1
|
Hao P, Yang Z, So KF, Li X. A core scientific problem in the treatment of central nervous system diseases: newborn neurons. Neural Regen Res 2024; 19:2588-2601. [PMID: 38595278 PMCID: PMC11168522 DOI: 10.4103/nrr.nrr-d-23-01775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 01/06/2024] [Accepted: 02/22/2024] [Indexed: 04/11/2024] Open
Abstract
It has long been asserted that failure to recover from central nervous system diseases is due to the system's intricate structure and the regenerative incapacity of adult neurons. Yet over recent decades, numerous studies have established that endogenous neurogenesis occurs in the adult central nervous system, including humans'. This has challenged the long-held scientific consensus that the number of adult neurons remains constant, and that new central nervous system neurons cannot be created or renewed. Herein, we present a comprehensive overview of the alterations and regulatory mechanisms of endogenous neurogenesis following central nervous system injury, and describe novel treatment strategies that target endogenous neurogenesis and newborn neurons in the treatment of central nervous system injury. Central nervous system injury frequently results in alterations of endogenous neurogenesis, encompassing the activation, proliferation, ectopic migration, differentiation, and functional integration of endogenous neural stem cells. Because of the unfavorable local microenvironment, most activated neural stem cells differentiate into glial cells rather than neurons. Consequently, the injury-induced endogenous neurogenesis response is inadequate for repairing impaired neural function. Scientists have attempted to enhance endogenous neurogenesis using various strategies, including using neurotrophic factors, bioactive materials, and cell reprogramming techniques. Used alone or in combination, these therapeutic strategies can promote targeted migration of neural stem cells to an injured area, ensure their survival and differentiation into mature functional neurons, and facilitate their integration into the neural circuit. Thus can integration replenish lost neurons after central nervous system injury, by improving the local microenvironment. By regulating each phase of endogenous neurogenesis, endogenous neural stem cells can be harnessed to promote effective regeneration of newborn neurons. This offers a novel approach for treating central nervous system injury.
Collapse
Affiliation(s)
- Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Kwok-Fai So
- Guangdong-HongKong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong Province, China
- Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administration Region, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, Guangdong Province, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
2
|
Rust R, Nih LR, Liberale L, Yin H, El Amki M, Ong LK, Zlokovic BV. Brain repair mechanisms after cell therapy for stroke. Brain 2024; 147:3286-3305. [PMID: 38916992 PMCID: PMC11449145 DOI: 10.1093/brain/awae204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 06/04/2024] [Accepted: 06/08/2024] [Indexed: 06/27/2024] Open
Abstract
Cell-based therapies hold great promise for brain repair after stroke. While accumulating evidence confirms the preclinical and clinical benefits of cell therapies, the underlying mechanisms by which they promote brain repair remain unclear. Here, we briefly review endogenous mechanisms of brain repair after ischaemic stroke and then focus on how different stem and progenitor cell sources can promote brain repair. Specifically, we examine how transplanted cell grafts contribute to improved functional recovery either through direct cell replacement or by stimulating endogenous repair pathways. Additionally, we discuss recently implemented preclinical refinement methods, such as preconditioning, microcarriers, genetic safety switches and universal (immune evasive) cell transplants, as well as the therapeutic potential of these pharmacologic and genetic manipulations to further enhance the efficacy and safety of cell therapies. By gaining a deeper understanding of post-ischaemic repair mechanisms, prospective clinical trials may be further refined to advance post-stroke cell therapy to the clinic.
Collapse
Affiliation(s)
- Ruslan Rust
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Institute for Regenerative Medicine, University of Zurich, 8952 Schlieren, Switzerland
| | - Lina R Nih
- Department of Brain Health, University of Nevada, Las Vegas, NV 89154, USA
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Hao Yin
- Robarts Research Institute, Schulich School of Medicine and Dentistry, Western University, London, ON N6A 3K7, Canada
| | - Mohamad El Amki
- Department of Neurology, University Hospital and University of Zurich, 8091 Zurich, Switzerland
| | - Lin Kooi Ong
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, QLD 4350, Australia
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, CA 90033, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| |
Collapse
|
3
|
Zhu S, Liu X, Lu X, Liao Q, Luo H, Tian Y, Cheng X, Jiang Y, Liu G, Chen J. Biomaterials and tissue engineering in traumatic brain injury: novel perspectives on promoting neural regeneration. Neural Regen Res 2024; 19:2157-2174. [PMID: 38488550 PMCID: PMC11034597 DOI: 10.4103/1673-5374.391179] [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: 08/10/2023] [Revised: 10/13/2023] [Accepted: 11/20/2023] [Indexed: 04/24/2024] Open
Abstract
Traumatic brain injury is a serious medical condition that can be attributed to falls, motor vehicle accidents, sports injuries and acts of violence, causing a series of neural injuries and neuropsychiatric symptoms. However, limited accessibility to the injury sites, complicated histological and anatomical structure, intricate cellular and extracellular milieu, lack of regenerative capacity in the native cells, vast variety of damage routes, and the insufficient time available for treatment have restricted the widespread application of several therapeutic methods in cases of central nervous system injury. Tissue engineering and regenerative medicine have emerged as innovative approaches in the field of nerve regeneration. By combining biomaterials, stem cells, and growth factors, these approaches have provided a platform for developing effective treatments for neural injuries, which can offer the potential to restore neural function, improve patient outcomes, and reduce the need for drugs and invasive surgical procedures. Biomaterials have shown advantages in promoting neural development, inhibiting glial scar formation, and providing a suitable biomimetic neural microenvironment, which makes their application promising in the field of neural regeneration. For instance, bioactive scaffolds loaded with stem cells can provide a biocompatible and biodegradable milieu. Furthermore, stem cells-derived exosomes combine the advantages of stem cells, avoid the risk of immune rejection, cooperate with biomaterials to enhance their biological functions, and exert stable functions, thereby inducing angiogenesis and neural regeneration in patients with traumatic brain injury and promoting the recovery of brain function. Unfortunately, biomaterials have shown positive effects in the laboratory, but when similar materials are used in clinical studies of human central nervous system regeneration, their efficacy is unsatisfactory. Here, we review the characteristics and properties of various bioactive materials, followed by the introduction of applications based on biochemistry and cell molecules, and discuss the emerging role of biomaterials in promoting neural regeneration. Further, we summarize the adaptive biomaterials infused with exosomes produced from stem cells and stem cells themselves for the treatment of traumatic brain injury. Finally, we present the main limitations of biomaterials for the treatment of traumatic brain injury and offer insights into their future potential.
Collapse
Affiliation(s)
- Shihong Zhu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiaoyin Liu
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, Chengdu, Sichuan Province, China
| | - Xiyue Lu
- Department of Anesthesiology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Qiang Liao
- Department of Pharmacy, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Huiyang Luo
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
- Department of Anesthesiology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Yuan Tian
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Xu Cheng
- Department of Anesthesiology, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Yaxin Jiang
- Out-patient Department, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Guangdi Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| | - Jing Chen
- Department of Neurosurgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan Province, China
| |
Collapse
|
4
|
Wu N, Li W, Chen Q, Chen M, Chen S, Cheng C, Xie Y. Research Advances in Neuroblast Migration in Traumatic Brain Injury. Mol Neurobiol 2024; 61:1-13. [PMID: 38507029 DOI: 10.1007/s12035-024-04117-4] [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: 09/10/2023] [Accepted: 02/17/2024] [Indexed: 03/22/2024]
Abstract
Neuroblasts were first derived from the adult mammalian brains in the 1990s by Reynolds et al. Since then, persistent neurogenesis in the subgranular zone (SGZ) of the hippocampus and subventricular zone (SVZ) has gradually been recognized. To date, reviews on neuroblast migration have largely investigated glial cells and molecular signaling mechanisms, while the relationship between vasculature and cell migration remains a mystery. Thus, this paper underlines the partial biological features of neuroblast migration and unravels the significance and mechanisms of the vasculature in the process to further clarify theoretically the neural repair mechanism after brain injury. Neuroblast migration presents three modes according to the characteristics of cells that act as scaffolds during the migration process: gliophilic migration, neurophilic migration, and vasophilic migration. Many signaling molecules, including brain-derived neurotrophic factor (BDNF), stromal cell-derived factor 1 (SDF-1), vascular endothelial growth factor (VEGF), and angiopoietin-1 (Ang-1), affect vasophilic migration, synergistically regulating the migration of neuroblasts to target areas along blood vessels. However, the precise role of blood vessels in the migration of neuroblasts needs to be further explored. The in-depth study of neuroblast migration will most probably provide theoretical basis and breakthrough for the clinical treatment of brain injury diseases.
Collapse
Affiliation(s)
- Na Wu
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Wenlang Li
- Department of Anesthesiology, The Second Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Qiang Chen
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Meng Chen
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Siyuan Chen
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China
| | - Chongjie Cheng
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Yuzhong District, Chongqing, China
| | - Yimin Xie
- Department of Pediatric Surgery, Chongqing University Three Gorges Hospital, Wanzhou District, No. 165 Xincheng Road, Wanzhou District, Chongqing, 404100, China.
| |
Collapse
|
5
|
Liu HC, Huang CH, Chiang MR, Hsu RS, Chou TC, Lu TT, Lee IC, Liao LD, Chiou SH, Lin ZH, Hu SH. Sustained Release of Nitric Oxide-Mediated Angiogenesis and Nerve Repair by Mussel-Inspired Adaptable Microreservoirs for Brain Traumatic Injury Therapy. Adv Healthc Mater 2024; 13:e2302315. [PMID: 37713592 DOI: 10.1002/adhm.202302315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Traumatic brain injury (TBI) triggers inflammatory response and glial scarring, thus substantially hindering brain tissue repair. This process is exacerbated by the accumulation of activated immunocytes at the injury site, which contributes to scar formation and impedes tissue repair. In this study, a mussel-inspired nitric oxide-release microreservoir (MINOR) that combines the features of reactive oxygen species (ROS) scavengers and sustained NO release to promote angiogenesis and neurogenesis is developed for TBI therapy. The injectable MINOR fabricated using a microfluidic device exhibits excellent monodispersity and gel-like self-healing properties, thus allowing the maintenance of its structural integrity and functionality upon injection. Furthermore, polydopamine in the MINOR enhances cell adhesion, significantly reduces ROS levels, and suppresses inflammation. Moreover, a nitric oxide (NO) donor embedded into the MINOR enables the sustained release of NO, thus facilitating angiogenesis and mitigating inflammatory responses. By harnessing these synergistic effects, the biocompatible MINOR demonstrates remarkable efficacy in enhancing recovery in mice. These findings benefit future therapeutic interventions for patients with TBI.
Collapse
Affiliation(s)
- Hsiu-Ching Liu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Chu-Han Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Min-Ren Chiang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Ru-Siou Hsu
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Tsu-Chin Chou
- Institute of Analytical and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Tsai-Te Lu
- Institute of Biomedical Engineering, National Tsing Hua University, 300044, Hsinchu, Taiwan
- Department of Chemistry, Chung Yuan Christian University, Taoyuan, 320314, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, 300044, Taiwan
| | - I-Chi Lee
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| | - Lun-De Liao
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, 35053, Miaoli County, Taiwan
| | - Shih-Hwa Chiou
- Department of Medical Research, National Yang Ming Chiao Tung University, Taipei Veterans General Hospital, 112304, Taipei, Taiwan
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei, 112304, Taiwan
| | - Zhong-Hong Lin
- Department of Biomedical Engineering, National Taiwan University, 10617, Taipei, Taiwan
| | - Shang-Hsiu Hu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, 300044, Hsinchu, Taiwan
| |
Collapse
|
6
|
Bolan F, Dickie BR, Cook JR, Thomas JM, Pinteaux E, Allan SM, Saiani A, Lawrence CB. Intracerebral Administration of a Novel Self-Assembling Peptide Hydrogel Is Safe and Supports Cell Proliferation in Experimental Intracerebral Haemorrhage. Transl Stroke Res 2024; 15:986-1004. [PMID: 37853252 PMCID: PMC11364698 DOI: 10.1007/s12975-023-01189-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/08/2023] [Accepted: 08/15/2023] [Indexed: 10/20/2023]
Abstract
Intracerebral haemorrhage (ICH) is the deadliest form of stroke, but current treatment options are limited, meaning ICH survivors are often left with life-changing disabilities. The significant unmet clinical need and socioeconomic burden of ICH mean novel regenerative medicine approaches are gaining interest. To facilitate the regeneration of the ICH lesion, injectable biomimetic hydrogels are proposed as both scaffolds for endogenous repair and delivery platforms for pro-regenerative therapies. In this paper, the objective was to explore whether injection of a novel self-assembling peptide hydrogel (SAPH) Alpha2 was feasible, safe and could stimulate brain tissue regeneration, in a collagenase-induced ICH model in rats. Alpha2 was administered intracerebrally at 7 days post ICH and functional outcome measures, histological markers of damage and repair and RNA-sequencing were investigated for up to 8 weeks. The hydrogel Alpha2 was safe, well-tolerated and was retained in the lesion for several weeks, where it allowed infiltration of host cells. The hydrogel had a largely neutral effect on functional outcomes and expression of angiogenic and neurogenic markers but led to increased numbers of proliferating cells. RNAseq and pathway analysis showed that ICH altered genes related to inflammatory and phagocytic pathways, and these changes were also observed after administration of hydrogel. Overall, the results show that the novel hydrogel was safe when injected intracerebrally and had no negative effects on functional outcomes but increased cell proliferation. To elicit a regenerative effect, future studies could use a functionalised hydrogel or combine it with an adjunct therapy.
Collapse
Affiliation(s)
- Faye Bolan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, M13 9PT, UK
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Ben R Dickie
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, M13 9PT, UK
- Division of Informatics, Imaging and Data Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - James R Cook
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, M13 9PT, UK
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Josephine M Thomas
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, M13 9PT, UK
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Emmanuel Pinteaux
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, M13 9PT, UK
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Stuart M Allan
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, M13 9PT, UK
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Alberto Saiani
- Department of Materials, The University of Manchester, Manchester, M13 9PL, UK
- Manchester Institute of Biotechnology, The University of Manchester, Manchester, M13 9PL, UK
| | - Catherine B Lawrence
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance NHS Foundation Trust, The University of Manchester, Manchester, M13 9PT, UK.
