.ijbiomac.2023.128943] [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: 07/07/2023] [Revised: 12/06/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
Wound healing is a complex process involving the concerted action of many genes and signaling pathways, with angiogenesis being crucial for expediting wound closure. Dressings that possess pro-angiogenic properties are increasingly recognized as attractive candidates for wound care. Drawing inspiration from the active closure of wounds in embryos, we have developed a thermo-responsive hydrogel with mechanoactive properties, combining vascular regeneration and skin wound contraction to accelerate healing. The significant improvement in vascular reconstruction is attributed to the synergistic effect of arginine and deferoxamine (DFO) released from the hydrogels. Additionally, the contraction force of the hydrogel actively promotes skin closure in wounds. Remarkably, groups treated with hydroxybutyl chitosan methacrylate combined with arginine (HBC_m_Arg/DFO) exhibited increased vascularization, and greater wound maturity, leading to enhanced healing. These results highlight the synergistic impact of pro-angiogenic and mechanical properties of the HBC_m_Arg/DFO hydrogel in accelerating wound healing in rats.
Collapse
Affiliation(s)
- Jun Huang
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China
| | - Daohuan Lu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China
| | - Caixia Wu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China
| | - Dating Pei
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China
| | - Cuiping Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China
| | - Huilong Guo
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China
| | - Shan Yu
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China
| | - Botao Gao
- Institute of Biological and Medical Engineering, Guangdong Academy of Sciences, Guangzhou 510632, PR China; National Engineering Research Center for Healthcare Devices, Guangzhou 510632, PR China; Guangdong Key Lab of Medical Electronic Instruments and Polymer Material Products, Guangzhou 510632, PR China.
| |
Collapse
|
29
|
Qin Y, Li S, Liang L, Zhao S, Ye F. Rational synthesis of FeNiCo-LDH nanozyme for colorimetric detection of deferoxamine mesylate. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2023; 303:123156. [PMID: 37506456 DOI: 10.1016/j.saa.2023.123156] [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: 06/02/2023] [Revised: 06/30/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023]
Abstract
The accurate surveillance and sensitive detection of deferoxamine mesylate (DFO) is of great significance to ensure the safety of thalassemia major patients. Herein, we report a new nanozyme-based colorimetric sensor platform for DFO detection. First, a metal-organic framework (ZIF-67) was used as a precursor for the synthesis of FeNiCo-LDH (Layered Double Hydroxide, LDH) via an ion exchange reaction stirring at room temperature. The results of electron microscopy and nitrogen adsorption-desorption showed that FeNiCo-LDH exhibited a 3D hollow and mesopores structure, which supplied more exposed active sites and faster transfer of mass. The as-prepared FeNiCo-LDH showed superior peroxidase-like activity with a low Km and high υmax. It can catalyze the decomposition of H2O2 to generate reactive oxygen species (ROS) and further react with 3,3',5,5'-tetramethylbenzidine (TMB) to form blue oxidized TMB (oxTMB), which has a characteristic absorption at 652 nm. Once DFO was introduced, it can complex with FeNiCo-LDH and inhibit the peroxidase-like activity of FeNiCo-LDH, making the color of oxTMB lighter. The quantitative range of DFO was 0.8-28 μM with a detection limit of 0.71 μM. This established method was applied to the detection of DFO content in urine samples of thalassemia patients, and the spiked recoveries were falling between 97.7% and 109.6%, with a relative standard deviation was less than 5%, providing a promising tool for the clinical medication of thalassemia patients.
Collapse
Affiliation(s)
- Yuan Qin
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Shuishi Li
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Ling Liang
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Shulin Zhao
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China
| | - Fanggui Ye
- State Key Laboratory for the Chemistry and Molecular Engineering of Medicinal Resources, School of Chemistry and Pharmaceutical Science, Guangxi Normal University, Guilin 541004, PR China.
| |
Collapse
|
30
|
Halliwell B, Watt F, Minqin R. Iron and atherosclerosis: Lessons learned from rabbits relevant to human disease. Free Radic Biol Med 2023; 209:165-170. [PMID: 37852545 DOI: 10.1016/j.freeradbiomed.2023.10.383] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 10/02/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
The role of iron in promoting atherosclerosis, and hence the cardiovascular, neurodegenerative and other diseases that result from atherosclerosis, has been fiercely controversial. Many studies have been carried out on various rodent models of atherosclerosis, especially on apoE-knockout (apoE-/-) mice, which develop atherosclerosis more readily than normal mice. These apoE-/- mouse studies generally support a role for iron in atherosclerosis development, although there are conflicting results. The purpose of the current article is to describe studies on another animal model that is not genetically manipulated; New Zealand White (NZW) rabbits fed a high-cholesterol diet. This may be a better model than the apoE-/- mice for human atherosclerosis, although it has been given much less attention. Studies on NZW rabbits support the view that iron promotes atherosclerosis, although some uncertainties remain, which need to be resolved by further experimentation.
Collapse
Affiliation(s)
- Barry Halliwell
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Neurobiology Research Programme, National University of Singapore, Centre for Life Sciences, #05-01A, 28 Medical Drive, 117456, Singapore.
| | - Frank Watt
- Department of Physics, National University of Singapore, Faculty of Science, 2 Science Drive 3, Blk S12, Level 2, 117551, Singapore.
| | - Ren Minqin
- Department of Physics, National University of Singapore, Faculty of Science, 2 Science Drive 3, Blk S12, Level 2, 117551, Singapore.