- Division of Neuroscience, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
7
|
Wilson KL, Joseph NI, Onweller LA, Anderson AR, Darling NJ, David-Bercholz J, Segura T. SDF-1 Bound Heparin Nanoparticles Recruit Progenitor Cells for Their Differentiation and Promotion of Angiogenesis after Stroke. Adv Healthc Mater 2024; 13:e2302081. [PMID: 38009291 PMCID: PMC11128481 DOI: 10.1002/adhm.202302081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 11/22/2023] [Indexed: 11/28/2023]
Abstract
Angiogenesis after stroke is correlated with enhanced tissue repair and functional outcomes. The existing body of research in biomaterials for stroke focuses on hydrogels for the delivery of stem cells, growth factors, or small molecules or drugs. Despite the ability of hydrogels to enhance all these delivery methods, no material has significantly regrown vasculature within the translatable timeline of days to weeks after stroke. Here, two novel biomaterial formulations of granular hydrogels are developed for tissue regeneration after stroke: highly porous microgels (i.e., Cryo microgels) and microgels bound with heparin-norbornene nanoparticles with covalently bound SDF-1α. The combination of these materials results in perfused vessels throughout the stroke core in only 10 days, in addition to increased neural progenitor cell recruitment, maintenance, and increased neuronal differentiation.
Collapse
Affiliation(s)
- Katrina L. Wilson
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Neica I. Joseph
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Lauren A. Onweller
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Alexa R. Anderson
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
| | - Nicole J. Darling
- Department of Chemical and Biomolecular Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | | | - Tatiana Segura
- Department of Biomedical Engineering, Duke University, Durham NC 27708-0281, USA
- Department of Neurology, Duke University, Durham, NC, 27708-0281 USA
- Department of Dermatology, Duke University, Durham, NC, 27708-0281 USA
| |
Collapse
|
8
|
Tigner TJ, Dampf G, Tucker A, Huang YC, Jagrit V, Clevenger AJ, Mohapatra A, Raghavan SA, Dulin JN, Alge DL. Clickable Granular Hydrogel Scaffolds for Delivery of Neural Progenitor Cells to Sites of Spinal Cord Injury. Adv Healthc Mater 2024; 13:e2303912. [PMID: 38470994 PMCID: PMC11390979 DOI: 10.1002/adhm.202303912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 02/27/2024] [Indexed: 03/14/2024]
Abstract
Spinal cord injury (SCI) is a serious condition with limited treatment options. Neural progenitor cell (NPC) transplantation is a promising treatment option, and the identification of novel biomaterial scaffolds that support NPC engraftment and therapeutic activity is a top research priority. The objective of this study is to evaluate in situ assembled poly (ethylene glycol) (PEG)-based granular hydrogels for NPC delivery in a murine model of SCI. Microgel precursors are synthesized by using thiol-norbornene click chemistry to react four-armed PEG-amide-norbornene with enzymatically degradable and cell adhesive peptides. Unreacted norbornene groups are utilized for in situ assembly into scaffolds using a PEG-di-tetrazine linker. The granular hydrogel scaffolds exhibit good biocompatibility and do not adversely affect the inflammatory response after SCI. Moreover, when used to deliver NPCs, the granular hydrogel scaffolds supported NPC engraftment, do not adversely affect the immune response to the NPC grafts, and successfully support graft differentiation toward neuronal or astrocytic lineages as well as axonal extension into the host tissue. Collectively, these data establish PEG-based granular hydrogel scaffolds as a suitable biomaterial platform for NPC delivery and justify further testing, particularly in the context of more severe SCI.
Collapse
Affiliation(s)
- Thomas J Tigner
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Gabrielle Dampf
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Ashley Tucker
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Yu-Chi Huang
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Vipin Jagrit
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
| | - Abigail J Clevenger
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Arpita Mohapatra
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Shreya A Raghavan
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
| | - Jennifer N Dulin
- Department of Biology, Texas A&M University, College Station, TX, 77843-3258, USA
- Texas A&M Institute for Neuroscience, Texas A&M University, College Station, TX, 77843-3474, USA
| | - Daniel L Alge
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843-3120, USA
- Department of Materials Science and Engineering, Texas A&M University, College Station, TX, 77843-3003, USA
| |
Collapse
|
9
|
Du Z, Qiao F, Tong L, Zhang W, Mou X, Zhao X, Maitz MF, Wang H, Huang N, Yang Z. Mimicking Mytilus edulis foot protein: A versatile strategy for robust biomedical coatings. Innovation (N Y) 2024; 5:100671. [PMID: 39114479 PMCID: PMC11305295 DOI: 10.1016/j.xinn.2024.100671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 06/28/2024] [Indexed: 08/10/2024] Open
Abstract
Universal coatings with versatile surface adhesion, good mechanochemical robustness, and the capacity for secondary modification are of great scientific interest. However, incorporating these advantages into a system is still a great challenge. Here, we report a series of catechol-decorated polyallylamines (CPAs), denoted as pseudo-Mytilus edulis foot protein 5 (pseudo-Mefp-5), that mimic not only the catechol and amine groups but also the backbone of Mefp-5. CPAs can fabricate highly adhesive, robust, multifunctional polyCPA (PCPA) coatings based on synergetic catechol-polyamine chemistry as universal building blocks. Due to the interpenetrating entangled network architectures, these coatings exhibit high chemical robustness against harsh conditions (HCl, pH 1; NaOH, pH 14; H2O2, 30%), good mechanical robustness, and wear resistance. In addition, PCPA coatings provide abundant grafting sites, enabling the fabrication of various functional surfaces through secondary modification. Furthermore, the versatility, multifaceted robustness, and scalability of PCPA coatings indicate their great potential for surface engineering, especially for withstanding harsh conditions in multipurpose biomedical applications.
Collapse
Affiliation(s)
- Zeyu Du
- School of Materials Science and Engineering, Department of Cardiology, Third People’s Hospital of Chengdu Affiliated with Southwest Jiaotong University, Southwest Jiaotong University, Chengdu 610031, China
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, China
| | - Feng Qiao
- School of Materials Science and Engineering, Department of Cardiology, Third People’s Hospital of Chengdu Affiliated with Southwest Jiaotong University, Southwest Jiaotong University, Chengdu 610031, China
| | - Liping Tong
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wentai Zhang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, China
| | - Xiaohui Mou
- School of Materials Science and Engineering, Department of Cardiology, Third People’s Hospital of Chengdu Affiliated with Southwest Jiaotong University, Southwest Jiaotong University, Chengdu 610031, China
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, China
| | - Xin Zhao
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, China
| | - Manfred F. Maitz
- School of Materials Science and Engineering, Department of Cardiology, Third People’s Hospital of Chengdu Affiliated with Southwest Jiaotong University, Southwest Jiaotong University, Chengdu 610031, China
- Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Hohe Strasse 6, 01069 Dresden, Germany
| | - Huaiyu Wang
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Nan Huang
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, China
- GuangZhou Nanchuang Mount Everest Company for Medical Science and Technology, Guangzhou 510670, China
| | - Zhilu Yang
- School of Materials Science and Engineering, Department of Cardiology, Third People’s Hospital of Chengdu Affiliated with Southwest Jiaotong University, Southwest Jiaotong University, Chengdu 610031, China
- Dongguan Key Laboratory of Smart Biomaterials and Regenerative Medicine, The Tenth Affiliated Hospital of Southern Medical University, Dongguan 523059, China
| |
Collapse
|
10
|
Wang Y, Chang C, Wang R, Li X, Bao X. The advantages of multi-level omics research on stem cell-based therapies for ischemic stroke. Neural Regen Res 2024; 19:1998-2003. [PMID: 38227528 DOI: 10.4103/1673-5374.390959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 10/11/2023] [Indexed: 01/17/2024] Open
Abstract
Stem cell transplantation is a potential therapeutic strategy for ischemic stroke. However, despite many years of preclinical research, the application of stem cells is still limited to the clinical trial stage. Although stem cell therapy can be highly beneficial in promoting functional recovery, the precise mechanisms of action that are responsible for this effect have yet to be fully elucidated. Omics analysis provides us with a new perspective to investigate the physiological mechanisms and multiple functions of stem cells in ischemic stroke. Transcriptomic, proteomic, and metabolomic analyses have become important tools for discovering biomarkers and analyzing molecular changes under pathological conditions. Omics analysis could help us to identify new pathways mediated by stem cells for the treatment of ischemic stroke via stem cell therapy, thereby facilitating the translation of stem cell therapies into clinical use. In this review, we summarize the pathophysiology of ischemic stroke and discuss recent progress in the development of stem cell therapies for the treatment of ischemic stroke by applying multi-level omics. We also discuss changes in RNAs, proteins, and metabolites in the cerebral tissues and body fluids under stroke conditions and following stem cell treatment, and summarize the regulatory factors that play a key role in stem cell therapy. The exploration of stem cell therapy at the molecular level will facilitate the clinical application of stem cells and provide new treatment possibilities for the complete recovery of neurological function in patients with ischemic stroke.
Collapse
Affiliation(s)
- Yiqing Wang
- 4+4 Doctor Medical Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chuheng Chang
- 4+4 Doctor Medical Program, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoguang Li
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
11
|
Kim KM, D'Elia AM, Rodell CB. Hydrogel-based approaches to target hypersensitivity mechanisms underlying autoimmune disease. Adv Drug Deliv Rev 2024; 212:115395. [PMID: 39004347 DOI: 10.1016/j.addr.2024.115395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 06/23/2024] [Accepted: 07/08/2024] [Indexed: 07/16/2024]
Abstract
A robust adaptive immune response is essential for combatting pathogens. In the wrong context such as due to genetic and environmental factors, however, the same mechanisms crucial for self-preservation can lead to a loss of self-tolerance. Resulting autoimmunity manifests in the development of a host of organ-specific or systemic autoimmune diseases, hallmarked by aberrant immune responses and tissue damage. The prevalence of autoimmune diseases is on the rise, medical management of which focuses primarily on pharmacological immunosuppression that places patients at a risk of side effects, including opportunistic infections and tumorigenesis. Biomaterial-based drug delivery systems confer many opportunities to address challenges associated with conventional disease management. Hydrogels, in particular, can protect encapsulated cargo (drug or cell therapeutics) from the host environment, afford their presentation in a controlled manner, and can be tailored to respond to disease conditions or support treatment via multiplexed functionality. Moreover, localized delivery to affected sites by these approaches has the potential to concentrate drug action at the site, reduce off-target exposure, and enhance patient compliance by reducing the need for frequent administration. Despite their many benefits for the management of autoimmune disease, such biomaterial-based approaches focus largely on the downstream effects of hypersensitivity mechanisms and have a limited capacity to eradicate the disease. In contrast, direct targeting of mechanisms of hypersensitivity reactions uniquely enables prophylaxis or the arrest of disease progression by mitigating the basis of autoimmunity. One promising approach is to induce self-antigen-specific tolerance, which specifically subdues damaging autoreactivity while otherwise retaining the normal immune responses. In this review, we will discuss hydrogel-based systems for the treatment of autoimmune disease, with a focus on those that target hypersensitivity mechanisms head-on. As the field continues to advance, it will expand the range of therapeutic choices for people coping with autoimmune diseases, providing fresh prospects for better clinical outcomes and improved quality of life.
Collapse
Affiliation(s)
- Kenneth M Kim
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA.
| | - Arielle M D'Elia
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| | - Christopher B Rodell
- Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA, USA; School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Weber RZ, Buil BA, Rentsch NH, Bosworth A, Zhang M, Kisler K, Tackenberg C, Zlokovic BV, Rust R. A molecular brain atlas reveals cellular shifts during the repair phase of stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.21.608971. [PMID: 39229128 PMCID: PMC11370539 DOI: 10.1101/2024.08.21.608971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Ischemic stroke triggers a cascade of pathological events that affect multiple cell types and often lead to incomplete functional recovery. Despite advances in single-cell technologies, the molecular and cellular responses that contribute to long-term post-stroke impairment remain poorly understood. To gain better insight into the underlying mechanisms, we generated a single-cell transcriptomic atlas from distinct brain regions using a mouse model of permanent focal ischemia at one month post-injury. Our findings reveal cell- and region-specific changes within the stroke-injured and peri-infarct brain tissue. For instance, GABAergic and glutamatergic neurons exhibited upregulated genes in signaling pathways involved in axon guidance and synaptic plasticity, and downregulated pathways associated with aerobic metabolism. Using cell-cell communication analysis, we identified increased strength in predicted interactions within stroke tissue among both neural and non-neural cells via signaling pathways such as those involving collagen, protein tyrosine phosphatase receptor, neuronal growth regulator, laminin, and several cell adhesion molecules. Furthermore, we found a strong correlation between mouse transcriptome responses after stroke and those observed in human nonfatal brain stroke lesions. Common molecular features were linked to inflammatory responses, extracellular matrix organization, and angiogenesis. Our findings provide a detailed resource for advancing our molecular understanding of stroke pathology and for discovering therapeutic targets in the repair phase of stroke recovery.
Collapse
Affiliation(s)
- Rebecca Z Weber
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Beatriz Achón Buil
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Nora H Rentsch
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Allison Bosworth
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Mingzi Zhang
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Kassandra Kisler
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Christian Tackenberg
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
| | - Berislav V Zlokovic
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| | - Ruslan Rust
- Institute for Regenerative Medicine, University of Zurich, Schlieren, Switzerland
- Neuroscience Center Zurich, ETH Zurich and University of Zurich, Zurich, Switzerland
- Department of Physiology and Neuroscience, University of Southern California, Los Angeles, USA
- Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, USA
| |
Collapse
|
13
|
Fernández-Serra R, Lekouaghet A, Peracho L, Yonesi M, Alcázar A, Chioua M, Marco-Contelles J, Pérez-Rigueiro J, Rojo FJ, Panetsos F, Guinea GV, González-Nieto D. Permselectivity of Silk Fibroin Hydrogels for Advanced Drug Delivery Neurotherapies. Biomacromolecules 2024; 25:5233-5250. [PMID: 39018332 PMCID: PMC11323009 DOI: 10.1021/acs.biomac.4c00629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/20/2024] [Accepted: 05/21/2024] [Indexed: 07/19/2024]
Abstract
A promising trend in tissue engineering is using biomaterials to improve the control of drug concentration in targeted tissue. These vehicular systems are of specific interest when the required treatment time window is higher than the stability of therapeutic molecules in the body. Herein, the capacity of silk fibroin hydrogels to release different molecules and drugs in a sustained manner was evaluated. We found that a biomaterial format, obtained by an entirely aqueous-based process, could release molecules of variable molecular weight and charge with a preferential delivery of negatively charged molecules. Although the theoretical modeling suggested that drug delivery was more likely to be driven by Fickian diffusion, the external media had a considerable influence on the release, with lipophilic organic solvents such as acetonitrile-methanol (ACN-MeOH) intensifying the release of hydrophobic molecules. Second, we found that silk fibroin could be used as a vehicular system to treat a variety of brain disorders as this biomaterial sustained the release of different factors with neurotrophic (brain-derived neurotrophic factor) (BDNF), chemoattractant (C-X-C motif chemokine 12) (CXCL12), anti-inflammatory (TGF-β-1), and angiogenic (VEGF) capacities. Finally, we demonstrated that this biomaterial hydrogel could release cholesteronitrone ISQ201, a nitrone with antioxidant capacity, showing neuroprotective activity in an in vitro model of ischemia-reoxygenation. Given the slow degradation rate shown by silk fibroin in many biological tissues, including the nervous system, our study expands the restricted list of drug delivery-based biomaterial systems with therapeutic capacity for both short- and especially long-term treatment windows and has merit for use with brain pathologies.
Collapse
Affiliation(s)
- Rocío Fernández-Serra
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
| | - Amira Lekouaghet
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
| | - Lorena Peracho
- Department
of Research, Hospital Universitario Ramón
y Cajal, Madrid 28034, Spain
- Proteomics
Unit, Instituto Ramón y Cajal de
Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Mahdi Yonesi
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
| | - Alberto Alcázar
- Department
of Research, Hospital Universitario Ramón
y Cajal, Madrid 28034, Spain
- Proteomics
Unit, Instituto Ramón y Cajal de
Investigación Sanitaria (IRYCIS), Madrid 28034, Spain
| | - Mourad Chioua
- Laboratory
of Medicinal Chemistry, Institute of General
Organic Chemistry (CSIC), Madrid 28006, Spain
| | - José Marco-Contelles
- Laboratory
of Medicinal Chemistry, Institute of General
Organic Chemistry (CSIC), Madrid 28006, Spain
- Center
for
Biomedical Network Research on Rare Diseases (CIBERER), CIBER, ISCIII, Madrid 28029, Spain
| | - José Pérez-Rigueiro
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Departamento
de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
| | - Francisco J. Rojo
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Departamento
de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid 28040, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
| | - Fivos Panetsos
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
- Neurocomputing
and Neurorobotics Research Group, Faculty of Biology and Faculty of
Optics, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Gustavo V. Guinea
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Departamento
de Ciencia de Materiales, ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid 28040, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Biomaterials
and Regenerative Medicine Group, Instituto de Investigación
Sanitaria del Hospital Clínico San Carlos (IdISSC), Calle Prof. Martín Lagos s/n, Madrid 28040, Spain
| | - Daniel González-Nieto
- Center
for Biomedical Technology, Universidad Politécnica
de Madrid, Pozuelo de Alarcón 28223, Spain
- Silk
Biomed SL, Calle Navacerrada
18, Urb. Puerto Galapagar. Galapagar 28260, Spain
- Centro
de Investigación Biomédica en Red de Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), Instituto de Salud Carlos III, Madrid 28029, Spain
- Departamento de
Tecnología Fotónica y Bioingeniería,
ETSI Telecomunicaciones, Universidad Politécnica
de Madrid, Madrid 28040, Spain
| |
Collapse
|
14
|
Wang Y, Ran G, Zhang Q, Zhang Q. The association between social support and prosocial behavior: A three-level meta-analysis. Psych J 2024. [PMID: 39034601 DOI: 10.1002/pchj.792] [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: 03/14/2023] [Accepted: 06/18/2024] [Indexed: 07/23/2024]
Abstract
Numerous studies have examined the relationship between social support and prosocial behavior and have concluded that social support is an important factor in generating prosocial behavior. However, different studies have produced different conclusions, and the moderating effect on the relationship is not entirely clear. The current study uses a three-level meta-analysis method to clarify the relationship between social support and prosocial behavior, and explores the moderating variables that affect the relationship between the two variables. Through a systematic literature search, a total of 92 studies, 418 effect sizes, and 74,378 participants were obtained. The main effects test found a significant positive correlation between social support and prosocial behavior. Tests of the moderating effects indicated that the relationship between social support and prosocial behavior was moderated by year of publication, source of social support, measurement of social support and measurement of prosocial behavior. In summary, social support plays an important role in prosocial behavior, and exploring their relationship is beneficial to families, schools and society in guiding individuals' prosocial behavior.
Collapse
Affiliation(s)
- Yinlan Wang
- Department of Psychology, School of Education, China West Normal University, Nanchong, China
| | - Guangming Ran
- Department of Psychology, School of Education, China West Normal University, Nanchong, China
| | - Qi Zhang
- College of Preschool and Primary Education, China West Normal University, Nanchong, China
| | - Qiongzhi Zhang
- Department of Psychology, School of Education, China West Normal University, Nanchong, China
| |
Collapse
|
15
|
Michór P, Renardson L, Li S, Boltze J. Neurorestorative Approaches for Ischemic StrokeChallenges, Opportunities, and Recent Advances. Neuroscience 2024; 550:69-78. [PMID: 38763225 DOI: 10.1016/j.neuroscience.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/21/2024]
Abstract
Despite recent advances in acute stroke management, most patients experiencing a stroke will suffer from residual brain damage and functional impairment. Addressing those residual deficits would require neurorestoration, i.e., rebuilding brain tissue to repair the structural brain damage caused by stroke. However, there are major pathobiological, anatomical and technological hurdles making neurorestorative approaches remarkably challenging, and true neurorestoration after larger ischemic lesions could not yet be achieved. On the other hand, there has been steady advancement in our understanding of the limits of tissue regeneration in the adult mammalian brain as well as of the fundamental organization of brain tissue growth during embryo- and ontogenesis. This has been paralleled by the development of novel animal models to study stroke, advancement of biomaterials that can be used to support neurorestoration, and in stem cell technologies. This review gives a detailed explanation of the major hurdles so far preventing the achievement of neurorestoration after stroke. It will also describe novel concepts and advancements in biomaterial science, brain organoid culturing, and animal modeling that may enable the investigation of post-stroke neurorestorative approaches in translationally relevant setups. Finally, there will be a review of recent achievements in experimental studies that have the potential to be the starting point of research and development activities that may eventually bring post-stroke neurorestoration within reach.
Collapse
Affiliation(s)
- Paulina Michór
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom
| | - Lydia Renardson
- University of Warwick, Warwick Medical School, Coventry CV4 7AL, United Kingdom
| | - Shen Li
- Department of Neurology and Psychiatry, Beijing Shijitan Hospital, Capital Medical University, Beijing, China; Beijing Institute of Brain Disorders, Capital Medical University, Beijing, China
| | - Johannes Boltze
- University of Warwick, School of Life Sciences, Coventry CV4 7AL, United Kingdom.
| |
Collapse
|
16
|
Kang H, Huang Y, Peng H, Zhang X, Liu Y, Liu Y, Xia Y, Liu S, Wu Y, Wang S, Lei T, Zhang H. Mesenchymal Stem Cell-Loaded Hydrogel Improves Surgical Treatment for Chronic Cerebral Ischemia. Transl Stroke Res 2024:10.1007/s12975-024-01274-5. [PMID: 38977638 DOI: 10.1007/s12975-024-01274-5] [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: 05/09/2024] [Revised: 06/11/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
Chronic cerebral ischemia (CCI) results in a prolonged insufficient blood supply to the brain tissue, leading to impaired neuronal function and subsequent impairment of cognitive and motor abilities. Our previous research showed that in mice with bilateral carotid artery stenosis, the collateral neovascularization post Encephalo-myo-synangiosis (EMS) treatment could be facilitated by bone marrow mesenchymal stem cells (MSCs) transplantation. Considering the advantages of biomaterials, we synthesized and modified a gelatin hydrogel for MSCs encapsulation. We then applied this hydrogel on the brain surface during EMS operation in rats with CCI, and evaluated its impact on cognitive performance and collateral circulation. Consequently, MSCs encapsulated in hydrogel significantly augment the therapeutic effects of EMS, potentially by promoting neovascularization, facilitating neuronal differentiation, and suppressing neuroinflammation. Furthermore, taking advantage of multi-RNA-sequencing and in silico analysis, we revealed that MSCs loaded in hydrogel regulate PDCD4 and CASP2 through the overexpression of miR-183-5p and miR-96-5p, thereby downregulating the expression of apoptosis-related proteins and inhibiting early apoptosis. In conclusion, a gelatin hydrogel to enhance the functionality of MSCs has been developed, and its combination with EMS treatment can improve the therapeutic effect in rats with CCI, suggesting its potential clinical benefit.
Collapse
Affiliation(s)
- Huayu Kang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yimin Huang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huan Peng
- Cellular Signaling Laboratory, International Research Center for Sensory Biology and Technology of MOST, Key Laboratory of Molecular Biophysics of MOE, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, Hubei, China
| | - Xincheng Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuan Liu
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yanchao Liu
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yuze Xia
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Shengwen Liu
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yaqi Wu
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Sheng Wang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Ting Lei
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Huaqiu Zhang
- Department of Neurosurgery, Tongji Hospital of Tongji Medical College of Huazhong University Science and Technology, Jiefang Avenue 1095, Wuhan, 430030, China.
- Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
17
|
Singhal R, Sarangi MK, Rath G. Injectable Hydrogels: A Paradigm Tailored with Design, Characterization, and Multifaceted Approaches. Macromol Biosci 2024; 24:e2400049. [PMID: 38577905 DOI: 10.1002/mabi.202400049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/22/2024] [Indexed: 04/06/2024]
Abstract
Biomaterials denoting self-healing and versatile structural integrity are highly curious in the biomedicine segment. The injectable and/or printable 3D printing technology is explored in a few decades back, which can alter their dimensions temporarily under shear stress, showing potential healing/recovery tendency with patient-specific intervention toward the development of personalized medicine. Thus, self-healing injectable hydrogels (IHs) are stunning toward developing a paradigm for tissue regeneration. This review comprises the designing of IHs, rheological characterization and stability, several benchmark consequences for self-healing IHs, their translation into tissue regeneration of specific types, applications of IHs in biomedical such as anticancer and immunomodulation, wound healing and tissue/bone regeneration, antimicrobial potentials, drugs, gene and vaccine delivery, ocular delivery, 3D printing, cosmeceuticals, and photothermal therapy as well as in other allied avenues like agriculture, aerospace, electronic/electrical industries, coating approaches, patents associated with therapeutic/nontherapeutic avenues, and numerous futuristic challenges and solutions.
Collapse
Affiliation(s)
- Rishika Singhal
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Manoj Kumar Sarangi
- Department of Pharmaceutics, Amity Institute of Pharmacy, Amity University, Malhaur Railway Station Road, Gomti Nagar, Lucknow, Uttar Pradesh, 201313, India
| | - Goutam Rath
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Siksha 'O' Anusandhan University, Bhubaneswar, Odisha, 751030, India
| |
Collapse
|
18
|
Luo W, Zhang H, Wan R, Cai Y, Liu Y, Wu Y, Yang Y, Chen J, Zhang D, Luo Z, Shang X. Biomaterials-Based Technologies in Skeletal Muscle Tissue Engineering. Adv Healthc Mater 2024; 13:e2304196. [PMID: 38712598 DOI: 10.1002/adhm.202304196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 04/26/2024] [Indexed: 05/08/2024]
Abstract
For many clinically prevalent severe injuries, the inherent regenerative capacity of skeletal muscle remains inadequate. Skeletal muscle tissue engineering (SMTE) seeks to meet this clinical demand. With continuous progress in biomedicine and related technologies including micro/nanotechnology and 3D printing, numerous studies have uncovered various intrinsic mechanisms regulating skeletal muscle regeneration and developed tailored biomaterial systems based on these understandings. Here, the skeletal muscle structure and regeneration process are discussed and the diverse biomaterial systems derived from various technologies are explored in detail. Biomaterials serve not merely as local niches for cell growth, but also as scaffolds endowed with structural or physicochemical properties that provide tissue regenerative cues such as topographical, electrical, and mechanical signals. They can also act as delivery systems for stem cells and bioactive molecules that have been shown as key participants in endogenous repair cascades. To achieve bench-to-bedside translation, the typical effect enabled by biomaterial systems and the potential underlying molecular mechanisms are also summarized. Insights into the roles of biomaterials in SMTE from cellular and molecular perspectives are provided. Finally, perspectives on the advancement of SMTE are provided, for which gene therapy, exosomes, and hybrid biomaterials may hold promise to make important contributions.
Collapse
Affiliation(s)
- Wei Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Hanli Zhang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Renwen Wan
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yuxi Cai
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yinuo Liu
- The Second Clinical Medical College of Nanchang University, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yang Wu
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Yimeng Yang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Jiani Chen
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Deju Zhang
- Food and Nutritional Sciences, School of Biological Sciences, The University of Hong Kong, Pokfulam Road, Hong Kong, 999077, Hong Kong
| | - Zhiwen Luo
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| | - Xiliang Shang
- Department of Sports Medicine Huashan Hospital, Fudan University, Shanghai, 200040, P. R. China
| |
Collapse
|
19
|
Wasnik K, Gupta PS, Singh G, Maity S, Patra S, Pareek D, Kumar S, Rai V, Prakash R, Acharya A, Maiti P, Mukherjee S, Mastai Y, Paik P. Neurogenic and angiogenic poly( N-acryloylglycine)- co-(acrylamide)- co-( N-acryloyl-glutamate) hydrogel: preconditioning effect under oxidative stress and use in neuroregeneration. J Mater Chem B 2024; 12:6221-6241. [PMID: 38835196 DOI: 10.1039/d4tb00243a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Traumatic injuries, neurodegenerative diseases and oxidative stress serve as the early biomarkers for neuronal damage and impede angiogenesis and subsequently neuronal growth. Considering this, the present work aimed to develop a poly(N-acryloylglycine)-co-(acrylamide)-co-(N-acryloylglutamate) hydrogel [p(NAG-Ac-NAE)] with angiogenesis/neurogenesis properties. As constituents of this polymer modulate their vital role in biological functions, inhibitory neurotransmitter glycine regulates neuronal homeostasis, and glutamatergic signalling regulates angiogenesis. The p(NAG-Ac-NAE) hydrogel is a highly branched, biodegradable and pH-responsive polymer with a very high swelling behavior of 6188%. The mechanical stability (G', 2.3-2.7 kPa) of this polymeric hydrogel is commendable in the differentiation of mature neurons. This hydrogel is biocompatible (as tested in HUVEC cells) and helps to proliferate PC12 cells (152.7 ± 13.7%), whereas it is cytotoxic towards aggressive cancers such as glioblastoma (LN229 cells) and triple negative breast cancer (TNBC; MDA-MB-231 cells) and helps to maintain the healthy cytoskeleton framework structure of primary cortical neurons by facilitating the elongation of the axonal pathway. Furthermore, FACS results revealed that the synthesized hydrogel potentiates neurogenesis by inducing the cell cycle (G0/G1) and arresting the sub-G1 phase by limiting apoptosis. Additionally, RT-PCR results revealed that this hydrogel induced an increased level of HIF-1α expression, providing preconditioning effects towards neuronal cells under oxidative stress by scavenging ROS and initiating neurogenic and angiogenic signalling. This hydrogel further exhibits more pro-angiogenic activities by increasing the expression of VEGF isoforms compared to previously reported hydrogels. In conclusion, the newly synthesized p(NAG-Ac-NAE) hydrogel can be one of the potential neuroregenerative materials for vasculogenesis-assisted neurogenic applications and paramount for the management of neurodegenerative diseases.