| |
Collapse
|
31
|
Tsai LT, Weng TI, Chang TY, Lan KC, Chiang CK, Liu SH. Inhibition of Indoxyl Sulfate-Induced Reactive Oxygen Species-Related Ferroptosis Alleviates Renal Cell Injury In Vitro and Chronic Kidney Disease Progression In Vivo. Antioxidants (Basel) 2023; 12:1931. [PMID: 38001784 PMCID: PMC10669521 DOI: 10.3390/antiox12111931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/27/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
The accumulation of the uremic toxin indoxyl sulfate (IS) is a key pathological feature of chronic kidney disease (CKD). The effect of IS on ferroptosis and the role of IS-related ferroptosis in CKD are not well understood. We used a renal tubular cell model and an adenine-induced CKD mouse model to explore whether IS induces ferroptosis and injury and affects iron metabolism in the renal cells and the kidneys. Our results showed that exposure to IS induced several characteristics for ferroptosis, including iron accumulation, an impaired antioxidant system, elevated reactive oxygen species (ROS) levels, and lipid peroxidation. Exposure to IS triggered intracellular iron accumulation by upregulating transferrin and transferrin receptors, which are involved in cellular iron uptake. We also observed increased levels of the iron storage protein ferritin. The effects of IS-induced ROS generation, lipid peroxidation, ferroptosis, senescence, ER stress, and injury/fibrosis were effectively alleviated by treatments with an iron chelator deferoxamine (DFO) in vitro and the adsorbent charcoal AST-120 (scavenging the IS precursor) in vivo. Our findings suggest that IS triggers intracellular iron accumulation and ROS generation, leading to the induction of ferroptosis, senescence, ER stress, and injury/fibrosis in CKD kidneys. AST-120 administration may serve as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Li-Ting Tsai
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
| | - Te-I Weng
- Department of Forensic Medicine, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - Ting-Yu Chang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
| | - Kuo-Cheng Lan
- Department of Emergency Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei 114, Taiwan
| | - Chih-Kang Chiang
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
- Departments of Integrated Diagnostics & Therapeutics and Internal Medicine, College of Medicine and Hospital, National Taiwan University, Taipei 100, Taiwan
| | - Shing-Hwa Liu
- Institute of Toxicology, College of Medicine, National Taiwan University, Taipei 100, Taiwan; (L.-T.T.); (T.-Y.C.); (C.-K.C.)
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung 404, Taiwan
- Department of Pediatrics, College of Medicine, National Taiwan University & Hospital, Taipei 100, Taiwan
| |
Collapse
|
32
|
Saberi A, Kouhjani M, Mohammadi M, Hosta-Rigau L. Novel scaffold platforms for simultaneous induction osteogenesis and angiogenesis in bone tissue engineering: a cutting-edge approach. J Nanobiotechnology 2023; 21:351. [PMID: 37770928 PMCID: PMC10536787 DOI: 10.1186/s12951-023-02115-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 09/15/2023] [Indexed: 09/30/2023] Open
Abstract
Despite the recent advances in the development of bone graft substitutes, treatment of critical size bone defects continues to be a significant challenge, especially in the elderly population. A current approach to overcome this challenge involves the creation of bone-mimicking scaffolds that can simultaneously promote osteogenesis and angiogenesis. In this context, incorporating multiple bioactive agents like growth factors, genes, and small molecules into these scaffolds has emerged as a promising strategy. To incorporate such agents, researchers have developed scaffolds incorporating nanoparticles, including nanoparticulate carriers, inorganic nanoparticles, and exosomes. Current paper provides a summary of the latest advancements in using various bioactive agents, drugs, and cells to synergistically promote osteogenesis and angiogenesis in bone-mimetic scaffolds. It also discusses scaffold design properties aimed at maximizing the synergistic effects of osteogenesis and angiogenesis, various innovative fabrication strategies, and ongoing clinical studies.
Collapse
Affiliation(s)
- Arezoo Saberi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Maryam Kouhjani
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Marzieh Mohammadi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Leticia Hosta-Rigau
- DTU Health Tech, Centre for Nanomedicine and Theranostics, Technical University of Denmark, Produktionstorvet, Building 423, 2800, Kgs. Lyngby, Denmark.
| |
Collapse
|
33
|
Wang J, Wang T, Fang M, Wang Z, Xu W, Teng B, Yuan Q, Hu X. Advances of nanotechnology for intracerebral hemorrhage therapy. Front Bioeng Biotechnol 2023; 11:1265153. [PMID: 37771570 PMCID: PMC10523393 DOI: 10.3389/fbioe.2023.1265153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 09/01/2023] [Indexed: 09/30/2023] Open
Abstract
Intracerebral hemorrhage (ICH), the most devastating subtype of stoke, is of high mortality at 5 years and even those survivors usually would suffer permanent disabilities. Fortunately, various preclinical active drugs have been approached in ICH, meanwhile, the therapeutic effects of these pharmaceutical ingredients could be fully boosted with the assistance of nanotechnology. In this review, besides the pathology of ICH, some ICH therapeutically available active drugs and their employed nanotechnologies, material functions, and therapeutic principles were comprehensively discussed hoping to provide novel and efficient strategies for ICH therapy in the future.
Collapse
Affiliation(s)
- Jiayan Wang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Tianyou Wang
- State Key Laboratory of Polymer Materials Engineering, College of Polymer Science and Engineering, Sichuan University, Chengdu, China
| | - Mei Fang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Zexu Wang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Wei Xu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Bang Teng
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Qijuan Yuan
- School of Materials Science and Engineering, Xihua University, Chengdu, China
| | - Xin Hu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Zhang Y, Yu L, Qiu R, Cao L, Ye G, Lin R, Wang Y, Wang G, Hu B, Hou H. 3D hypoxia-mimicking and anti-synechia hydrogel enabling promoted neovascularization for renal injury repair and regeneration. Mater Today Bio 2023; 21:100694. [PMID: 37346780 PMCID: PMC10279555 DOI: 10.1016/j.mtbio.2023.100694] [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: 02/15/2023] [Revised: 05/02/2023] [Accepted: 06/06/2023] [Indexed: 06/23/2023] Open
Abstract
In-situ renal tissue engineering is promising yet challenging for renal injury repair and regeneration due to the highly vascularized structure of renal tissue and complex high-oxidative stress and ischemic microenvironment. Herein, a novel biocompatible 3D porous hydrogel (DFO-gel) with sustained release capacity of hypoxia mimicking micromolecule drug deferoxamine (DFO) was developed for in-situ renal injury repair. In vitro and in vivo experimental results demonstrated that the developed DFO-gels can exert the synchronous benefit of scavenging excess reactive oxygen species (ROS) regulating inflammatory microenvironment and promoting angiogenesis for effective renal injury repair by up-regulating hypoxia-inducible factor-1 alpha (HIF-1α) and vascular endothelial growth factor (VEGF). The in-situ neogenesis of neonatal glomerular- and tubular-like structures in the implanted areas in the partially nephrectomized rats also suggested the potential for promoting renal injury repair and regeneration. This multifunctional hydrogel can not only exhibit the sustained release and promoted bio-uptake capacity for DFO, but also improve the renal injured microenvironment by alleviating oxidative and inflammatory stress, accelerating neovascularization, and promoting efficient anti-synechia. We believe this work offers a promising strategy for renal injury repair and regeneration.
Collapse
Affiliation(s)
- Yuehang Zhang
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Division of Nephrology, The Second Affiliated Hospital of Kunming Medical University, Kunming Medical University, Kunming, Yunnan, 650500, PR China
| | - Lei Yu
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Renjie Qiu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Lisha Cao
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Genlan Ye
- The Fifth Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Rurong Lin
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Yongqin Wang
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Guobao Wang
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Bianxiang Hu
- Division of Nephrology, State Key Lab for Organ Failure Research, National Clinical Research Center of Kidney Disease, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| | - Honghao Hou
- Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, PR China
| |
Collapse
|
35
|
Zhang T, Zhao J, Guan Y, Li X, Bai J, Song X, Jia Z, Chen S, Li C, Xu Y, Peng J, Wang Y. Deferoxamine promotes peripheral nerve regeneration by enhancing Schwann cell function and promoting axon regeneration of dorsal root ganglion. Neuroscience 2023:S0306-4522(23)00249-X. [PMID: 37286159 DOI: 10.1016/j.neuroscience.2023.05.029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2022] [Revised: 05/13/2023] [Accepted: 05/27/2023] [Indexed: 06/09/2023]
Abstract
Deferoxamine (DFO) is a potent iron chelator for clinical treatment of various diseases. Recent studies have also shown its potential to promote vascular regeneration during peripheral nerve regeneration. However, the effect of DFO on the Schwann cell function and axon regeneration remains unclear. In this study, we investigated the effects of different concentrations of DFO on Schwann cell viability, proliferation, migration, expression of key functional genes, and axon regeneration of dorsal root ganglia (DRG) through a series of in vitro experiments. We found that DFO improves Schwann cell viability, proliferation, and migration in the early stages, with an optimal concentration of 25 μM. DFO also upregulates the expression of myelin-related genes and nerve growth-promoting factors in Schwann cells, while inhibiting the expression of Schwann cell dedifferentiation genes. Moreover, the appropriate concentration of DFO promotes axon regeneration in DRG. Our findings demonstrate that DFO, with suitable concentration and duration of action, can positively affect multiple stages of peripheral nerve regeneration, thereby improving the effectiveness of nerve injury repair. This study also enriches the theory of DFO promoting peripheral nerve regeneration and provides a basis for the design of sustained-release DFO nerve grafts.
Collapse
Affiliation(s)
- Tieyuan Zhang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of Chinese PLA, Beijing, 100853, China
| | - Jinjuan Zhao
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China
| | - Yanjun Guan
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of Chinese PLA, Beijing, 100853, China
| | - Xiangling Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; The School of Medicine, Jinzhou Medical University, Jinzhou, 121099, China
| | - Jun Bai
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of Chinese PLA, Beijing, 100853, China
| | - Xiangyu Song
- Hebei North University, Zhangjiakou, 075000, China
| | - Zhibo Jia
- Hebei North University, Zhangjiakou, 075000, China
| | - Shengfeng Chen
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Guizhou Medical University, Guiyang, 550025, China
| | - Chaochao Li
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Medical School of Chinese PLA, Beijing, 100853, China
| | - Yifan Xu
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226007, China
| | - Yu Wang
- Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma and War Injuries PLA, Department of Orthopedics, the Fourth Medical Center, Chinese PLA General Hospital, Beijing, 100048, China; Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226007, China.
| |
Collapse
|
36
|
Nasir NJM, Heemskerk H, Jenkins J, Hamadee NH, Bunte R, Tucker-Kellogg L. Myoglobin-derived iron causes wound enlargement and impaired regeneration in pressure injuries of muscle. eLife 2023; 12:85633. [PMID: 37267120 DOI: 10.7554/elife.85633] [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: 01/31/2023] [Accepted: 04/25/2023] [Indexed: 06/04/2023] Open
Abstract
The reasons for poor healing of pressure injuries are poorly understood. Vascular ulcers are worsened by extracellular release of hemoglobin, so we examined the impact of myoglobin (Mb) iron in murine muscle pressure injuries (mPI). Tests used Mb-knockout or treatment with deferoxamine iron chelator (DFO). Unlike acute injuries from cardiotoxin, mPI regenerated poorly with a lack of viable immune cells, persistence of dead tissue (necro-slough), and abnormal deposition of iron. However, Mb-knockout or DFO-treated mPI displayed a reversal of the pathology: decreased tissue death, decreased iron deposition, decrease in markers of oxidative damage, and higher numbers of intact immune cells. Subsequently, DFO treatment improved myofiber regeneration and morphology. We conclude that myoglobin iron contributes to tissue death in mPI. Remarkably, a large fraction of muscle death in untreated mPI occurred later than, and was preventable by, DFO treatment, even though treatment started 12 hr after pressure was removed. This demonstrates an opportunity for post-pressure prevention to salvage tissue viability.