Collapse
Affiliation(s)
- Kirti Wasnik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Prem Shankar Gupta
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Gurmeet Singh
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Somedutta Maity
- School of Engineering Sciences and Technology, University of Hyderabad, Telangana State 500 046, India
| | - Sukanya Patra
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Divya Pareek
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Sandeep Kumar
- Department of Zoology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Vipin Rai
- Department of Biochemistry, Institute of Sciences, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Ravi Prakash
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Arbind Acharya
- Department of Zoology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221005, India
| | - Pralay Maiti
- School of Material Science, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| | - Yitzhak Mastai
- Department of Chemistry and the Institute of Nanotechnology, Bar-Ilan University, Ramat-Gan, 52900, Israel
| | - Pradip Paik
- School of Biomedical Engineering, Indian Institute of Technology, Banaras Hindu University (BHU), Varanasi, Uttar Pradesh 221 005, India.
| |
Collapse
|
20
|
Wang Y, Huang R, Lu Y, Liu M, Mo R. Immuno-protective vesicle-crosslinked hydrogel for allogenic transplantation. Nat Commun 2024; 15:5176. [PMID: 38890279 PMCID: PMC11189436 DOI: 10.1038/s41467-024-49135-x] [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: 09/16/2023] [Accepted: 05/24/2024] [Indexed: 06/20/2024] Open
Abstract
The longevity of grafts remains a major challenge in allogeneic transplantation due to immune rejection. Systemic immunosuppression can impair graft function and can also cause severe adverse effects. Here, we report a local immuno-protective strategy to enhance post-transplant persistence of allografts using a mesenchymal stem cell membrane-derived vesicle (MMV)-crosslinked hydrogel (MMV-Gel). MMVs are engineered to upregulate expression of Fas ligand (FasL) and programmed death ligand 1 (PD-L1). The MMVs are retained within the hydrogel by crosslinking. The immuno-protective microenvironment of the hydrogel protects allografts by presenting FasL and PD-L1. The binding of these ligands to T effector cells, the dominant contributors to graft destruction and rejection, results in apoptosis of T effector cells and generation of regulatory T cells. We demonstrate that implantation with MMV-Gel prolongs the survival and function of grafts in mouse models of allogeneic pancreatic islet cells and skin transplantation.
Collapse
Affiliation(s)
- Yuqian Wang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Renqi Huang
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Yougong Lu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Mingqi Liu
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China
| | - Ran Mo
- State Key Laboratory of Natural Medicines, Jiangsu Key Laboratory of Drug Discovery for Metabolic Diseases and Jiangsu Key Laboratory of Drug Design and Optimization, Center of Advanced Pharmaceuticals and Biomaterials, School of Life Science and Technology, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
21
|
Li X, Li Y, Zhang X, Xu J, Kang J, Li B, Zhao B, Wang L. Cross-Linking Methods of the Silk Protein Hydrogel in Oral and Craniomaxillofacial Tissue Regeneration. Tissue Eng Regen Med 2024; 21:529-544. [PMID: 38294593 PMCID: PMC11087422 DOI: 10.1007/s13770-023-00624-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 12/16/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024] Open
Abstract
BACKGROUND Craniomaxillofacial tissue defects are clinical defects involving craniomaxillofacial and oral soft and hard tissues. They are characterized by defect-shaped irregularities, bacterial and inflammatory environments, and the need for functional recovery. Conventional clinical treatments are currently unable to achieve regeneration of high-quality oral craniomaxillofacial tissue. As a natural biomaterial, silk fibroin (SF) has been widely studied in biomedicine and has broad prospects for use in tissue regeneration. Hydrogels made of SF showed excellent water retention, biocompatibility, safety and the ability to combine with other materials. METHODS To gain an in-depth understanding of the current development of SF, this article reviews the structure, preparation and application prospects in oral and craniomaxillofacial tissue regenerative medicine. It first briefly introduces the structure of SF and then summarizes the principles, advantages and disadvantages of the different cross-linking methods (physical cross-linking, chemical cross-linking and double network structure) of SF. Finally, the existing research on the use of SF in tissue engineering and the prospects of using SF with different cross-linking methods in oral and craniomaxillofacial tissue regeneration are also discussed. CONCLUSIONS This review is intended to show the advantages of SF hydrogels in tissue engineering and provides theoretical support for establishing novel and viable silk protein hydrogels for regeneration.
Collapse
Affiliation(s)
- Xiujuan Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Yuanjiao Li
- School of Medical Sciences, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xinsong Zhang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Jie Xu
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Jie Kang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Bing Li
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China
| | - Bin Zhao
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
| | - Lu Wang
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, Shanxi, China.
| |
Collapse
|
22
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [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: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
23
|
Cao Y, Yu Y, Pan L, Han W, Zeng F, Wang J, Mei Q, Liu C. Sulfated Polysaccharide-Based Nanocarrier Drives Microenvironment-Mediated Cerebral Neurovascular Remodeling for Ischemic Stroke Treatment. NANO LETTERS 2024; 24:5214-5223. [PMID: 38649327 DOI: 10.1021/acs.nanolett.4c00650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2024]
Abstract
Stroke is a leading cause of global mortality and severe disability. However, current strategies used for treating ischemic stroke lack specific targeting capabilities, exhibit poor immune escape ability, and have limited drug release control. Herein, we developed an ROS-responsive nanocarrier for targeted delivery of the neuroprotective agent rapamycin (RAPA) to mitigate ischemic brain damage. The nanocarrier consisted of a sulfated chitosan (SCS) polymer core modified with a ROS-responsive boronic ester enveloped by a red blood cell membrane shell incorporating a stroke homing peptide. When encountering high levels of intracellular ROS in ischemic brain tissues, the release of SCS combined with RAPA from nanoparticle disintegration facilitates effective microglia polarization and, in turn, maintains blood-brain barrier integrity, reduces cerebral infarction, and promotes cerebral neurovascular remodeling in a mouse stroke model involving transient middle cerebral artery occlusion (tMCAO). This work offers a promising strategy to treat ischemic stroke therapy.
Collapse
Affiliation(s)
- Yinli Cao
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Yuanman Yu
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Lina Pan
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Weili Han
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
| | - Feng Zeng
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, People's Republic of China
| | - Jing Wang
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| | - Qiyong Mei
- School of Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Neurosurgery, Changzheng Hospital, Naval Medical University, 415 Fengyang Road, Shanghai 200003, People's Republic of China
| | - Changsheng Liu
- The State Key Laboratory of Bioreactor Engineering, Frontiers Science Center for Materiobiology and Dynamic Chemistry, East China University of Science and Technology, Shanghai 200237, People's Republic of China
| |
Collapse
|
24
|
Castro-Ribeiro ML, Castro VIB, Vieira de Castro J, Pires RA, Reis RL, Costa BM, Ferreira H, Neves NM. The Potential of the Fibronectin Inhibitor Arg-Gly-Asp-Ser in the Development of Therapies for Glioblastoma. Int J Mol Sci 2024; 25:4910. [PMID: 38732135 PMCID: PMC11084566 DOI: 10.3390/ijms25094910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 04/23/2024] [Accepted: 04/27/2024] [Indexed: 05/13/2024] Open
Abstract
Glioblastoma (GBM) is the most lethal and common malignant primary brain tumor in adults. An important feature that supports GBM aggressiveness is the unique composition of its extracellular matrix (ECM). Particularly, fibronectin plays an important role in cancer cell adhesion, differentiation, proliferation, and chemoresistance. Thus, herein, a hydrogel with mechanical properties compatible with the brain and the ability to disrupt the dynamic and reciprocal interaction between fibronectin and tumor cells was produced. High-molecular-weight hyaluronic acid (HMW-HA) functionalized with the inhibitory fibronectin peptide Arg-Gly-Asp-Ser (RGDS) was used to produce the polymeric matrix. Liposomes encapsulating doxorubicin (DOX) were also included in the hydrogel to kill GBM cells. The resulting hydrogel containing liposomes with therapeutic DOX concentrations presented rheological properties like a healthy brain. In vitro assays demonstrated that unmodified HMW-HA hydrogels only caused GBM cell killing after DOX incorporation. Conversely, RGDS-functionalized hydrogels displayed per se cytotoxicity. As GBM cells produce several proteolytic enzymes capable of disrupting the peptide-HA bond, we selected MMP-2 to illustrate this phenomenon. Therefore, RGDS internalization can induce GBM cell apoptosis. Importantly, RGDS-functionalized hydrogel incorporating DOX efficiently damaged GBM cells without affecting astrocyte viability, proving its safety. Overall, the results demonstrate the potential of the RGDS-functionalized hydrogel to develop safe and effective GBM treatments.
Collapse
Affiliation(s)
- Maria L. Castro-Ribeiro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal; (M.L.C.-R.); (V.I.B.C.); (J.V.d.C.); (R.A.P.); (R.L.R.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
| | - Vânia I. B. Castro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal; (M.L.C.-R.); (V.I.B.C.); (J.V.d.C.); (R.A.P.); (R.L.R.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
| | - Joana Vieira de Castro
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal; (M.L.C.-R.); (V.I.B.C.); (J.V.d.C.); (R.A.P.); (R.L.R.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
| | - Ricardo A. Pires
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal; (M.L.C.-R.); (V.I.B.C.); (J.V.d.C.); (R.A.P.); (R.L.R.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
| | - Rui L. Reis
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal; (M.L.C.-R.); (V.I.B.C.); (J.V.d.C.); (R.A.P.); (R.L.R.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
| | - Bruno M. Costa
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
- Life and Health Sciences Research Institute (ICVS), School of Medicine, Campus Gualtar, University of Minho, 4710-057 Braga, Portugal
| | - Helena Ferreira
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal; (M.L.C.-R.); (V.I.B.C.); (J.V.d.C.); (R.A.P.); (R.L.R.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
| | - Nuno M. Neves
- 3B’s Research Group, I3Bs—Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Guimarães, Portugal; (M.L.C.-R.); (V.I.B.C.); (J.V.d.C.); (R.A.P.); (R.L.R.)
- ICVS/3B’s—PT Government Associate Laboratory, 4710-057/4805-017 Braga/Guimarães, Portugal;
| |
Collapse
|
25
|
Luo P, Liu W, Ye Z, Zhang Y, Zhang Z, Yi J, Zeng R, Yang S, Tu M. 26SCS-Loaded SilMA/Col Composite Sponge with Well-Arranged Layers Promotes Angiogenesis-Based Diabetic Wound Repair by Mediating Macrophage Inflammatory Response. Molecules 2024; 29:1832. [PMID: 38675654 PMCID: PMC11053466 DOI: 10.3390/molecules29081832] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/10/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Diabetic wound healing is a significant clinical challenge because abnormal immune cells in the wound cause chronic inflammation and impair tissue regeneration. Therefore, regulating the behavior and function of macrophages may be conducive to improving treatment outcomes in diabetic wounds. Herein, sulfated chitosan (26SCS)-containing composite sponges (26SCS-SilMA/Col-330) with well-arranged layers and high porosity were constructed based on collagen and silk fibroin, aiming to induce an appropriate inflammatory response and promote angiogenesis. The results indicated that the ordered topological structure of composite sponges could trigger the pro-inflammatory response of Mφs in the early stage, and rapid release of 26SCS in the early and middle stages (within the concentration range of 1-3 mg/mL) induced a positive inflammatory response; initiated the pro-inflammatory reaction of Mφs within 3 days; shifted M1 Mφs to the M2 phenotype within 3-7 days; and significantly up-regulated the expression of two typical angiogenic growth factors, namely VEGF and PDGF-BB, on day 7, leading to rapid HUVEC migration and angiogenesis. In vivo data also demonstrated that on the 14th day after surgery, the 26SCS-SilMA/Col-330-implanted areas exhibited less inflammation, faster re-epithelialization, more abundant collagen deposition and a greater number of blood vessels in the skin tissue. The composite sponges with higher 26SCS contents (the (5.0) 26SCS-SilMA/Col-330 and the (7.5) 26SCS-SilMA/Col-330) could better orchestrate the phenotype and function of Mφs and facilitate wound healing. These findings highlight that the 26SCS-SilMA/Col-330 sponges developed in this work might have great potential as a novel dressing for the treatment of diabetic wounds.
Collapse
Affiliation(s)
- Pin Luo
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Wei Liu
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Zhangyao Ye
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Yuyu Zhang
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Zekun Zhang
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Jing Yi
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Rong Zeng
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Shenyu Yang
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| | - Mei Tu
- College of Chemistry and Materials Science, Jinan University, Huangpu Road 601, Guangzhou 510632, China; (P.L.); (W.L.); (Z.Y.); (Y.Z.); (Z.Z.); (J.Y.); (R.Z.); (S.Y.)
- Engineering Research Center of Artificial Organs and Materials, Ministry of Education, Jinan University, Guangzhou 510632, China
| |
Collapse
|
26
|
Zareein A, Mahmoudi M, Jadhav SS, Wilmore J, Wu Y. Biomaterial engineering strategies for B cell immunity modulations. Biomater Sci 2024; 12:1981-2006. [PMID: 38456305 PMCID: PMC11019864 DOI: 10.1039/d3bm01841e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 02/23/2024] [Indexed: 03/09/2024]
Abstract
B cell immunity has a penetrating effect on human health and diseases. Therapeutics aiming to modulate B cell immunity have achieved remarkable success in combating infections, autoimmunity, and malignancies. However, current treatments still face significant limitations in generating effective long-lasting therapeutic B cell responses for many conditions. As the understanding of B cell biology has deepened in recent years, clearer regulation networks for B cell differentiation and antibody production have emerged, presenting opportunities to overcome current difficulties and realize the full therapeutic potential of B cell immunity. Biomaterial platforms have been developed to leverage these emerging concepts to augment therapeutic humoral immunity by facilitating immunogenic reagent trafficking, regulating T cell responses, and modulating the immune microenvironment. Moreover, biomaterial engineering tools have also advanced our understanding of B cell biology, further expediting the development of novel therapeutics. In this review, we will introduce the general concept of B cell immunobiology and highlight key biomaterial engineering strategies in the areas including B cell targeted antigen delivery, sustained B cell antigen delivery, antigen engineering, T cell help optimization, and B cell suppression. We will also discuss our perspective on future biomaterial engineering opportunities to leverage humoral immunity for therapeutics.
Collapse
Affiliation(s)
- Ali Zareein
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Mina Mahmoudi
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
| | - Shruti Sunil Jadhav
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
| | - Joel Wilmore
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Yaoying Wu
- Department of Biomedical Engineering, Syracuse University, Syracuse, NY, USA.
- The BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, USA
- Department of Microbiology & Immunology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
27
|
Adewumi HO, Berniac GI, McCarthy EA, O'Shea TM. Ischemic and hemorrhagic stroke lesion environments differentially alter the glia repair potential of neural progenitor cell and immature astrocyte grafts. Exp Neurol 2024; 374:114692. [PMID: 38244885 DOI: 10.1016/j.expneurol.2024.114692] [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: 08/14/2023] [Revised: 01/03/2024] [Accepted: 01/15/2024] [Indexed: 01/22/2024]
Abstract
Using cell grafting to direct glia-based repair mechanisms in adult CNS injuries represents a potential therapeutic strategy for supporting functional neural parenchymal repair. However, glia repair directed by neural progenitor cell (NPC) grafts is dramatically altered by increasing lesion size, severity, and mode of injury. To address this, we studied the interplay between astrocyte differentiation and cell proliferation of NPC in vitro to generate proliferating immature astrocytes (ImA) using hysteretic conditioning. ImA maintain proliferation rates at comparable levels to NPC but showed robust immature astrocyte marker expression including Gfap and Vimentin. ImA demonstrated enhanced resistance to myofibroblast-like phenotypic transformations upon exposure to serum enriched environments in vitro compared to NPC and were more effective at scratch wound closure in vitro compared to quiescent astrocytes. Glia repair directed by ImA at acute ischemic striatal stroke lesions was equivalent to NPC but better than quiescent astrocyte grafts. While ischemic injury environments supported enhanced survival of grafts compared to healthy striatum, hemorrhagic lesions were hostile towards both NPC and ImA grafts leading to poor survival and ineffective modulation of natural wound repair processes. Our findings demonstrate that lesion environments, rather than transcriptional pre-graft states, determine the survival, cell-fate, and glia repair competency of cell grafts applied to acute CNS injuries.
Collapse
Affiliation(s)
- Honour O Adewumi
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA
| | - Gabriela I Berniac
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA
| | - Emily A McCarthy
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA
| | - Timothy M O'Shea
- Department of Biomedical Engineering, Boston University, Boston, MA 02215-2407, USA.
| |
Collapse
|
28
|
Wu X, Zhang T, Jia J, Chen Y, Zhang Y, Fang Z, Zhang C, Bai Y, Li Z, Li Y. Perspective insights into versatile hydrogels for stroke: From molecular mechanisms to functional applications. Biomed Pharmacother 2024; 173:116309. [PMID: 38479180 DOI: 10.1016/j.biopha.2024.116309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 02/13/2024] [Accepted: 02/17/2024] [Indexed: 03/27/2024] Open
Abstract
As the leading killer of life and health, stroke leads to limb paralysis, speech disorder, dysphagia, cognitive impairment, mental depression and other symptoms, which entail a significant financial burden to society and families. At present, physiology, clinical medicine, engineering, and materials science, advanced biomaterials standing on the foothold of these interdisciplinary disciplines provide new opportunities and possibilities for the cure of stroke. Among them, hydrogels have been endowed with more possibilities. It is well-known that hydrogels can be employed as potential biosensors, medication delivery vectors, and cell transporters or matrices in tissue engineering in tissue engineering, and outperform many traditional therapeutic drugs, surgery, and materials. Therefore, hydrogels become a popular scaffolding treatment option for stroke. Diverse synthetic hydrogels were designed according to different pathophysiological mechanisms from the recently reported literature will be thoroughly explored. The biological uses of several types of hydrogels will be highlighted, including pro-angiogenesis, pro-neurogenesis, anti-oxidation, anti-inflammation and anti-apoptosis. Finally, considerations and challenges of using hydrogels in the treatment of stroke are summarized.
Collapse
Affiliation(s)
- Xinghan Wu
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China; Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Tiejun Zhang
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jing Jia
- Department of Pharmacy, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Yining Chen
- Key laboratory for Leather Chemistry and Engineering of the Education Ministry, Sichuan University, Chengdu, Sichuan 610065, China
| | - Ying Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhenwei Fang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Chenyu Zhang
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yang Bai
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Zhengjun Li
- Department of Dermatology, Qilu Hospital, Shandong University, Jinan 250012, China
| | - Yuwen Li
- Department of Pharmacy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China.
| |
Collapse
|
29
|
Olteanu G, Neacșu SM, Joița FA, Musuc AM, Lupu EC, Ioniță-Mîndrican CB, Lupuliasa D, Mititelu M. Advancements in Regenerative Hydrogels in Skin Wound Treatment: A Comprehensive Review. Int J Mol Sci 2024; 25:3849. [PMID: 38612660 PMCID: PMC11012090 DOI: 10.3390/ijms25073849] [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: 01/30/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/14/2024] Open
Abstract
This state-of-the-art review explores the emerging field of regenerative hydrogels and their profound impact on the treatment of skin wounds. Regenerative hydrogels, composed mainly of water-absorbing polymers, have garnered attention in wound healing, particularly for skin wounds. Their unique properties make them well suited for tissue regeneration. Notable benefits include excellent water retention, creating a crucially moist wound environment for optimal healing, and facilitating cell migration, and proliferation. Biocompatibility is a key feature, minimizing adverse reactions and promoting the natural healing process. Acting as a supportive scaffold for cell growth, hydrogels mimic the extracellular matrix, aiding the attachment and proliferation of cells like fibroblasts and keratinocytes. Engineered for controlled drug release, hydrogels enhance wound healing by promoting angiogenesis, reducing inflammation, and preventing infection. The demonstrated acceleration of the wound healing process, particularly beneficial for chronic or impaired healing wounds, adds to their appeal. Easy application and conformity to various wound shapes make hydrogels practical, including in irregular or challenging areas. Scar minimization through tissue regeneration is crucial, especially in cosmetic and functional regions. Hydrogels contribute to pain management by creating a protective barrier, reducing friction, and fostering a soothing environment. Some hydrogels, with inherent antimicrobial properties, aid in infection prevention, which is a crucial aspect of successful wound healing. Their flexibility and ability to conform to wound contours ensure optimal tissue contact, enhancing overall treatment effectiveness. In summary, regenerative hydrogels present a promising approach for improving skin wound healing outcomes across diverse clinical scenarios. This review provides a comprehensive analysis of the benefits, mechanisms, and challenges associated with the use of regenerative hydrogels in the treatment of skin wounds. In this review, the authors likely delve into the application of rational design principles to enhance the efficacy and performance of hydrogels in promoting wound healing. Through an exploration of various methodologies and approaches, this paper is poised to highlight how these principles have been instrumental in refining the design of hydrogels, potentially revolutionizing their therapeutic potential in addressing skin wounds. By synthesizing current knowledge and highlighting potential avenues for future research, this review aims to contribute to the advancement of regenerative medicine and ultimately improve clinical outcomes for patients with skin wounds.
Collapse
Affiliation(s)
- Gabriel Olteanu
- Department of Clinical Laboratory and Food Safety, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania; (G.O.); (M.M.)
| | - Sorinel Marius Neacșu
- Department of Pharmaceutical Technology and Bio-Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania; (S.M.N.); (D.L.)
| | - Florin Alexandru Joița
- Department of Pharmaceutical Technology and Bio-Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania; (S.M.N.); (D.L.)
| | | | - Elena Carmen Lupu
- Department of Mathematics and Informatics, Faculty of Pharmacy, “Ovidius” University of Constanta, 900001 Constanta, Romania;
| | - Corina-Bianca Ioniță-Mîndrican
- Department of Toxicology, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania;
| | - Dumitru Lupuliasa
- Department of Pharmaceutical Technology and Bio-Pharmacy, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020945 Bucharest, Romania; (S.M.N.); (D.L.)
| | - Magdalena Mititelu
- Department of Clinical Laboratory and Food Safety, Faculty of Pharmacy, Carol Davila University of Medicine and Pharmacy, 020956 Bucharest, Romania; (G.O.); (M.M.)
| |
Collapse
|
30
|
Politrón-Zepeda GA, Fletes-Vargas G, Rodríguez-Rodríguez R. Injectable Hydrogels for Nervous Tissue Repair-A Brief Review. Gels 2024; 10:190. [PMID: 38534608 PMCID: PMC10970171 DOI: 10.3390/gels10030190] [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: 01/18/2024] [Revised: 02/25/2024] [Accepted: 03/06/2024] [Indexed: 03/28/2024] Open
Abstract
The repair of nervous tissue is a critical research field in tissue engineering because of the degenerative process in the injured nervous system. In this review, we summarize the progress of injectable hydrogels using in vitro and in vivo studies for the regeneration and repair of nervous tissue. Traditional treatments have not been favorable for patients, as they are invasive and inefficient; therefore, injectable hydrogels are promising for the treatment of damaged tissue. This review will contribute to a better understanding of injectable hydrogels as potential scaffolds and drug delivery system for neural tissue engineering applications.
Collapse
Affiliation(s)
- Gladys Arline Politrón-Zepeda
- Ingeniería en Sistemas Biológicos, Centro Universitario de los Valles (CUVALLES), Universidad de Guadalajara, Carretera Guadalajara-Ameca Km. 45.5, Ameca 46600, Jalisco, Mexico;
| | - Gabriela Fletes-Vargas
- Departamento de Ciencias Clínicas, Centro Universitario de los Altos (CUALTOS), Universidad de Guadalajara, Carretera Tepatitlán-Yahualica de González Gallo, Tepatitlán de Morelos 47620, Jalisco, Mexico;
| | - Rogelio Rodríguez-Rodríguez
- Departamento de Ciencias Naturales y Exactas, Centro Universitario de los Valles (CUVALLES), Universidad de Guadalajara, Carretera Guadalajara-Ameca Km. 45.5, Ameca 46600, Jalisco, Mexico
| |
Collapse
|
31
|
Martínez-Ramírez J, Toldos-Torres M, Benayas E, Villar-Gómez N, Fernández-Méndez L, Espinosa FM, García R, Veintemillas-Verdaguer S, Morales MDP, Serrano MC. Hybrid hydrogels support neural cell culture development under magnetic actuation at high frequency. Acta Biomater 2024; 176:156-172. [PMID: 38281674 DOI: 10.1016/j.actbio.2024.01.030] [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: 10/21/2023] [Revised: 01/08/2024] [Accepted: 01/22/2024] [Indexed: 01/30/2024]
Abstract
The combination of hydrogels and magnetic nanoparticles, scarcely explored to date, offers a wide range of possibilities for innovative therapies. Herein, we have designed hybrid 3D matrices integrating natural polymers, such as collagen, chitosan (CHI) and hyaluronic acid (HA), to provide soft and flexible 3D networks mimicking the extracellular matrix of natural tissues, and iron oxide nanoparticles (IONPs) that deliver localized heat when exposed to an alternating magnetic field (AMF). First, colloidally stable nanoparticles with a hydrodynamic radius of ∼20 nm were synthesized and coated with either CHI (NPCHI) or HA (NPHA). Then, collagen hydrogels were homogeneously loaded with these coated-IONPs resulting in soft (E0 ∼ 2.6 kPa), biodegradable and magnetically responsive matrices. Polymer-coated IONPs in suspension preserved primary neural cell viability and neural differentiation even at the highest dose (0.1 mg Fe/mL), regardless of the coating, even boosting neuronal interconnectivity at lower doses. Magnetic hydrogels maintained high neural cell viability and sustained the formation of highly interconnected and differentiated neuronal networks. Interestingly, those hydrogels loaded with the highest dose of NPHA (0.25 mgFe/mg polymer) significantly impaired non-neuronal differentiation with respect to those with NPCHI. When evaluated under AMF, cell viability slightly diminished in comparison with control hydrogels magnetically stimulated, but not compared to their counterparts without stimulation. Neuronal differentiation under AMF was only affected on collagen hydrogels with the highest dose of NPHA, while non-neuronal differentiation regained control values. Taken together, NPCHI-loaded hydrogels displayed a superior performance, maybe benefited from their higher nanomechanical fluidity. STATEMENT OF SIGNIFICANCE: Hydrogels and magnetic nanoparticles are undoubtedly useful biomaterials for biomedical applications. Nonetheless, the combination of both has been scarcely explored to date. In this study, we have designed hybrid 3D matrices integrating both components as promising magnetically responsive platforms for neural therapeutics. The resulting collagen scaffolds were soft (E0 ∼ 2.6 kPa) and biodegradable hydrogels with capacity to respond to external magnetic stimuli. Primary neural cells proved to grow on these substrates, preserving high viability and neuronal differentiation percentages even under the application of a high-frequency alternating magnetic field. Importantly, those hydrogels loaded with chitosan-coated iron oxide nanoparticles displayed a superior performance, likely related to their higher nanomechanical fluidity.