Collapse
Affiliation(s)
- Nurul Jannah Mohamed Nasir
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Hans Heemskerk
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- BioSyM and CAMP Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, CREATE, Singapore, Singapore
| | - Julia Jenkins
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | | | - Ralph Bunte
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Lisa Tucker-Kellogg
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Centre for Computational Biology, Duke-NUS Medical School, Singapore, Singapore
- BioSyM and CAMP Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, CREATE, Singapore, Singapore
| |
Collapse
|
37
|
Shams S, Stilhano RS, Silva EA. Harnessing EGLN1 Gene Editing to Amplify HIF-1α and Enhance Human Angiogenic Response. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.29.542734. [PMID: 37398294 PMCID: PMC10312464 DOI: 10.1101/2023.05.29.542734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Therapeutic angiogenesis has been the focus of hundreds of clinical trials but approval for human treatment remains elusive. Current strategies often rely on the upregulation of a single proangiogenic factor, which fails to recapitulate the complex response needed in hypoxic tissues. Hypoxic oxygen tensions dramatically decrease the activity of hypoxia inducible factor prolyl hydroxylase 2 (PHD2), the primary oxygen sensing portion of the hypoxia inducible factor 1 alpha (HIF-1α) proangiogenic master regulatory pathway. Repressing PHD2 activity increases intracellular levels of HIF-1α and impacts the expression of hundreds of downstream genes directly associated with angiogenesis, cell survival, and tissue homeostasis. This study explores activating the HIF-1α pathway through Sp Cas9 knockout of the PHD2 encoding gene EGLN1 as an innovative in situ therapeutic angiogenesis strategy for chronic vascular diseases. Our findings demonstrate that even low editing rates of EGLN1 lead to a strong proangiogenic response regarding proangiogenic gene transcription, protein production, and protein secretion. In addition, we show that secreted factors of EGLN1 edited cell cultures may enhance human endothelial cell neovascularization activity in the context of proliferation and motility. Altogether, this study reveals that EGLN1 gene editing shows promise as a potential therapeutic angiogenesis strategy.
Collapse
|
38
|
Park D, Lee SJ, Choi DK, Park JW. Therapeutic Agent-Loaded Fibrous Scaffolds for Biomedical Applications. Pharmaceutics 2023; 15:pharmaceutics15051522. [PMID: 37242764 DOI: 10.3390/pharmaceutics15051522] [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: 04/05/2023] [Revised: 04/28/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Tissue engineering is a sophisticated field that involves the integration of various disciplines, such as clinical medicine, material science, and life science, to repair or regenerate damaged tissues and organs. To achieve the successful regeneration of damaged or diseased tissues, it is necessary to fabricate biomimetic scaffolds that provide structural support to the surrounding cells and tissues. Fibrous scaffolds loaded with therapeutic agents have shown considerable potential in tissue engineering. In this comprehensive review, we examine various methods for fabricating bioactive molecule-loaded fibrous scaffolds, including preparation methods for fibrous scaffolds and drug-loading techniques. Additionally, we delved into the recent biomedical applications of these scaffolds, such as tissue regeneration, inhibition of tumor recurrence, and immunomodulation. The aim of this review is to discuss the latest research trends in fibrous scaffold manufacturing methods, materials, drug-loading methods with parameter information, and therapeutic applications with the goal of contributing to the development of new technologies or improvements to existing ones.
Collapse
Affiliation(s)
- Dongsik Park
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Su Jin Lee
- Drug Manufacturing Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Dong Kyu Choi
- New Drug Development Center (NDDC), Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| | - Jee-Woong Park
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (K-MEDI Hub), Daegu 41061, Republic of Korea
| |
Collapse
|
39
|
Wang J, Zhang Z, Shi F, Li Y, Tang Y, Liu C, Wang Y, Chen J, Jiang X, Yang H, Sun L, Chen Q, Ao L, Han F, Liu J, Cao J. PM 2.5 caused ferroptosis in spermatocyte via overloading iron and disrupting redox homeostasis. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162089. [PMID: 36781135 DOI: 10.1016/j.scitotenv.2023.162089] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/03/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
Fine particulate matter (PM2.5) has been reported to cause various types of damage to male reproductive system, but the research on the underlying mechanisms is still insufficient. This study attempted to explore the underlying mechanisms of this widely concerning environmental health problem through in vivo and in vitro exposure models. Significant pathological damage and abnormal mitochondria in spermatocytes were observed in the real-time PM2.5 exposure animal model. In addition, significant alterations in key biomarkers of iron metabolism and ferroptosis were found in testis tissues. Notably decreased cell viability was found in vitro. Moreover, the ferroptosis pathway was significantly enriched in the transcriptome enrichment analysis. Subsequent experiments showed that the two core events of ferroptosis, iron overload and lipid peroxidation, occurred in spermatocytes after PM2.5 treatment. Moreover, lipid metabolic genes (Acsl4 and Aloxe3) and the antioxidant gene Gpx4 were found to be key target genes of ferroptosis caused by PM2.5 in spermatocytes. Importantly, further studies showed that the damaging effect could be reversed by the iron chelator deferoxamine mesylate (DFOM) and the lipid peroxidation inhibitor ferrostatin-1 (Fer-1), which further confirmed the role of ferroptosis in PM2.5 toxicity. Our study revealed the vital role of ferroptosis in PM2.5-induced male reproductive damage, providing novel insights into the air pollution-induced decrease in male fertility.
Collapse
Affiliation(s)
- Jiankang Wang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Zhonghao Zhang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Fuquan Shi
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yingqing Li
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Ying Tang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Chang Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Yimeng Wang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jianping Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Xiao Jiang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Huan Yang
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Lei Sun
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Qing Chen
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Lin Ao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Fei Han
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China
| | - Jinyi Liu
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| | - Jia Cao
- Key Lab of Medical Protection for Electromagnetic Radiation, Ministry of Education of China, Institute of Toxicology, College of Preventive Medicine, Army Medical University (Third Military Medical University), Chongqing 400038, China.
| |
Collapse
|
40
|
Zhu Z, Liu X, Li P, Wang H, Zhang Y, Liu M, Ren J. Renal Clearable Quantum Dot-Drug Conjugates Modulate Labile Iron Species and Scavenge Free Radicals for Attenuating Chemotherapeutic Drug-Induced Acute Kidney Injury. ACS APPLIED MATERIALS & INTERFACES 2023; 15:21854-21865. [PMID: 37115671 DOI: 10.1021/acsami.3c00714] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Chemotherapeutic drug-induced acute kidney injury (AKI) involves pathologically increased labile iron species in the kidneys that mediate the excessive generation of reactive oxygen species (ROS) to induce ferroptosis and apoptosis, subsequently driving renal dysfunction. Herein, we report renal clearable quantum dot-drug conjugates (QDCs) composed of carbon quantum dot (CDs), deferoxamine (DFO), and poly(ethylene glycol) (PEG) for attenuating chemotherapeutic drug-induced AKI. The CDs component in QDCs can not only provide DFO with high renal specificity to effectively remove the pathological labile iron species in the kidneys to block the source of ROS generation but also exert high antioxidative effects to avoid renal oxidative damage caused by the ROS that have been overproduced. In cisplatin-induced AKI mice, QDCs can inhibit ferroptosis and apoptosis with high efficacy for AKI treatment. This study will provide a new paradigm to realize enhanced therapeutic efficacy for AKI by simultaneously removing the pathological labile iron species and eliminating overproduced ROS in the kidneys to achieve the goal of addressing both symptoms and root causes.