Collapse
Affiliation(s)
- Julia Martínez-Ramírez
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Marta Toldos-Torres
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Esther Benayas
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Natalia Villar-Gómez
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Laura Fernández-Méndez
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Francisco M Espinosa
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Ricardo García
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - Sabino Veintemillas-Verdaguer
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - María Del Puerto Morales
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain
| | - María Concepción Serrano
- Instituto de Ciencia de Materiales de Madrid, Consejo Superior de Investigaciones Científicas, Calle Sor Juana Inés de la Cruz 3, Madrid 28049, Spain.
| |
Collapse
|
32
|
Yang P, Lu Y, Gou W, Qin Y, Tan J, Luo G, Zhang Q. Glycosaminoglycans' Ability to Promote Wound Healing: From Native Living Macromolecules to Artificial Biomaterials. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305918. [PMID: 38072674 PMCID: PMC10916610 DOI: 10.1002/advs.202305918] [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: 08/21/2023] [Revised: 10/25/2023] [Indexed: 03/07/2024]
Abstract
Glycosaminoglycans (GAGs) are important for the occurrence of signaling molecules and maintenance of microenvironment within the extracellular matrix (ECM) in living tissues. GAGs and GAG-based biomaterial approaches have been widely explored to promote in situ tissue regeneration and repair by regulating the wound microenvironment, accelerating re-epithelialization, and controlling ECM remodeling. However, most approaches remain unacceptable for clinical applications. To improve insights into material design and clinical translational applications, this review highlights the innate roles and bioactive mechanisms of native GAGs during in situ wound healing and presents common GAG-based biomaterials and the adaptability of application scenarios in facilitating wound healing. Furthermore, challenges before the widespread commercialization of GAG-based biomaterials are shared, to ensure that future designed and constructed GAG-based artificial biomaterials are more likely to recapitulate the unique and tissue-specific profile of native GAG expression in human tissues. This review provides a more explicit and clear selection guide for researchers designing biomimetic materials, which will resemble or exceed their natural counterparts in certain functions, thereby suiting for specific environments or therapeutic goals.
Collapse
Affiliation(s)
- Peng Yang
- Institute of Burn ResearchState Key Laboratory of TraumaBurn and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Yifei Lu
- Institute of Burn ResearchState Key Laboratory of TraumaBurn and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Weiming Gou
- Institute of Burn ResearchState Key Laboratory of TraumaBurn and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Yiming Qin
- Department of Dermatology and Laboratory of DermatologyClinical Institute of Inflammation and ImmunologyFrontiers Science Center for Disease‐Related Molecular NetworkWest China HospitalSichuan UniversityChengdu610041China
| | - Jianglin Tan
- Institute of Burn ResearchState Key Laboratory of TraumaBurn and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Gaoxing Luo
- Institute of Burn ResearchState Key Laboratory of TraumaBurn and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Qing Zhang
- Institute of Burn ResearchState Key Laboratory of TraumaBurn and Combined InjurySouthwest HospitalThird Military Medical UniversityChongqing400038China
| |
Collapse
|
33
|
Shan BH, Wu FG. Hydrogel-Based Growth Factor Delivery Platforms: Strategies and Recent Advances. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2210707. [PMID: 37009859 DOI: 10.1002/adma.202210707] [Citation(s) in RCA: 50] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 03/25/2023] [Indexed: 06/19/2023]
Abstract
Growth factors play a crucial role in regulating a broad variety of biological processes and are regarded as powerful therapeutic agents in tissue engineering and regenerative medicine in the past decades. However, their application is limited by their short half-lives and potential side effects in physiological environments. Hydrogels are identified as having the promising potential to prolong the half-lives of growth factors and mitigate their adverse effects by restricting them within the matrix to reduce their rapid proteolysis, burst release, and unwanted diffusion. This review discusses recent progress in the development of growth factor-containing hydrogels for various biomedical applications, including wound healing, brain tissue repair, cartilage and bone regeneration, and spinal cord injury repair. In addition, the review introduces strategies for optimizing growth factor release including affinity-based delivery, carrier-assisted delivery, stimuli-responsive delivery, spatial structure-based delivery, and cellular system-based delivery. Finally, the review presents current limitations and future research directions for growth factor-delivering hydrogels.
Collapse
Affiliation(s)
- Bai-Hui Shan
- State Key Laboratory of Digital Medical Engineering Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Digital Medical Engineering Jiangsu Key Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| |
Collapse
|
34
|
Duan H, Li S, Hao P, Hao F, Zhao W, Gao Y, Qiao H, Gu Y, Lv Y, Bao X, Chiu K, So KF, Yang Z, Li X. Activation of endogenous neurogenesis and angiogenesis by basic fibroblast growth factor-chitosan gel in an adult rat model of ischemic stroke. Neural Regen Res 2024; 19:409-415. [PMID: 37488905 PMCID: PMC10503635 DOI: 10.4103/1673-5374.375344] [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: 07/14/2022] [Revised: 01/04/2023] [Accepted: 04/12/2023] [Indexed: 07/26/2023] Open
Abstract
Attempts have been made to use cell transplantation and biomaterials to promote cell proliferation, differentiation, migration, and survival, as well as angiogenesis, in the context of brain injury. However, whether bioactive materials can repair the damage caused by ischemic stroke by activating endogenous neurogenesis and angiogenesis is still unknown. In this study, we applied chitosan gel loaded with basic fibroblast growth factor to the stroke cavity 7 days after ischemic stroke in rats. The gel slowly released basic fibroblast growth factor, which improved the local microenvironment, activated endogenous neural stem/progenitor cells, and recruited these cells to migrate toward the penumbra and stroke cavity and subsequently differentiate into neurons, while enhancing angiogenesis in the penumbra and stroke cavity and ultimately leading to partial functional recovery. This study revealed the mechanism by which bioactive materials repair ischemic strokes, thus providing a new strategy for the clinical application of bioactive materials in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Shulun Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Fei Hao
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Yudan Gao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hui Qiao
- Department of Epidemiology and Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Yiming Gu
- Department of Physical Education, Capital University of Economics and Businessm, Beijing, China
| | - Yang Lv
- Department of Epidemiology and Statistics, School of Public Health and Management, Ningxia Medical University, Yinchuan, Ningxia Hui Autonomous Region, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kin Chiu
- Department of Psychology, State Key Lab of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administration Region, China
| | - Kwok-Fai So
- Guangdong-Hongkong-Macau Institute of CNS Regeneration, Ministry of Education CNS Regeneration Collaborative Joint Laboratory, Jinan University, Guangzhou, Guangdong Province, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, Guangdong Province, China
- Department of Ophthalmology and State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong Special Administration Region, China
- Center for Brain Science and Brain-Inspired Intelligence, Guangdong-Hong Kong-Macao Greater Bay Area, Guangzhou, Guangdong Province, China
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaoguang Li
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
- Department of Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
35
|
Mahmoudi N, Wang Y, Moriarty N, Ahmed NY, Dehorter N, Lisowski L, Harvey AR, Parish CL, Williams RJ, Nisbet DR. Neuronal Replenishment via Hydrogel-Rationed Delivery of Reprogramming Factors. ACS NANO 2024; 18:3597-3613. [PMID: 38221746 DOI: 10.1021/acsnano.3c11337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
The central nervous system's limited capacity for regeneration often leads to permanent neuronal loss following injury. Reprogramming resident reactive astrocytes into induced neurons at the site of injury is a promising strategy for neural repair, but challenges persist in stabilizing and accurately targeting viral vectors for transgene expression. In this study, we employed a bioinspired self-assembling peptide (SAP) hydrogel for the precise and controlled release of a hybrid adeno-associated virus (AAV) vector, AAVDJ, carrying the NeuroD1 neural reprogramming transgene. This method effectively mitigates the issues of high viral dosage at the target site, off-target delivery, and immunogenic reactions, enhancing the vector's targeting and reprogramming efficiency. In vitro, this vector successfully induced neuron formation, as confirmed by morphological, histochemical, and electrophysiological analyses. In vivo, SAP-mediated delivery of AAVDJ-NeuroD1 facilitated the trans-differentiation of reactive host astrocytes into induced neurons, concurrently reducing glial scarring. Our findings introduce a safe and effective method for treating central nervous system injuries, marking a significant advancement in regenerative neuroscience.
Collapse
Affiliation(s)
- Negar Mahmoudi
- Laboratory of Advanced Biomaterials, the John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
- ANU College of Engineering & Computer Science, Acton, ACT 2601, Australia
| | - Yi Wang
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC 3010, Australia
| | - Niamh Moriarty
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Noorya Y Ahmed
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Nathalie Dehorter
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2601, Australia
- The Queensland Brain Institute, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Leszek Lisowski
- Translational Vectorology Research Unit, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Vector and Genome Engineering Facility, Children's Medical Research Institute, Faculty of Medicine and Health, The University of Sydney, Westmead, NSW 2145, Australia
- Australian Genome Therapeutics Centre, Children's Medical Research Institute and Sydney Children's Hospitals Network, Westmead, NSW 2145, Australia
- Laboratory of Molecular Oncology and Innovative Therapies, Military Institute of Medicine, 04-141 Warsaw, Poland
| | - Alan R Harvey
- School of Human Sciences, The University of Western Australia, and Perron Institute for Neurological and Translational Science, Perth, WA 6009, Australia
| | - Clare L Parish
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, Melbourne, VIC 3010, Australia
| | - Richard J Williams
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- IMPACT, School of Medicine, Deakin University, Geelong, VIC 3217, Australia
| | - David R Nisbet
- Laboratory of Advanced Biomaterials, the John Curtin School of Medical Research, Australian National University, Canberra, ACT 2601, Australia
- The Graeme Clark Institute, The University of Melbourne, Melbourne, VIC 3010, Australia
- Department of Biomedical Engineering, Faculty of Engineering and Information Technology, The University of Melbourne, Melbourne, VIC 3010, Australia
- Melbourne Medical School, Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
36
|
Li Y, Yang KD, Kong DC, Li XM, Duan HY, Ye JF. Harnessing filamentous phages for enhanced stroke recovery. Front Immunol 2024; 14:1343788. [PMID: 38299142 PMCID: PMC10829096 DOI: 10.3389/fimmu.2023.1343788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 02/02/2024] Open
Abstract
Stroke poses a critical global health challenge, leading to substantial morbidity and mortality. Existing treatments often miss vital timeframes and encounter limitations due to adverse effects, prompting the pursuit of innovative approaches to restore compromised brain function. This review explores the potential of filamentous phages in enhancing stroke recovery. Initially antimicrobial-centric, bacteriophage therapy has evolved into a regenerative solution. We explore the diverse role of filamentous phages in post-stroke neurological restoration, emphasizing their ability to integrate peptides into phage coat proteins, thereby facilitating recovery. Experimental evidence supports their efficacy in alleviating post-stroke complications, immune modulation, and tissue regeneration. However, rigorous clinical validation is essential to address challenges like dosing and administration routes. Additionally, genetic modification enhances their potential as injectable biomaterials for complex brain tissue issues. This review emphasizes innovative strategies and the capacity of filamentous phages to contribute to enhanced stroke recovery, as opposed to serving as standalone treatment, particularly in addressing stroke-induced brain tissue damage.
Collapse
Affiliation(s)
- Yang Li
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
- School of Nursing, Jilin University, Changchun, China
| | - Kai-di Yang
- School of Nursing, Jilin University, Changchun, China
| | - De-cai Kong
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiao-meng Li
- School of Nursing, Jilin University, Changchun, China
| | - Hao-yu Duan
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| | - Jun-feng Ye
- General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
37
|
Aqel S, Al-Thani N, Haider MZ, Abdelhady S, Al Thani AA, Kobeissy F, Shaito AA. Biomaterials in Traumatic Brain Injury: Perspectives and Challenges. BIOLOGY 2023; 13:21. [PMID: 38248452 PMCID: PMC10813103 DOI: 10.3390/biology13010021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 01/23/2024]
Abstract
Traumatic brain injury (TBI) is a leading cause of mortality and long-term impairment globally. TBI has a dynamic pathology, encompassing a variety of metabolic and molecular events that occur in two phases: primary and secondary. A forceful external blow to the brain initiates the primary phase, followed by a secondary phase that involves the release of calcium ions (Ca2+) and the initiation of a cascade of inflammatory processes, including mitochondrial dysfunction, a rise in oxidative stress, activation of glial cells, and damage to the blood-brain barrier (BBB), resulting in paracellular leakage. Currently, there are no FDA-approved drugs for TBI, but existing approaches rely on delivering micro- and macromolecular treatments, which are constrained by the BBB, poor retention, off-target toxicity, and the complex pathology of TBI. Therefore, there is a demand for innovative and alternative therapeutics with effective delivery tactics for the diagnosis and treatment of TBI. Tissue engineering, which includes the use of biomaterials, is one such alternative approach. Biomaterials, such as hydrogels, including self-assembling peptides and electrospun nanofibers, can be used alone or in combination with neuronal stem cells to induce neurite outgrowth, the differentiation of human neural stem cells, and nerve gap bridging in TBI. This review examines the inclusion of biomaterials as potential treatments for TBI, including their types, synthesis, and mechanisms of action. This review also discusses the challenges faced by the use of biomaterials in TBI, including the development of biodegradable, biocompatible, and mechanically flexible biomaterials and, if combined with stem cells, the survival rate of the transplanted stem cells. A better understanding of the mechanisms and drawbacks of these novel therapeutic approaches will help to guide the design of future TBI therapies.
Collapse
Affiliation(s)
- Sarah Aqel
- Medical Research Center, Hamad Medical Corporation, Doha P.O. Box 3050, Qatar
| | - Najlaa Al-Thani
- Research and Development Department, Barzan Holdings, Doha P.O. Box 7178, Qatar
| | - Mohammad Z. Haider
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Samar Abdelhady
- Faculty of Medicine, Alexandria University, Alexandria 21544, Egypt;
| | - Asmaa A. Al Thani
- Biomedical Research Center and Department of Biomedical Sciences, College of Health Science, QU Health, Qatar University, Doha P.O. Box 2713, Qatar;
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Morehouse School of Medicine, 720 Westview Dr. SW, Atlanta, GA 30310, USA
| | - Abdullah A. Shaito
- Biomedical Research Center, Department of Biomedical Sciences at College of Health Sciences, College of Medicine, Qatar University, Doha P.O. Box 2713, Qatar
| |
Collapse
|
38
|
Morris GP, Sutherland BA. The presence of functional blood vessels in the ischemic core provides a therapeutic target for stroke recovery. Neural Regen Res 2023; 18:2653-2654. [PMID: 37449607 DOI: 10.4103/1673-5374.373703] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/18/2023] Open
Affiliation(s)
- Gary P Morris
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| | - Brad A Sutherland
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
39
|
Fan JS, Wang M, Chen N, Sun BC, Zhang QB, Li Y, Huang MJ. Association between inflammatory bowel disease and risk of stroke: a systematic review and meta-analysis of cohort studies. Front Neurol 2023; 14:1204727. [PMID: 38046580 PMCID: PMC10693426 DOI: 10.3389/fneur.2023.1204727] [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: 04/12/2023] [Accepted: 10/25/2023] [Indexed: 12/05/2023] Open
Abstract
Background/objectives Recently, four meta-analyses have explored the association between inflammatory bowel disease (IBD) and the risk of stroke. These studies have demonstrated that people with IBD may be at an increased risk of stroke. However, some limitations such as high heterogeneity and the lack of uniformity in the types of research, especially the reuse of some sample sizes, cannot be neglected. These factors reduce the credibility of their research conclusions. Therefore, we conducted a meta-analysis to explore this possible association. Methods PubMed, Embase, and Web of Science were searched from inception to 30 June 2023. A random effects model with the generic inverse variance method was used in this meta-analysis. The Review Manager software was used to obtain all relative risks (RRs) and their 95% confidence intervals (CIs). Publication bias was tested, and sensitivity and subgroup analyses were conducted to explore possible heterogeneities. Results This meta-analysis included 12 cohort studies (involving 4,495,055 individuals). Meta-analysis of these data has shown that IBD was associated with an increased risk of stroke (RR = 1.19, 95%CI:1.14-1.24, p < 0.00001). Our results were stable and robust in subgroup and sensitivity analyses. Conclusions Our results suggest that IBD is associated with an increased risk of stroke. To reduce the incidence of stroke, patients with IBD are encouraged to undergo stroke risk assessments, especially for young female patients; assessing the risk of ischemic stroke is of particular importance. Prospective studies considering stroke subtypes, IBD severity and treatments, regions, and other confounding factors are needed to further explore the nature of each association. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD42022373656.