Collapse
Affiliation(s)
- Zitong Zhu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Xinchen Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, Hospital of Stomatology, Jilin University, Changchun 130021, Jilin, P. R. China
| | - Penghui Li
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Huan Wang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
| | - Yanjie Zhang
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Mengmeng Liu
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| | - Jinsong Ren
- State Key Laboratory of Rare Earth Resources Utilization and Laboratory of Chemical Biology, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei 230026, Anhui, P. R. China
| |
Collapse
|
41
|
Jiang Y, Guo Y, Feng X, Yang P, Liu Y, Dai X, Zhao F, Lei D, Li X, Liu Y, Li Y. Iron metabolism disorder regulated by BMP signaling in hypoxic pulmonary hypertension. Biochim Biophys Acta Mol Basis Dis 2023; 1869:166589. [PMID: 36343841 DOI: 10.1016/j.bbadis.2022.166589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2022] [Revised: 10/22/2022] [Accepted: 10/25/2022] [Indexed: 11/06/2022]
Abstract
BACKGROUNDS AND AIMS Unexplained iron deficiency is associated with poorer survival in patients with pulmonary hypertension (PH). Bone morphogenetic protein (BMP) signaling and BMP protein type II receptor (BMPR2) expression are important in the pathogenesis of PH. BMP6 in hepatocytes is a central transcriptional regulator of the iron hormone hepcidin that controls systemic iron balance. This study aimed to investigate the effects of BMP signaling on iron metabolism and its implication in hypoxia-induced PH. METHODS AND RESULTS PH was induced in Sprague-Dawley Rats under hypoxia for 4 weeks. Compared with the control group, right ventricular systolic pressure and right ventricle hypertrophy index were both markedly increased, and serum iron level was significantly decreased with iron metabolic disorder in the hypoxia group. In cultured human pulmonary artery endothelial cells (HPAECs), hypoxia increased oxidative stress and apoptosis, which were reversed by supplementation with Fe agent. Meanwhile, iron chelator deferoxamine triggered oxidative stress and apoptosis in HPAECs, and treatment with antioxidant alleviated iron-deficiency-induced apoptosis by reducing reactive oxygen species production. Expression of hepcidin, BMP6 and hypoxia-inducible factor (HIF)-1α were significantly upregulated, while expression of BMPR2 was downregulated in hepatocytes in the hypoxia group, both in vivo and in vitro. Expression of hepcidin and HIF-1α were significantly increased by BMP6, while pretreatment with siRNA-BMPR2 augmented the enhanced expression of hepcidin and HIF-1α induced by BMP6. CONCLUSIONS Iron deficiency promoted oxidative stress and apoptosis in HPAECs in hypoxia-induced PH, and enhanced expression of hepcidin regulated by BMP6/BMPR2 signaling may contribute to iron metabolic disorder.
Collapse
Affiliation(s)
- Yujie Jiang
- Department of Health Management, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yingfan Guo
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xuexiang Feng
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Pingting Yang
- Department of Health Management, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yi Liu
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xuejing Dai
- Department of Health Management, The Third Xiangya Hospital of Central South University, Changsha, China; Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Feilong Zhao
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Dongyu Lei
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Xiaohui Li
- Department of Pharmacology, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Yuan Liu
- Department of Anesthesiology, The Second Xiangya Hospital of Central South University, Changsha, China.
| | - Ying Li
- Department of Health Management, The Third Xiangya Hospital of Central South University, Changsha, China.
| |
Collapse
|
42
|
Li J, Chen X, Ren L, Chen X, Wu T, Wang Y, Ren X, Cheng B, Xia J. Type H vessel/platelet-derived growth factor receptor β + perivascular cell disintegration is involved in vascular injury and bone loss in radiation-induced bone damage. Cell Prolif 2023:e13406. [PMID: 36694343 DOI: 10.1111/cpr.13406] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 01/04/2023] [Accepted: 01/10/2023] [Indexed: 01/26/2023] Open
Abstract
Collapse of the microvascular system is a prerequisite for radiation-induced bone loss. Since type H vessels, a specific bone vessel subtype surrounded by platelet-derived growth factor receptor β+ (PDGFRβ+ ) perivascular cells (PVCs), has been recently identified to couple angiogenesis and osteogenesis, we hypothesize that type H vessel injury initiates PDGFRβ+ PVC dysfunction, which contributes to the abnormal angiogenesis and osteogenesis after irradiation. In this study, we found that radiation led to the decrease of both type H endothelial cell (EC) and PDGFRβ+ PVC numbers. Remarkably, results from lineage tracing showed that PDGFRβ+ PVCs detached from microvessels and converted the lineage commitment from osteoblasts to adipocytes, leading to vascular injury and bone loss after irradiation. These phenotype transitions above were further verified to be associated with the decrease in hypoxia-inducible factor-1α (HIF-1α)/PDGF-BB/PDGFRβ signalling between type H ECs and PDGFRβ+ PVCs. Pharmacological blockade of HIF-1α/PDGF-BB/PDGFRβ signalling induced a phenotype similar to radiation-induced bone damage, while the rescue of this signalling significantly alleviated radiation-induced bone injury. Our findings show that the decrease in HIF-1α/PDGF-BB/PDGFRβ signalling between type H ECs and PDGFRβ+ PVCs after irradiation affects the homeostasis of EC-PVC coupling and plays a part in vascular damage and bone loss, which has broad implications for effective translational therapies.