Collapse
Affiliation(s)
- Jin-Shan Fan
- Department of Intensive Care Unit (ICU), Qian Jiang Central Hospital of Hubei Province, Qian Jiang Hospital Affiliated to Renmin Hospital of Wuhan University, Qian Jiang Clinical Medical College, Health Science Center, Yangtze University, Qianjiang, China
| | - Meng Wang
- Department of Neurology, The Third Clinical Medical College of China, Three Gorges University, Gezhouba Central Hospital of Sinopharm, Yichang, China
| | - Ni Chen
- Department of Ophthalmology, The Third Clinical Medical College of China, Three Gorges University, Gezhouba Central Hospital of Sinopharm, Yichang, China
| | - Bai-chao Sun
- Department of Ophthalmology, The Third Clinical Medical College of China, Three Gorges University, Gezhouba Central Hospital of Sinopharm, Yichang, China
| | - Qi-Bing Zhang
- Department of Neurology, The Third Clinical Medical College of China, Three Gorges University, Gezhouba Central Hospital of Sinopharm, Yichang, China
| | - Yong Li
- Department of Neurology, The Third Clinical Medical College of China, Three Gorges University, Gezhouba Central Hospital of Sinopharm, Yichang, China
| | - Ming-Jie Huang
- Department of Ophthalmology, The Third Clinical Medical College of China, Three Gorges University, Gezhouba Central Hospital of Sinopharm, Yichang, China
| |
Collapse
|
40
|
Ghuman H, Kim K, Barati S, Ganguly K. Emergence of task-related spatiotemporal population dynamics in transplanted neurons. Nat Commun 2023; 14:7320. [PMID: 37951968 PMCID: PMC10640594 DOI: 10.1038/s41467-023-43081-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/31/2023] [Indexed: 11/14/2023] Open
Abstract
Loss of nervous system tissue after severe brain injury is a main determinant of poor functional recovery. Cell transplantation is a promising method to restore lost tissue and function, yet it remains unclear if transplanted neurons can demonstrate the population level dynamics important for movement control. Here we present a comprehensive approach for long-term single neuron monitoring and manipulation of transplanted embryonic cortical neurons after cortical injury in adult male mice performing a prehension task. The observed patterns of population activity in the transplanted network strongly resembled that of healthy networks. Specifically, the task-related spatiotemporal activity patterns of transplanted neurons could be represented by latent factors that evolve within a low dimensional manifold. We also demonstrate reliable modulation of the transplanted networks using minimally invasive epidural stimulation. Our approach may allow greater insight into how restoration of cell-type specific network dynamics in vivo can restore motor function.
Collapse
Affiliation(s)
- Harman Ghuman
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Kyungsoo Kim
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Sapeeda Barati
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Karunesh Ganguly
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
- Neurology Service, San Francisco Veterans Affairs Medical Center, San Francisco, CA, USA.
| |
Collapse
|
41
|
Wang Z, Zheng D, Tan YS, Yuan Q, Yuan F, Zhang S. Enabling Survival of Transplanted Neural Precursor Cells in the Ischemic Brain. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302527. [PMID: 37867250 PMCID: PMC10667812 DOI: 10.1002/advs.202302527] [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: 04/20/2023] [Revised: 07/24/2023] [Indexed: 10/24/2023]
Abstract
There is no effective therapy for ischemic stroke following the acute stage. Neural transplantation offers a potential option for repairing the ischemic lesion. However, this strategy is hindered by the poor survival of the neural precursor cells (NPCs) that are transplanted into the inflammatory ischemic core. Here, a chemical cocktail consisting of fibrinogen and maraviroc is developed to promote the survival of the transplanted NPCs in the ischemic core of the mouse cerebral cortex. The grafted NPCs survive in the presence of the cocktail but not fibrinogen or maraviroc alone at day 7. The surviving NPCs divide and differentiate to mature neurons by day 30, reconstituting the infarct cortex with vascularization. Molecular analysis in vivo and in vitro shows that blocking the activation of CCR5 on the NPCs protects the NPCs from apoptosis induced by pro-inflammatory factors, revealing the underlying protective effect of the cocktail for NPCs. The findings open an avenue to enable survival of the transplanted NPCs under the inflammatory neurological conditions like stroke.
Collapse
Affiliation(s)
- Zhifu Wang
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Danyi Zheng
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Ye Sing Tan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Qiang Yuan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Fang Yuan
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
| | - Su‐Chun Zhang
- Program in Neuroscience & Behavioral Disorders, GK Goh Centre for NeuroscienceDuke‐NUS Medical SchoolSingapore169857Singapore
- Department of NeuroscienceDepartment of NeurologyWaisman CenterUniversity of Wisconsin‐MadisonMadisonWI53705USA
| |
Collapse
|
42
|
Liu Z, Zhang S, Ran Y, Geng H, Gao F, Tian G, Feng Z, Xi J, Ye L, Su W. Nanoarchitectonics of tannic acid based injectable hydrogel regulate the microglial phenotype to enhance neuroplasticity for poststroke rehabilitation. Biomater Res 2023; 27:108. [PMID: 37908012 PMCID: PMC10617113 DOI: 10.1186/s40824-023-00444-0] [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: 08/13/2023] [Accepted: 10/08/2023] [Indexed: 11/02/2023] Open
Abstract
BACKGROUND Stroke is the second leading cause of mortality and disability worldwide. Poststroke rehabilitation is still unsatisfactory in clinics, which brings great pain and economic burdens to stroke patients. In this study, an injectable hydrogel in which tannic acid (TA) acts as not only a building block but also a therapeutic drug, was developed for poststroke rehabilitation. METHODS TA is used as a building block to form an injectable hydrogel (TA gel) with carboxymethyl chitosan (CMCS) by multivalent hydrogen bonds. The morphology, rheological properties, and TA release behavior of the hydrogel were characterized. The abilities of the TA gel to modulate microglial (BV2 cells) polarization and subsequently enhance the neuroplasticity of neuro cells (N2a cells) were assessed in vitro. The TA gel was injected into the cavity of stroke mice to evaluate motor function recovery, microglial polarization, and neuroplasticity in vivo. The molecular pathway through which TA modulates microglial polarization was also explored both in vitro and in vivo. RESULTS The TA gel exhibited sustainable release behavior of TA. The TA gel can suppress the expression of CD16 and IL-1β, and upregulate the expression of CD206 and TGF-β in oxygen and glucose-deprived (OGD) BV2 cells, indicating the regulation of OGD BV2 cells to an anti-inflammatory phenotype in vitro. This finding further shows that the decrease in synaptophysin and PSD95 in OGD N2a cells is effectively recovered by anti-inflammatory BV2 cells. Furthermore, the TA gel decreased CD16/iNOS expression and increased CD206 expression in the peri-infarct area of stroke mice, implying anti-inflammatory polarization of microglia in vivo. The colocalization of PSD95 and Vglut1 stains, as well as Golgi staining, showed the enhancement of neuroplasticity by the TA gel. Spontaneously, the TA gel successfully recovered the motor function of stroke mice. The western blot results in vitro and in vivo suggested that the TA gel regulated microglial polarization via the NF-κB pathway. CONCLUSION The TA gel serves as an effective brain injectable implant to treat stroke and shows promising potential to promote poststroke rehabilitation in the clinic.
Collapse
Affiliation(s)
- Zongjian Liu
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, China
| | - Shulei Zhang
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanyuan Ran
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, China
| | - Huimin Geng
- Department of Neurosurgery, Qilu Hospital of Shandong University, Shandong University, Jinan, Shandong, 250012, China.
| | - Fuhai Gao
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, China
| | - Guiqin Tian
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, China
| | - Zengguo Feng
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Jianing Xi
- Beijing Rehabilitation Hospital, Capital Medical University, Beijing, 100144, China
| | - Lin Ye
- School of Materials Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China.
| | - Wei Su
- Beijing Tsinghua Chang Gung Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 102218, China.
| |
Collapse
|
43
|
Nguyen JN, Mohan EC, Pandya G, Ali U, Tan C, Kofler JK, Shapiro L, Marrelli SP, Chauhan A. CD13 facilitates immune cell migration and aggravates acute injury but promotes chronic post-stroke recovery. J Neuroinflammation 2023; 20:232. [PMID: 37817190 PMCID: PMC10566099 DOI: 10.1186/s12974-023-02918-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Accepted: 10/01/2023] [Indexed: 10/12/2023] Open
Abstract
INTRODUCTION Acute stroke leads to the activation of myeloid cells. These cells express adhesion molecules and transmigrate to the brain, thereby aggravating injury. Chronically after stroke, repair processes, including angiogenesis, are activated and enhance post-stroke recovery. Activated myeloid cells express CD13, which facilitates their migration into the site of injury. However, angiogenic blood vessels which play a role in recovery also express CD13. Overall, the specific contribution of CD13 to acute and chronic stroke outcomes is unknown. METHODS CD13 expression was estimated in both mice and humans after the ischemic stroke. Young (8-12 weeks) male wild-type and global CD13 knockout (KO) mice were used for this study. Mice underwent 60 min of middle cerebral artery occlusion (MCAO) followed by reperfusion. For acute studies, the mice were euthanized at either 24- or 72 h post-stroke. For chronic studies, the Y-maze, Barnes maze, and the open field were performed on day 7 and day 28 post-stroke. Mice were euthanized at day 30 post-stroke and the brains were collected for assessment of inflammation, white matter injury, tissue loss, and angiogenesis. Flow cytometry was performed on days 3 and 7 post-stroke to quantify infiltrated monocytes and neutrophils and CXCL12/CXCR4 signaling. RESULTS Brain CD13 expression and infiltrated CD13+ monocytes and neutrophils increased acutely after the stroke. The brain CD13+lectin+ blood vessels increased on day 15 after the stroke. Similarly, an increase in the percentage area CD13 was observed in human stroke patients at the subacute time after stroke. Deletion of CD13 resulted in reduced infarct volume and improved neurological recovery after acute stroke. However, CD13KO mice had significantly worse memory deficits, amplified gliosis, and white matter damage compared to wild-type animals at chronic time points. CD13-deficient mice had an increased percentage of CXCL12+cells but a reduced percentage of CXCR4+cells and decreased angiogenesis at day 30 post-stroke. CONCLUSIONS CD13 is involved in the trans-migration of monocytes and neutrophils after stroke, and acutely, led to decreased infarct size and improved behavioral outcomes. However, loss of CD13 led to reductions in post-stroke angiogenesis by reducing CXCL12/CXCR4 signaling.
Collapse
Affiliation(s)
- Justin N Nguyen
- University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Eric C Mohan
- University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Gargee Pandya
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Uzma Ali
- Baylor University, Waco, TX, USA
| | - Chunfeng Tan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Julia K Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Linda Shapiro
- Center for Vascular Biology, The University of Connecticut Health Center, Farmington, CT, USA
| | - Sean P Marrelli
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA.
| |
Collapse
|
44
|
Yang X, Qi Y, Wang C, Zwang TJ, Rommelfanger NJ, Hong G, Lieber CM. Laminin-coated electronic scaffolds with vascular topography for tracking and promoting the migration of brain cells after injury. Nat Biomed Eng 2023; 7:1282-1292. [PMID: 37814007 DOI: 10.1038/s41551-023-01101-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 08/30/2023] [Indexed: 10/11/2023]
Abstract
In the adult brain, neural stem cells are largely restricted into spatially discrete neurogenic niches, and hence areas of neuron loss during neurodegenerative disease or following a stroke or traumatic brain injury do not typically repopulate spontaneously. Moreover, understanding neural activity accompanying the neural repair process is hindered by a lack of minimally invasive devices for the chronic measurement of the electrophysiological dynamics in damaged brain tissue. Here we show that 32 individually addressable platinum microelectrodes integrated into laminin-coated branched polymer scaffolds stereotaxically injected to span a hydrogel-filled cortical lesion and deeper regions in the brains of mice promote neural regeneration while allowing for the tracking of migrating host brain cells into the lesion. Chronic measurements of single-unit activity and neural-circuit analyses revealed the establishment of spiking activity in new neurons in the lesion and their functional connections with neurons deeper in the brain. Electronic implants mimicking the topographical and surface properties of brain vasculature may aid the stimulation and tracking of neural-circuit restoration following injury.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- Department of Psychiatry and Behavioral Sciences and Department of Chemistry, Stanford University, Stanford, CA, USA.
| | - Yue Qi
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Beijing Graphene Institute, Beijing, China
| | - Chonghe Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Theodore J Zwang
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
- Department of Neurology, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Charlestown, MA, USA
| | | | - Guosong Hong
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| | - Charles M Lieber
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, USA.
- Lieber Research Group, Lexington, MA, USA.
| |
Collapse
|
45
|
Qin W, Wan Q, Yan J, Han X, Lu W, Ma Z, Ye T, Li Y, Li C, Wang C, Tay FR, Niu L, Jiao K. Effect of Extracellular Ribonucleic Acids on Neurovascularization in Osteoarthritis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301763. [PMID: 37395388 PMCID: PMC10502862 DOI: 10.1002/advs.202301763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 06/14/2023] [Indexed: 07/04/2023]
Abstract
Osteoarthritis is a degenerative disease characterized by abnormal neurovascularization at the osteochondral junctions, the regulatory mechanisms of which remain poorly understood. In the present study, a murine osteoarthritic model with augmented neurovascularization at the osteochondral junction is used to examine this under-evaluated facet of degenerative joint dysfunction. Increased extracellular RNA (exRNA) content is identified in neurovascularized osteoarthritic joints. It is found that the amount of exRNA is positively correlated with the extent of neurovascularization and the expression of vascular endothelial growth factor (VEGF). In vitro binding assay and molecular docking demonstrate that synthetic RNAs bind to VEGF via electrostatic interactions. The RNA-VEGF complex promotes the migration and function of endothelial progenitor cells and trigeminal ganglion cells. The use of VEGF and VEGFR2 inhibitors significantly inhibits the amplification of the RNA-VEGF complex. Disruption of the RNA-VEGF complex by RNase and polyethyleneimine reduces its in vitro activities, as well as prevents excessive neurovascularization and osteochondral deterioration in vivo. The results of the present study suggest that exRNAs may be potential targets for regulating nerve and blood vessel ingrowth under physiological and pathological joint conditions.
Collapse
Affiliation(s)
- Wen‐pin Qin
- Department of StomatologyTangdu hospitalThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Qian‐Qian Wan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Jian‐Fei Yan
- Department of StomatologyTangdu hospitalThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Xiao‐Xiao Han
- Department of StomatologyTangdu hospitalThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Wei‐Cheng Lu
- Department of StomatologyTangdu hospitalThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Zhang‐Yu Ma
- Department of StomatologyTangdu hospitalThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Tao Ye
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Yu‐Tao Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Chang‐Jun Li
- Department of EndocrinologyEndocrinology Research CenterThe Xiangya Hospital of Central South UniversityChangshaHunan410008P. R. China
| | - Chen Wang
- Department of StomatologyThe Eighth Medical Center of PLA General HospitalHaidian DistrictBeijingP. R. China100091
| | - Franklin R. Tay
- Dental College of GeorgiaAugusta UniversityAugustaGA30912USA
| | - Li‐Na Niu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| | - Kai Jiao
- Department of StomatologyTangdu hospitalThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of StomatologySchool of StomatologyThe Fourth Military Medical UniversityXi'anShaanxi710032P. R. China
| |
Collapse
|
46
|
Mu J, Hao P, Duan H, Zhao W, Wang Z, Yang Z, Li X. Non-human primate models of focal cortical ischemia for neuronal replacement therapy. J Cereb Blood Flow Metab 2023; 43:1456-1474. [PMID: 37254891 PMCID: PMC10414004 DOI: 10.1177/0271678x231179544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 03/13/2023] [Accepted: 04/26/2023] [Indexed: 06/01/2023]
Abstract
Despite the high prevalence, stroke remains incurable due to the limited regeneration capacity in the central nervous system. Neuronal replacement strategies are highly diverse biomedical fields that attempt to replace lost neurons by utilizing exogenous stem cell transplants, biomaterials, and direct neuronal reprogramming. Although these approaches have achieved encouraging outcomes mostly in the rodent stroke model, further preclinical validation in non-human primates (NHP) is still needed prior to clinical trials. In this paper, we briefly review the recent progress of promising neuronal replacement therapy in NHP stroke studies. Moreover, we summarize the key characteristics of the NHP as highly valuable translational tools and discuss (1) NHP species and their advantages in terms of genetics, physiology, neuroanatomy, immunology, and behavior; (2) various methods for establishing NHP focal ischemic models to study the regenerative and plastic changes associated with motor functional recovery; and (3) a comprehensive analysis of experimentally and clinically accessible outcomes and a potential adaptive mechanism. Our review specifically aims to facilitate the selection of the appropriate NHP cortical ischemic models and efficient prognostic evaluation methods in preclinical stroke research design of neuronal replacement strategies.
Collapse
Affiliation(s)
- Jiao Mu
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing, China
| | - Peng Hao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Hongmei Duan
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Wen Zhao
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zijue Wang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Zhaoyang Yang
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| | - Xiaoguang Li
- Beijing Key Laboratory for Biomaterials and Neural Regeneration, School of Engineering Medicine, Beihang University, Beijing, China
- Department of Neurobiology, School of Basic Medical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
47
|
Fan Z, Ardicoglu R, Batavia AA, Rust R, von Ziegler L, Waag R, Zhang J, Desgeorges T, Sturman O, Dang H, Weber R, Roszkowski M, Moor AE, Schwab ME, Germain PL, Bohacek J, De Bock K. The vascular gene Apold1 is dispensable for normal development but controls angiogenesis under pathological conditions. Angiogenesis 2023; 26:385-407. [PMID: 36933174 PMCID: PMC10328887 DOI: 10.1007/s10456-023-09870-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Accepted: 02/06/2023] [Indexed: 03/19/2023]
Abstract
The molecular mechanisms of angiogenesis have been intensely studied, but many genes that control endothelial behavior and fate still need to be described. Here, we characterize the role of Apold1 (Apolipoprotein L domain containing 1) in angiogenesis in vivo and in vitro. Single-cell analyses reveal that - across tissues - the expression of Apold1 is restricted to the vasculature and that Apold1 expression in endothelial cells (ECs) is highly sensitive to environmental factors. Using Apold1-/- mice, we find that Apold1 is dispensable for development and does not affect postnatal retinal angiogenesis nor alters the vascular network in adult brain and muscle. However, when exposed to ischemic conditions following photothrombotic stroke as well as femoral artery ligation, Apold1-/- mice display dramatic impairments in recovery and revascularization. We also find that human tumor endothelial cells express strikingly higher levels of Apold1 and that Apold1 deletion in mice stunts the growth of subcutaneous B16 melanoma tumors, which have smaller and poorly perfused vessels. Mechanistically, Apold1 is activated in ECs upon growth factor stimulation as well as in hypoxia, and Apold1 intrinsically controls EC proliferation but not migration. Our data demonstrate that Apold1 is a key regulator of angiogenesis in pathological settings, whereas it does not affect developmental angiogenesis, thus making it a promising candidate for clinical investigation.
Collapse
Affiliation(s)
- Zheng Fan
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland
- Institute of Anatomy, University of Zürich, Winterthurerstrasse 190, 8057, Zurich, Switzerland
| | - Raphaela Ardicoglu
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland
- Department of Health Sciences and Technology, Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, ETH Zürich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zürich, University of Zürich, Zurich, Switzerland
| | - Aashil A Batavia
- Department of Pathology and Molecular Pathology, University and University Hospital Zürich, Zurich, Switzerland
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Ruslan Rust
- Department of Health Sciences and Technology, Institute for Regenerative Medicine, University of Zürich, ETH Zürich, Zurich, Switzerland
| | - Lukas von Ziegler
- Department of Health Sciences and Technology, Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, ETH Zürich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zürich, University of Zürich, Zurich, Switzerland
| | - Rebecca Waag
- Department of Health Sciences and Technology, Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, ETH Zürich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zürich, University of Zürich, Zurich, Switzerland
| | - Jing Zhang
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland
| | - Thibaut Desgeorges
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland
| | - Oliver Sturman
- Department of Health Sciences and Technology, Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, ETH Zürich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zürich, University of Zürich, Zurich, Switzerland
| | - Hairuo Dang
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland
- DKFZ-ZMBH Alliance, Im Neuenheimer Feld 282, 69120, Heidelberg, Germany
| | - Rebecca Weber
- Department of Health Sciences and Technology, Institute for Regenerative Medicine, University of Zürich, ETH Zürich, Zurich, Switzerland
| | - Martin Roszkowski
- Department of Health Sciences and Technology, Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, ETH Zürich, Zurich, Switzerland
- Neuroscience Center Zurich, ETH Zürich, University of Zürich, Zurich, Switzerland
| | - Andreas E Moor
- Department of Biosystems Science and Engineering, ETH Zürich, Basel, Switzerland
| | - Martin E Schwab
- Department of Health Sciences and Technology, Institute for Regenerative Medicine, University of Zürich, ETH Zürich, Zurich, Switzerland
| | - Pierre-Luc Germain
- Neuroscience Center Zurich, ETH Zürich, University of Zürich, Zurich, Switzerland
- Department of Health Sciences and Technology, Computational Neurogenomics, Institute for Neuroscience, ETH Zürich, Zurich, Switzerland
- Department for Molecular Life Sciences, Laboratory of Statistical Bioinformatics, University of Zürich, Zurich, Switzerland
| | - Johannes Bohacek
- Department of Health Sciences and Technology, Laboratory of Molecular and Behavioral Neuroscience, Institute for Neuroscience, ETH Zürich, Zurich, Switzerland.
- Neuroscience Center Zurich, ETH Zürich, University of Zürich, Zurich, Switzerland.
| | - Katrien De Bock
- Department of Health Sciences and Technology, Laboratory of Exercise and Health, ETH Zürich, Zurich, Switzerland.
| |
Collapse
|
48
|
Wilson KL, Onweller LA, Joseph NI, David-Bercholz J, Darling NJ, Segura T. SDF-1 Bound Heparin Nanoparticles Recruit Progenitor Cells for Their Differentiation and Promotion of Angiogenesis After Stroke. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.07.05.547800. [PMID: 37461490 PMCID: PMC10349963 DOI: 10.1101/2023.07.05.547800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2023]
Abstract
Angiogenesis after stroke is correlated with enhanced tissue repair and functional outcomes. The existing body of research in biomaterials for stroke focuses on hydrogels for the delivery of stem cells, growth factors, or small molecules or drugs. Despite the ability of hydrogels to enhance all these delivery methods, no material has significantly regrown vasculature within the translatable timeline of days to weeks after stroke. Here we developed 2 novel biomaterials for tissue regeneration after stroke, a highly porous granular hydrogel termed Cryo microgels, and heparin-norbornene nanoparticles with covalently bound SDF-1α. The combination of these materials resulted in fully revascularized vessels throughout the stroke core in only 10 days, as well as increased neural progenitor cell migration and maintenance and increased neurons.
Collapse
|
49
|
Wang J, Wang Y, Xiaohalati X, Su Q, Liu J, Cai B, Yang W, Wang Z, Wang L. A Bioinspired Manganese-Organic Framework Ameliorates Ischemic Stroke through its Intrinsic Nanozyme Activity and Upregulating Endogenous Antioxidant Enzymes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206854. [PMID: 37129343 PMCID: PMC10369237 DOI: 10.1002/advs.202206854] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 03/21/2023] [Indexed: 05/03/2023]
Abstract
Following stroke, oxidative stress induced by reactive oxygen species (ROS) aggravates neuronal damage and enlarges ischemic penumbra, which is devastating to stroke patients. Nanozyme-based antioxidants are emerging to treat stroke through scavenging excessive ROS. However, most of nanozymes cannot efficiently scavenge ROS in neuronal cytosol and mitochondria, due to low-uptake abilities of neurons and barriers of organelle membranes, significantly limiting nanozymes' neuroprotective effects. To overcome this limitation, a manganese-organic framework modified with polydopamine (pDA-MNOF), capable of not only mimicking catalytic activities of natural SOD2's catalytic domain but also upregulating two endogenous antioxidant enzymes in neurons is fabricated. With such a dual anti-ROS effect, this nanozyme robustly decreases cellular ROS and effectively protects them from ROS-induced injury. STAT-3 signaling is found to play a vital role in pDA-MNOF activating the two antioxidant enzymes, HO1 and SOD2. In vivo pDA-MNOF treatment significantly improves the survival of middle cerebral artery occlusion (MCAo) mice by reducing infarct volume and more importantly, promotes animal behavioral recovery. Further, pDA-MNOF activates vascular endothelial growth factor expression, a downstream target of STAT3 signaling, thus enhancing angiogenesis. Taken together, the biochemical, cell-biological, and animal-level behavioral data demonstrate the potentiality of pDA-MNOF as a dual ROS-scavenging agent for stroke treatment.
Collapse
Affiliation(s)
- Jian Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Yang Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Xiakeerzhati Xiaohalati
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Qiangfei Su
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Jingwei Liu
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Bo Cai
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Wen Yang
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Zheng Wang
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| | - Lin Wang
- Department of Clinical LaboratoryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
- Hubei Key Laboratory of Regenerative Medicine and Multi‐disciplinary Translational ResearchResearch Center for Tissue Engineering and Regenerative MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022P. R. China
| |
Collapse
|
50
|
Jiang L, Wu X, Wang Y, Liu C, Wu Y, Wang J, Xu N, He Z, Wang S, Zhang H, Wang X, Lu X, Tan Q, Sun X. Photothermal Controlled-Release Immunomodulatory Nanoplatform for Restoring Nerve Structure and Mechanical Nociception in Infectious Diabetic Ulcers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300339. [PMID: 37148168 PMCID: PMC10369251 DOI: 10.1002/advs.202300339] [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: 01/16/2023] [Revised: 03/04/2023] [Indexed: 05/08/2023]
Abstract
Infectious diabetic ulcers (IDU) require anti-infection, angiogenesis, and nerve regeneration therapy; however, the latter has received comparatively less research attention than the former two. In particular, there have been few reports on the recovery of mechanical nociception. In this study, a photothermal controlled-release immunomodulatory hydrogel nanoplatform is tailored for the treatment of IDU. Due to a thermal-sensitive interaction between polydopamine-reduced graphene oxide (pGO) and the antibiotic mupirocin, excellent antibacterial efficacy is achieved through customized release kinetics. In addition, Trem2+ macrophages recruited by pGO regulate collagen remodeling and restore skin adnexal structures to alter the fate of scar formation, promote angiogenesis, accompanied by the regeneration of neural networks, which ensures the recovery of mechanical nociception and may prevent the recurrence of IDU at the source. In all, a full-stage strategy from antibacterial, immune regulation, angiogenesis, and neurogenesis to the recovery of mechanical nociception, an indispensable neural function of skin, is introduced to IDU treatment, which opens up an effective and comprehensive therapy for refractory IDU.
Collapse
Affiliation(s)
- Le Jiang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Xiangyi Wu
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Yifan Wang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Chunlin Liu
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Yixian Wu
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Jingyun Wang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Nan Xu
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Zhijun He
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Shuqin Wang
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Hao Zhang
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| | - Xiong Lu
- Key Lab of Advanced Technologies of MaterialsMinistry of EducationSchool of Materials Science and EngineeringSouthwest Jiaotong UniversityChengduSichuan610031China
| | - Qian Tan
- Department of Burns and Plastic SurgeryNanjing Drum Tower HospitalThe Affiliated Hospital of Nanjing University Medical SchoolNo. 321, Zhongshan RoadNanjingJiangsu210008China
| | - Xiaodan Sun
- State Key Laboratory of New Ceramics and Fine ProcessingSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
- Key Laboratory of Advanced Materials of Ministry of Education of ChinaSchool of Materials Science and EngineeringTsinghua UniversityBeijing100084P. R. China
| |
Collapse
|