Collapse
Affiliation(s)
- Jiayan Li
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xiaodan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lin Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xijuan Chen
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Tong Wu
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yun Wang
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xianyue Ren
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China.,Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
43
|
Gao S, Zhang W, Zhai X, Zhao X, Wang J, Weng J, Li J, Chen X. An antibacterial and proangiogenic double-layer drug-loaded microneedle patch for accelerating diabetic wound healing. Biomater Sci 2023; 11:533-541. [PMID: 36472206 DOI: 10.1039/d2bm01588a] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Diabetic wounds are difficult to heal because of bacterial infections and insufficient angiogenesis. Herein, we report a double-layer drug-loaded microneedle patch with antibacterial and angiogenesis-promoting properties for diabetic wound healing. The double-layer microneedle comprises the hyaluronic acid (HA)-loaded antibacterial drug tetracycline hydrochloride (TCH) as the tip and a mixture of chitosan and silk fibroin containing the angiogenic drug deferoxamine (DFO) as the substrate. In the double-layer drug-loaded microneedle system (DMN@TCH/DFO), rapid dissolution of HA at the tip releases TCH to promote early antibacterial activity. The substrate exhibits excellent swelling properties, facilitating the absorption of tissue fluid from the wound to promote wound contraction. Simultaneously, DFO is released to promote angiogenesis. Therefore, DMN@TCH/DFO exhibited adequate mechanical properties, excellent swelling and biocompatibility, antibacterial properties, and angiogenesis-promoting capabilities. In a wound model of diabetic rats, DMN@TCH/DFO reduced inflammatory responses, promoted angiogenesis, and facilitated collagen deposition, thereby accelerating diabetic wound healing. Overall, DMN@TCH/DFO can accelerate the healing of diabetic wounds and has clinical application prospects.
Collapse
Affiliation(s)
- Shan Gao
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Wanlin Zhang
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Xingxing Zhai
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Xue Zhao
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| | - Jianxin Wang
- Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jie Weng
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China. .,Key Laboratory of Advanced Technologies of Materials (Ministry of Education), School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, China
| | - Jianshu Li
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Xingyu Chen
- College of Medicine, Southwest Jiaotong University, Chengdu 610031, China.
| |
Collapse
|
44
|
Parker JB, Griffin MF, Downer MA, Akras D, Berry CE, Cotterell AC, Gurtner GC, Longaker MT, Wan DC. Chelating the valley of death: Deferoxamine's path from bench to wound clinic. Front Med (Lausanne) 2023; 10:1015711. [PMID: 36873870 PMCID: PMC9975168 DOI: 10.3389/fmed.2023.1015711] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 01/18/2023] [Indexed: 02/17/2023] Open
Abstract
There is undisputable benefit in translating basic science research concretely into clinical practice, and yet, the vast majority of therapies and treatments fail to achieve approval. The rift between basic research and approved treatment continues to grow, and in cases where a drug is granted approval, the average time from initiation of human trials to regulatory marketing authorization spans almost a decade. Albeit with these hurdles, recent research with deferoxamine (DFO) bodes significant promise as a potential treatment for chronic, radiation-induced soft tissue injury. DFO was originally approved by the Food and Drug Administration (FDA) in 1968 for the treatment of iron overload. However, investigators more recently have posited that its angiogenic and antioxidant properties could be beneficial in treating the hypovascular and reactive-oxygen species-rich tissues seen in chronic wounds and radiation-induced fibrosis (RIF). Small animal experiments of various chronic wound and RIF models confirmed that treatment with DFO improved blood flow and collagen ultrastructure. With a well-established safety profile, and now a strong foundation of basic scientific research that supports its potential use in chronic wounds and RIF, we believe that the next steps required for DFO to achieve FDA marketing approval will include large animal studies and, if those prove successful, human clinical trials. Though these milestones remain, the extensive research thus far leaves hope for DFO to bridge the gap between bench and wound clinic in the near future.
Collapse
Affiliation(s)
- Jennifer B Parker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Michelle F Griffin
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Mauricio A Downer
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Deena Akras
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Charlotte E Berry
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Asha C Cotterell
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Geoffrey C Gurtner
- Department of Surgery, University of Arizona College of Medicine, Tucson, AZ, United States
| | - Michael T Longaker
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States.,Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, United States
| | - Derrick C Wan
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
45
|
Liu K, Li L, Chen J, Li Y, Wen W, Lu L, Li L, Li H, Liu M, Zhou C, Luo B. Bone ECM-like 3D Printing Scaffold with Liquid Crystalline and Viscoelastic Microenvironment for Bone Regeneration. ACS NANO 2022; 16:21020-21035. [PMID: 36469414 DOI: 10.1021/acsnano.2c08699] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Implanting a 3D printing scaffold is an effective therapeutic strategy for personalized bone repair. As the key factor for the success of bone tissue engineering, the scaffold should provide an appropriate bone regeneration microenvironment and excellent mechanical properties. In fact, the most ideal osteogenic microenvironment is undoubtedly provided by natural bone extracellular matrix (ECM), which exhibits liquid crystalline and viscoelastic characteristics. However, mimicking a bone ECM-like microenvironment in a 3D structure with outstanding mechanical properties is a huge challenge. Herein, we develop a facile approach to fabricate a bionic scaffold perfectly combining bone ECM-like microenvironment and robust mechanical properties. Creatively, 3D printing a poly(l-lactide) (PLLA) scaffold was effectively strengthened via layer-by-layer electrostatic self-assembly of chitin whiskers. More importantly, a kind of chitin whisker/chitosan composite hydrogel with bone ECM-like liquid crystalline state and viscoelasticity was infused into the robust PLLA scaffold to build the bone ECM-like microenvironment in 3D structure, thus highly promoting bone regeneration. Moreover, deferoxamine, an angiogenic factor, was encapsulated in the composite hydrogel and sustainably released, playing a long-term role in angiogenesis and thereby further promoting osteogenesis. This scaffold with bone ECM-like microenvironment and excellent mechanical properties can be considered as an effective implantation for bone repair.
Collapse
Affiliation(s)
- Kun Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
| | - Lin Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
| | - Jingsheng Chen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
| | - Yizhi Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
| | - Wei Wen
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou510632, PR China
| | - Lu Lu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou510632, PR China
| | - Lihua Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou510632, PR China
| | - Hong Li
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou510632, PR China
| | - Mingxian Liu
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou510632, PR China
| | - Changren Zhou
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou510632, PR China
| | - Binghong Luo
- Biomaterial Research Laboratory, Department of Material Science and Engineering, College of Chemistry and Materials, Jinan University, Guangzhou510632, PR China
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Guangzhou510632, PR China
| |
Collapse
|
46
|
Exploring Micromonospora as Phocoenamicins Producers. Mar Drugs 2022; 20:md20120769. [PMID: 36547916 PMCID: PMC9782249 DOI: 10.3390/md20120769] [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: 10/21/2022] [Revised: 12/01/2022] [Accepted: 12/03/2022] [Indexed: 12/13/2022] Open
Abstract
Over the past few years, new technological and scientific advances have reinforced the field of natural product discovery. The spirotetronate class of natural products has recently grown with the discovery of phocoenamicins, natural actinomycete derived compounds that possess different antibiotic activities. Exploring the MEDINA's strain collection, 27 actinomycete strains, including three marine-derived and 24 terrestrial strains, were identified as possible phocoenamicins producers and their taxonomic identification by 16S rDNA sequencing showed that they all belong to the Micromonospora genus. Using an OSMAC approach, all the strains were cultivated in 10 different media each, resulting in 270 fermentations, whose extracts were analyzed by LC-HRMS and subjected to High-throughput screening (HTS) against methicillin-resistant Staphylococcus aureus (MRSA), Mycobacterium tuberculosis H37Ra and Mycobacterium bovis. The combination of LC-UV-HRMS analyses, metabolomics analysis and molecular networking (GNPS) revealed that they produce several related spirotetronates not disclosed before. Variations in the culture media were identified as the most determining factor for phocoenamicin production and the best producer strains and media were established. Herein, we reported the chemically diverse production and metabolic profiling of Micromonospora sp. strains, including the known phocoenamicins and maklamicin, reported for the first time as being related to this family of compounds, as well as the bioactivity of their crude extracts. Although our findings do not confirm previous statements about phocoenamicins production only in unique marine environments, they have identified marine-derived Micromonospora species as the best producers of phocoenamicins in terms of both the abundance in their extracts of some major members of the structural class and the variety of molecular structures produced.
Collapse
|
47
|
Li Z, Cheng S, Li A, Song C, Jiang A, Xu F, Chi H, Yan J, Chen G. Fabrication of BMP-2-peptide-Deferoxamine- and QK-peptide-functionalized nanoscaffolds and their application for bone defect treatment. J Tissue Eng Regen Med 2022; 16:1223-1237. [PMID: 36349393 DOI: 10.1002/term.3364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 10/07/2022] [Accepted: 10/23/2022] [Indexed: 11/11/2022]
Abstract
The microenvironment in the healing process of large bone defects requires suitable conditions to promote osteogenesis and angiogenesis. Coaxial electrospinning is a mature method in bone tissue engineering (BTE) and allows functional modification. Appropriate modification methods can be used to improve the bioactivity of scaffolds for BTE. In this study, coaxial electrospinning with QK peptide (a Vascular endothelial growth factor mimetic peptide) and BMP-2 peptide-DFO (BD) was performed to produce double-modified PQBD scaffolds with vascularizing and osteogenic features. The morphology of coaxially electrospun scaffolds was verified by scanning electron microscopy (SEM) and transmission electron microscopy. Laser scanning confocal microscopy and Fourier transform infrared spectroscopy confirmed that BD covalently bound to the surface of the P and PQ scaffolds. In vitro, the PQBD scaffold promoted the adhesion and proliferation of bone marrow stromal cells (BMSCs). Both QK peptide and BD showed sustainable release and preservation of biological activity, enhancing the osteogenic differentiation of BMSCs and the migration of human umbilical vein endothelial cells and promoting angiogenesis. The combined ability of these factors to promote osteogenesis and angiogenesis is superior to that of each alone. In vivo, the PQBD scaffold was implanted into the bone defect, and after 8 weeks, the defect area was almost completely covered by new bone tissue. Histology showed more mature bone tissue and more blood vessels. PQBD scaffolds promote both angiogenesis and osteogenesis, offering a promising approach to enhance bone regeneration in the treatment of large bone defects.
Collapse
Affiliation(s)
- Zecheng Li
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Shi Cheng
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Ang Li
- The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Chengchao Song
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Anlong Jiang
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Fangxing Xu
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Hui Chi
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| | - Jinglong Yan
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Guanghua Chen
- Department of Orthopedics, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.,The Key Laboratory of Myocardial Ischemia, Harbin Medical University, Ministry of Education, Harbin, Heilongjiang, China
| |
Collapse
|
48
|
Han L, Dong X, Qiu T, Dou Z, Wu L, Dai H. Enhanced sciatic nerve regeneration by relieving iron-overloading and organelle stress with the nanofibrous P(MMD-co-LA)/DFO conduits. Mater Today Bio 2022; 16:100387. [PMID: 36042854 PMCID: PMC9420382 DOI: 10.1016/j.mtbio.2022.100387] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/24/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
Wallerian degeneration after peripheral nerve injury (PNI), that is, the autonomous degeneration of distal axons, leads to an imbalance of iron homeostasis and easily induces oxidative stress caused by iron overload. Inspired by the process of nerve degeneration and regeneration, the design of a functional electrospinning scaffold with iron chelating ability exhibited the importance of reconstructing a suitable microenvironment. Here, an electrospinning scaffold based on deferoxamine and poly(3(S)-methyl-morpholine-2,5-dione-co-lactone) (PDPLA/DFO) was constructed. This work aims to explore the promotion of nerve regeneration by the physiological regulation of the scaffold. In vitro, PDPLA/DFO films mitigated the reduction of glutathione and the inactivation of Glutathione peroxidase 4 caused by iron overload. In addition, they decreased reactive oxygen species, relieve the stress of the endoplasmic reticulum and mitochondria, and reduce cell apoptosis. In vivo, PDPLA/DFO conduits constructed the anti-inflammatory microenvironment and promoted cell survival by alleviating iron overload and organelle stress. In conclusion, PDPLA/DFO guidance conduits targeted the distal iron overload and promoted nerve regeneration. It provides novel ideas for designing nerve conduits targeting the distal microenvironment.
Collapse
Affiliation(s)
- Lei Han
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Xianzhen Dong
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Tong Qiu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Zhaona Dou
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Lin Wu
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
| | - Honglian Dai
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Biomedical Materials and Engineering Research Center of Hubei Province, Wuhan University of Technology, Wuhan, 430070, China
- Foshan Xianhu Laboratory of the Advanced Energy Science and Technology Guangdong Laboratory, Xianhu Hydrogen Valley, Foshan, 528200, China
| |
Collapse
|
49
|
Zhao Y, Chen H, Ran K, Zhang Y, Pan H, Shangguan J, Tong M, Yang J, Yao Q, Xu H. Porous hydroxyapatite scaffold orchestrated with bioactive coatings for rapid bone repair. BIOMATERIALS ADVANCES 2022; 144:213202. [PMID: 36434928 DOI: 10.1016/j.bioadv.2022.213202] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/09/2022] [Accepted: 11/14/2022] [Indexed: 11/18/2022]
Abstract
Current bioceramic scaffolds for critical-size bone defects are still facing various challenges such as the poor capability of self-resorption, vascularization and osteogenesis. Herein, a composite scaffold (HOD) is fabricated by integrating bioactive coatings of konjac glucomannan (KGM) and deferoxamine (DFO) into porous hydroxyapatite scaffold (HA), where KGM coating induces the self-resorption of HOD after implanting and DFO promoted the vascularization at the defected bone. Porous HA scaffolds with 200-400 μm of pore sizes were prepared and these bioactive coatings were successfully deposited on the scaffold, which was confirmed by SEM. MC3T3-E1 cells could be tightly attached to the pore wall of HOD and the obvious osteogenic differentiation was clearly displayed after 14 days of co-culture. Besides, HOD displayed the potential of promoting the vascularization of HUVECs. Importantly, the accelerated degradation of HOD was observed in a macrophage-associated acidic medium, which led to the self-resorption of HOD in vivo. Micro-CT images showed that HOD was gradually replaced by newly formed bone, achieving a balance between the new bone formation and the scaffold degradation. The rapid bone repairing of the femoral defects in rats was displayed for HOD in comparison to the HA scaffold. Moreover, the therapeutic mechanism of HOD was highly associated with promoted osteogenesis and vascularization. Collectively, the porous ceramic scaffold orchestrated with bioactive coatings may be a promising strategy for repairing of the large bone defect.
Collapse
Affiliation(s)
- Yingzheng Zhao
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| | - Hangbo Chen
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Kunjie Ran
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Yingying Zhang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Hanxiao Pan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Jianxun Shangguan
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Mengqi Tong
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Jiaojiao Yang
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Qing Yao
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China
| | - Helin Xu
- Department of pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| |
Collapse
|
50
|
Zeng Y, Huang C, Duan D, Lou A, Guo Y, Xiao T, Wei J, Liu S, Wang Z, Yang Q, Zhou L, Wu Z, Wang L. Injectable temperature-sensitive hydrogel system incorporating deferoxamine-loaded microspheres promotes H-type blood vessel-related bone repair of a critical size femoral defect. Acta Biomater 2022; 153:108-123. [PMID: 36115651 DOI: 10.1016/j.actbio.2022.09.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/01/2022]
Abstract
Insufficient vascularization is a major challenge in the repair of critical-sized bone defects. Deferoxamine (DFO) has been reported to play a potential role in promoting the formation of H-type blood vessels, a specialized vascular subtype with coupled angiogenesis and osteogenesis. However, whether DFO promotes the expression of H-type vessels in critical femoral defects with complete periosteal damage remains unknown. Moreover, stable drug loading systems need to be designed owing to the short half-life and high-dose toxic effects of DFO. In this study, we developed an injectable DFO-gelatin microspheres (GMs) hydrogel complex as a stable drug loading system for the treatment of critical femoral defects in rats. Our results showed that sustained release of DFO in critical femoral defects stimulated the generation of functional H-type vessels. The DFO-GMs hydrogel complex effectively promoted proliferation, formation, and migration of human umbilical vein endothelial cells in vitro. In vivo, the application of the DFO-GMs hydrogel complex expanded the distribution range and prolonged the expression time of H-type vessels in the defect area and was positively correlated with the number of osterix+ cells and new bone tissue. Topical application of the HIF-1α inhibitor PX-478 partially blocked the stimulation of H-type vessels by DFO, whereas the osteogenic potential of the latter was also weakened. Our results extended the local application of DFO and provided a theoretical basis for targeting H-type vessels to treat large femoral defects. STATEMENT OF SIGNIFICANCE: Abundant functional blood vessels are essential for bone repair. The H-type blood vessel is a functional subtype with angiogenesis and osteogenesis coupling potential. A drug loading system with long-term controlled release was first used to investigate the formation of H-type blood vessels in critical femoral defects and promotion of bone repair. Our results showed that the application of DFO-GMs hydrogel complex expanded the distribution range and expression time of H-type vessels, and was positively correlated with the number of osteoblasts and volume of new bone tissue. These results expanded the local application approach of DFO and provide a theoretical basis for targeting H-type vessels to treat large femoral defects.
Collapse
Affiliation(s)
- Yuwei Zeng
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Chuang Huang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Dongming Duan
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Aiju Lou
- Department of Rheumatology, Liwan Central Hospital of Guangzhou, 35 Liwan Road, Guangzhou 510030, China
| | - Yuan Guo
- Department of Stomatology, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Tianhua Xiao
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Jianguo Wei
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Song Liu
- Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Zhao Wang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Qihao Yang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China
| | - Lei Zhou
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| | - Zenghui Wu
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| | - Le Wang
- Guangzhou Key Laboratory of Spine Disease Prevention and Treatment, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China; Department of Orthopaedic Surgery, The Third Affiliated Hospital, Guangzhou Medical University, 63 Duobao Road, Guangzhou 510150, China.
| |
Collapse
|